Title of Invention

"AN ALBUMIN FUSION PROTEIN COMPRISING TWO OR MORE TANDEMLY ORIENTED GLUCAGON-LIKE-PEPTIDE 1 (GLP-1) POLYPEPTIDES"

Abstract An albumin fusion protein comprising two or more tandemly oriented Glucagon-Like-Peptide 1 (GLP-1) polypeptides, wherein said GLP-1 polypeptides are selected from wild-type GLP-1, GLP-1 (9-36), GLP-1 (7-36), and GLP-1(7-36(A8G)) fused to albumin-wherein said albumin increases the serum plasma half-life of unfused GLP-1 polypeptides, and wherein said fusion protein has GLP-1 activity, and wherein the albumin fusion protein further comprises a modified HSA/Kex2 leader sequence.
Full Text Albumin Fusion Proteins
REFERENCE TO SEQUENCE LISTING ON COMPACT DISC
|001] This application refers to a "Sequence Listing" listed below, which is provided as an electronic document on three identical compact discs (CD-R), labeled "Copy 1," "Copy 2," and "Copy 3." These compact discs each contain the file "PF6I2PCT SL.txt" (929,048 bytes, created on February 7, 2005), which is incorporated by reference in its entirety.
BACKGROUND OF THE INVENTION
[0002| 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.
[0003] 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 UP 330 451 and and 361 991.
[0004| 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 lyophilizcd 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.
[0005] 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.
[0006] Upon in vivo administration, therapeutic proteins in their native state or when recombinantly produced, such as interferons and growth hormones, exhibit a short plasma stability due to rapid clearance from the bloodstream. Accordingly, the therapeutic effects provided by these proteins are also short-lived. Thus, in order to sustain their desired therapeutic effect in vivo, the rapid clearance of these proteins from the blood dictates that the therapeutic molecules must be administered more frequently or at a higher dose. However, increasing the dosing schedule for administration of the therapeutic protein often results in an increase in injection site reactions, side-effects, and toxicity in the patient. Similarly, administration of the therapeutic protein at a higher dose also commonly results in an increase in toxicity and side-effects in the patient. |0007] The few practical solutions to increasing plasma stability of therapeutic molecules that have been proposed, including chemical conjugation, have provided limited benefit to the patient. Generally, in most cases, these chemically modified therapeutic molecules are still administered on a frequent dosing schedule, retaining significant injection site reactions, side-effects, and toxicity in patients. Accordingly, there is a need for an stabilized form of therapeutic molecules that retains a higher plasma stability in vivo than the native or recombinantly produced therapeutic alone and can be administered less frequently, thereby decreasing potential side-effects to the patient.
SUMMARY OF THE INVENTION
[0008] 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 pepude, or fragment or variant thereof) fused to albumin or a fragment (portion) or variant of albumin. The present invention also encompasses polynucleotides, comprising, or alternatively consisting of, nucleic acid molecules encoding proteins comprising a Therapeutic protein (e.g., a polypeptide, antibody, or peptide, or fragment or variant thereof) fused to albumin or a fragment (portion) or variant of albumin, that is sufficient to prolong the shelf life of the Therapeutic protein, to increase the plasma stability of the Therapeutic protein compared to its unfused state, and/or stabilize the Therapeutic protein and/or its activity in solution (or in a pharmaceutical composition) in vitro and/or in vivo. Albumin fusion proteins encoded by a polynuclcoiide 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. |0009| In a preferred aspect of the invention, albumin fusion proteins include, but are not limited to, those described in Table 2 and the polynudeotidL'S encoding such proteins. |OOIO| The invention also encompasses pharmaceutical formulations comprising an albumin fusion protein of the invention and a pharmaceutically acceptable diluent or earner. 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,
nmH.ontmg or diagnosing a disease or disease symptom in a patient, preferably a mamma!, most preferably a human, comprising the step of
administering the pharmaceutical formulation to the patient.
[0011] 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 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.
|0012| In one embodiment, an albumin fusion protein described in Table 1 or 2 has extended shelf life.
|0013| 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 I.
(0014) 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
|()015| Figure IA-D shows the amino acid sequence of the mature form of human albumin (SEQ ID N0:l) and a polynudeotide encoding it (SEQ ID NO:2). Nucleotides 1 to 1755 of SEQ ID N0:2 encode the mature form of human albumin (SEQ ID NO:1). |0016| Figure 2 shows the restriction map of the pPPCOOOS cloning vector ATCC deposit PTA-3278.
[0017] Figure 3 shows the restriction map of the pSAC35 yeast S. ccrcvisiae expression vector (Sleep el al., BioTcchnology 8:42 (1990)). [0018] Figure 4 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 76). Proteins were serially diluted from 5e-7 to le-14 g/ml in DMEM/10% FBS and used to treat 1SRE-SEAP/293F reporter cells. Afler 24 hours supernatants were removed from reporter cells and assayed for SEAP activity. IFNb albumin fusion protein was purified from three stable clones: 293F/#2011, CHO/#2011 and NSO/#2053. Mammalian derived IFNb, Avoncx, came from Biogen and was reported to have a specific activity of 2 Oc5 HJ/ug.
[0019] Figure 5 compares the anti-proliferative activity of IFN albumin fusion protein encoded by CID 3165 (PIT) 3165 protein) and rccombmant IFNa (rIFHa) on Hs294T melanoma cells. The cells were cultured with varying concentrations of either CID 3165 protein or rlFNa and proliferation was measured by BrdU incorporation after 3 days of culture. CID 3165 protein caused measurable inhibition of'ccll proliferation at concentrations above 10 ng/ml with 50% inhibition achieved at approximately 200 ng/ml. (•) = CID 3165 protein, (*) = rlFNa. [OUZO] Figure 6 shows the effect of various dilutions of IFNa albumin fusion proteins on SF,AP activity in the ISRE-SEAP/293F reporter cells. One preparation of IFNa fused upstream of albumin (*) was tested, as well as two different preparations of IFNa fused downstream of albumin (•) and (•).
[00211 Figure 7 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 (sec Example 78). Per time point: first bar = Vehicle control, 2™1 bar = 30 ug/kg CID 2249 protein day 1 iv, third bar = 30 ug/kg CID 2249 protein day 1 sc, 4* bar = 300 ug/kg CID 2249 protein day 1 sc, 5lh bar = 40 ug/kg recombinant IFNa day 1, 3 and 5 sc.
(0022| Figure 8 shows the dose-response relationship of BNP albumin fusion proteins encoded by DNA comprised in constructs CID 3691 and 3618 (CID 3691 and 3618 protein) on activating cGMP formation in NPR-A/293F reporter cells (see Examples 80 and 81). Doth recombinant BNP (•), as well as, two different preparations of BNP fused upstream of albumin (O) and (•) were tested.
|0023| Figure 9 shows the effect of BNP albumin fusion protein on mean arterial pressure in spontaneously hypertensive rats (see Example 80). Vehicle (D), recombinant BNP protein (•), or BNP albumin fusion protein (O) were delivered via tail vein injection. Systolic and diastolic blood pressures were recorded by cuff-tail method
|0024| Figure 10 shows the plasma cGMP levels in eleven- to 12-week-old male C57/BL6 mice after intravenous injection of recombinant BNP protein (•) or BNP albumin fusion protein (O (see Example 80). cGMP levels were determined from plasma prepared from tail bleeds collected at several time points after intravenous injection.
|0025] Figure 11 shows the blood glucose levels in fasted ~8-wcck old diabetic db/db mice 24 hours after single administration of tandem GLP-l(7-36A8G)2x-HSA fusion (CID 3610) (O), monomer GLP-1(7-36A8G)-HSA fusion (A), or HSA alone (•). Blood glucose levels were measured by an oral glucose tolerance test. Tandem GI.P-l(7-36A8G)2x-HSA fusion (CID 3610) (O) had an unexpected more potent glucosc-normalizing activity than Ihe monomer GLP-!(7-36A8G)-HSA fusion (A) 24 hours after a single administration in fasted ~8 week old diabetic db/db mice.
DETAILED DESCRIPTION
Definitions
[0026] The following definitions are provided to facilitate understanding of certain terms used throughout this specification. |0027) 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).
(0028) 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.
|0029] 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). |0030| 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 O- linked glycosylalion); 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".
[0031] 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
|0032| 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 lilac sequence is shaven as SCQ 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 valiant 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 polynucleotide encoding these albumin fusion proteins. [0033] 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 denim 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 extra cellular 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 polynucleotide encoding these albumin fusion proteins.
[0034] 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 Andover 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
|0035| As staled 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.
[0036] 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 arc linked with one another by chemical conjugation.
[0037| 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 arc not limited to, proteins, polypeptides, peptides, antibodies, and biologies. (The terms peptides, proteins, and polypeptides are used interchangeably herein.) It is specifically contemplated that the term 'Therapeutic protein" encompasses antibodies and fragments and variants thereof. 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.
|003H| 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.
|0039| 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, GUM, GLP-2, PACAP-27, PACAP-28, VIP, CD4M33, secretin, glicentin, oxyntomodulin, PHM, IFNn, 1|-N(S, ANP, BNP, NGF, BDNF, GDNF, and somatostatin.
|0040| 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 (Lcbleu et al., PNAS USA, 73:3107-3111 (1976); dresser et a]., Nature, 251:543-545 (1974); and Johnson, Texas Reports Iliol Med, 35:357-369 (1977)). Each interferon hybrid albumin fusion protein can be used to treat, prevent, or ameliorate viral infections (e.g., hepatitis (eg., HCV); or HIV), multiple sclerosis, or cancer
[0041] 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 A/D hybrid is fused at the common Bglll restriction siie 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 A/D hybrid would comprise the amino acid sequence:
CULHQlHSLOSRRTLMLI^QMRX|lSLrvSCLKDRHDFGFFQEEFGNQFQKAETIPVU1EMIQQa:74LFTTKX)SSAA\VDEDU.DK:FCTELYQQL NDLEACVMQEERVGETPLMNX2D!JILAVKJCYFRRITLYLTEKKYSPCAWEVVRAEIMRSLSLSTNLQERLRRK.E (SEQ ID N0:99), wherein the X, is R or K and the X2 is A or V (see, for example. Construct ID #2875). In an additional embodiment, the A/D hybrid is fused at the common PvullI restriction site, wherein the N-tcrmina! 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 A/D hybrid would comprise the amino acid sequence: CDLPQTHSLGSRRTLMLLj\QMRX|ISLFSCLICDRHDFGFPQEEFGNQFQICAE'fIPVLHEMIQQlFNLFSTKDSSAAWDETLLDlCFYTELYQQL NDLEACVMQEERVGETPLMNX2DSILAVICKYFRRrn.YLTEKKYSPCAWEVVRAEIMRSLSLSTNLQERLRRKE (SEQ ID NO:100), wherein the Xi is R or K. and the second X; is A or V (sec, for example, Construct ID #2872). These hybrids are further described in U.S. Patent No, 4,414,510, which is hereby incorporated by reference in its entirety.
|0042| In an additional embodiment, the alpha-alpha hybrid portion of the intcrferon hybrid albumin fusion protein consists, or alternatively comprises, of interferon alpha A fused to intcrferon 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 intcrferon 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:
CDLPQrHSLGSRRTLMLLAQMRXISLFSCLKDRHDFGFPQEEFGNQFQKAETIPVLHEMlQQIFNLFSTKDSSAAWDETLLDKFYTELYQQL NDMEACVIQEVGVEETPLMNVDSILAVKKYFQRITLYLTEKKYSPCAWEVVRAEIMRSFSLSICIFQERLRRKE (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, which is hereby incorporated by reference in its entirety. 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/D hybrid is fused at the common PvuIII 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: CDI.PQTHSLGSRRTlJv1LLAQMRX,ISLFSCLlCDRHDFGFPQEEFGNQFQKAETIPVLHEMIQQIFNLFSTICDSSAAWDETLLDKFYTELYQQL NDLEX2X,X,X5QEVGVIESPLMYEDSILAVRKYFQRITLYLTEKKYSSCAWEVVRAEIMRSFSLSINLQK.RLKSICE (SEQ ID NO: 102), wherein the X, is R or K- and X2 through Xs is SCVM or VLCD (see, for example, Construct ID #2873). These hybrids are further described in U.S. Patent No. 4,414,510, which is hereby incorporated by reference in its entirety.
|0043] In another embodiment, the interferon hybrid portion of the intcrferon hybrid albumin fusion protein comprises an interferon be.ta-inlerferon 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 intcrferon beta-l fused to intcrferon alpha D (also referred to as intcrferon alpha-1). In a further embodiment, the beta-l/alpha D hybrid is fused wherein the N-terminal portion corresponds to amino acids 1-73 of interferon beta-l and the C-termmal 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:
MSYNLLGFLQRSSNFQCQKLLWQLNGRLEYCLKDRMNFDIPEEIK.QLQQFQKEDAALTIYEMLQN1FAIFRQDSSAAWDEDLLD1CFCTELY QQLNDLEACVMQEERVGETPLMNXDSILAVKKYFRRITLYLTEKKYSPCAWEWRAOIMRSLSLSTNLQERLRRKE (SEQ ID NO: 103), wherein X is A or V. These hybrids are further described in U.S. Patent No. 4,758,428, which is hereby incorporated by reference in its entirety. |U044| In another embodiment, the interferon hybrid portion of the intcrferon hybrid albumin fusion protein comprises an interferon alpha-mterfeion beta hybrid (herein referred to as a alpha-beta hybrid). For example, the alpha-beta hybrid portion of the interferon hybrid albumin fusion piolcm consists, or alternatively comprises, of interferon alpha D (also referred to as interferon alpha-1) fused to interferon beta-l. In a further embodiment, the alpha D/bcta-l hybrid is fused wherein the N-terminal portion corresponds to amino acids 1-73 of interferon alpha D and the C-lerminal portion corresponds to amino acids 74-166 of interferon beta-l. For example, this alpha D/beta-l hybrid would have an amino acid sequence:
MCDI.PETHSLDNRRTLMLLAQMSR1SPSSCLMDRHDFGFPQEEFDGNQFQKAPAISVLHEI.1QQIFNLFTTKDSSSTGWNETIVENLLANVY 1IQINHLKTVLEEICLEK.EDFTRGKLMSSLHL1CRYYGR1UIYLKAK.EYSHCAWTIVRVE1LRNFYFINRLTGYLRN (SEQ ID NO: 104). These hybrids are further described in U.S. Patent No. 4,758,428, which is hereby incorporated by reference in its entirety.
|()045] 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 In the amino acid sequence of the intcrferon hybrid. Such modifications to the interferon hybrid albumin fusion proteins may be made, for example, in improve levels of production, increase stability, increase or decrease activity, or confer new biological properties.
[0046) 'I"he above-described intcrferon hybrid albumin fusion proteins are encompassed by the invention, as are host cells and vectors containing polynudeotides encoding the polypcptides. 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 unfus^d r'ntf rferon hybrid molecule
[0047] 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 I (column 2).
|OU48| In one embodiment, IFN-beta-HSA fusions are used to inhibit the activity of Ebola virus and the SARS virus (Toronto-2 strain). For example, 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. Further, the pharmacokinetics 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 ng/ml against Ebola and 2 ng/ml against the SARS virus. Array analysis showed that CID 2053 protein and [FN-beta induce the expression of a similar set of genes and trigger the IFN-stimuIated 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, the terminal half-life was 36-40 hours. Administration of CID 2053 protein induced sustained increases in serum neopterin levels and OASI mRNA expression. |0049| In a further embodiment, IFN-alpha-HSA fusions are used to inhibit viral agents classified under Category A- Filo (Ebola), Arena (Pichendc), Category B- Toga (VEE) or Category C- Bunya (Punto toro), Flavi (Yellow fever, West Nile). For example, CPE inhibition, neutral red staining and virus yield assays were employed to evaluate the antiviral 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 antiviral activity was achieved against all the RNA viruses evaluated with a favorable safety index. The IC50 values ranged from |()050| In a further embodiment, IFN-alpha-HSA fusions are used to treat patients with chronic Hepatitis C infection (HCV). Interferon alpha, also known as interferon alfa or leukocyte inferon, is the standard of care for treatment of patients infected with HCV. The term "interferon alpha" refers to a family of highly homologous related polypeptides with anti-viral activity. The interferon alpha portion of the IFN-alpha-HSA fusion consists or alternatively comprises any interferon alpha or fragment thereof known in the art. Non-limiting examples of interferon alpha encompassed by the invention include, but are not limited to, the interferon alpha proteins disclosed in the Therapeutic protein column of Table 1. In particular embodiments, the interferon alpha portion consists or alternatively comprises interferon alpha-2a, interferon alpha-2b, interferon alpha-2c, consensus interferon, intcrferon alfacon-1, interferon alpha-nl, interferon alpha-n3, any commercially available form of interferon alpha, such as, for example, INTRON* A (Schering Corp., Kcnilworth, N.J.), ROFERON* A (Hoffman-La Roche, Nutley, N.J.), Berofor alpha inteferon (Boehringcr Ingelheim Pharmaceutical, Inc., Ridgefied, Conn.), OMN1FERON™ (Viragen, Inc., Plantation, FL), MULTIFERON™ (Viragen, Inc., Plantation, FL) WELLFCRON* (GlaxoSmithKline, l^ndon, Great Britian), INFERGEN* (Amgen, Inc., Thousands Oaks, CA), SUMIFERON* (Sumitomo, Japan), BELEROFON* (Nautilus Biotech, France) or any purified interfcron alpha product or a fragment thereof In additional embodiments, the interferon alpha portion of the IFN-alpha-HSA fusion protein may be modified by the attachment of chemical moieties. For example, the inteferon alpha portion may be modified by pegylation. Accordingly, in additional embodiments, the intcrferon alpha portion of the IFN-alpha-HSA fusion protein consists or alternatively comprises pegylated forms of interferon alpha-2a, 2b, or consensus interferon and include, but arc not limited to, a commercially available pegylated intcrferon alpha, such as, for example, PEG-INT'RON* (Schering Corp., Kenilworth, N.J.), PEGASYS* (Hoffman-La Roche, Nutley, N.J.), PEG-OtvfNIFERON™ (Viragen, Inc., Plantation, FL) or a fragment thereof However, as used herein, "IFN-alpha-HSA" fusions refers to the HSA fused to any of the interfcron alpha proteins known in the art or a fragment thereof. [0051| Patients infected with HCV may fall within two categories based on previous exposure to an interferon regimen for treatment of the HCV infection. "Naive patients" are those patients who have never been treated with an interferon regimen. "Experienced patients" are those patients who have been treated or are currently being treated with an interferon regimen. "Non-responders" are experienced patients who have been previously treated with an intcrferon regimen but have failed to meet the primary endpoint of treatment such as an early viral load reduction (EVR) or an end-of-treatment response (ETR). However, as used herein, an "HCV patient" refers to a patient who is infected with HCV and who is either
naive, experienced, or a non-responder.
|0052] In addition, the Hepatite C virus can be classified intn four genotypes, genotype I, 2, 3, or 4. Generally, the Hepatitis C virus that infects ar, HCV patient comprises n single genotype However, the Hepatitis virus can comprise a combination of two or mom genotypes In addition, the genotype of Hepatitis C virus may also be a variant of one of the known HCV genotypes- In a further embodiment, the Hepatitis C virus of the HCV patient is genotype I or a variant thereof. However, as used herein, "HCV" refers to the Hepatitis C virus of any genotype, or combination or variants thereof.
[OOS3| The standard treatment regimen for patients with HCV involves treatment with inlerferon alpha in combination with an antiviral agent, such as, ribavirin. In general, the interferon alpha is administered daily, twice-a-week, or weekly and the ribavirin is administered daily. However, recent studies have also used inteferon alpha in combination with other antiviral agents blown in the art for the treatment of HCV. Thus, in a further embodiment the IFN-alpha-HSA fusion may be administered to :he HCV patient either alone or in combination with an antiviral agent, such as, for example, ribavirin.
(0054) As noted above, pharmokinetics of the CID 3165 protein support a dosing schedule of once every 2-4 weeks or greater. Thus, in a further embodiment, the HCV patients are treated with an IFN-alpha-HSA fusion by administration once every 2-4 weeks alone or in combination with an effective amount of an antiviral agent. In a preferred embodiment, Ihe HCV patients are treated with an IFN-alpha-HSA fusion by administration once every 2-4 weeks in combination with an effective amount of an antiviral agent. In an additional preferred embodiment, the IFN-alpha-HSA fusion is administered to the HCV patient once every 4 weeks. In an additional preferred embodiment, the IFN-alpha-HSA fusion is administered to the HCV patient more than once every 4 weeks. In additional embodiments, the IFN-alpha-HSA fusion is adminstered once every 4 weeks or more to an HCV patient, wherein the treatment also includes administration of an effective amount of an antiviral agent. |005S] In a another embodiment, IFN-alpha-HSA fusions may be used as a low-dose monotherapy for maintenance therapy of HCV. In a further additional embodiment, IFN-alpha-HSA fusions may used in combination with ribavirin and one or more olher antiviral agents for the treatement of HCV. Alternatively, in another embodiment, IFN-alpha-HSA fusions may be used in combination with one or more antiviral agents, other than ribavirin, for the treatment of HCV.
|0056] In an additional embodiment, IFN-alpha-HSA fusions may be used for the treatment of other viral infections. For example, in one embodiment, IFN-alpha-HSA fusions may be used for the treatment of Hepatitis B (HBV). In an additional embodiment, IFN-alpha-HSA fusions may be used for the treatment of Human Papilloma Virus (HPV). in a further embodiment, IFN-alpha-HSA fusions may be used in the treatment of cancer, including, but not limited to hairy cell leukemia, malignant melanoma, follicular lymphoma, chronic myelogenuus leukemia, AIDS related Kaposi's Sarcoma, multiple myeloma, or renal cell cancer.
[0057] In another embodiment, GLP-i-HSA fusions are used to regulate blood glucose levels in diabetic patients, (n a further embodiment, tandem fusions of wild-type or mutant GLP-1 are used to regulate blood glucose levels in diabetic patients. l;or example, the ability of monomer GLP-1(7-36A8G)-HSA and tandem GLP-1(7-36A8G)-HSA (CID 3610) fusions to regulate the blood glucose levels were evaluated utilizing an oral glucose tolerance test (1 gram glucose/kg by oral gavage), following subcutaneous injection of GLP-1-HSA protein in ~B week old diabetic db/db mice. This glucose tolerance test consisted of the subcutaneous injection of a GLP-1-HSA fusion followed by administration of 1 gram glucose/kg by oral gavage. Fasted diabetic db/db mice were administered equimolar doses (100 and 171 nrnol/kg) of either the monomer or tandem GLP-I-HSA fusion protein and oral glucose tolerance tests were performed 6 or 24 hours after a single administration. Quite surprisingly and unexpected, the tandem GLP-1 (7-36A8G)-HSA fusion (CID 3610) significantly reduced blood glucose at 6 hours afler injection when compared to the monomer GI.P-1(7-36A8G)-HSA fusion. In addition, the difference between monomer GLP-1(7-36A8G)-HSA and tandem GLP-I(7-36A8G)-HSA (CID 3610) fusions were even more dramatic when diabetic db/db mice were evaluated 24 hours afler injection. As shown in Figure 11, the tandem GLP-1(7-36A8G)-MSA fusion (CID 3610} (O) possessed a unexpectedly potent glucose-normalizing activily while fasting blood glucose levels for the single GLP-1(7-36A8G)-1ISA (A) fusion was similar to animals administered HSA alone (•) and clearly were diabetic in nature. |00581 As used herein, "therapeutic activily" or "activily" mjy 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. 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 I.
(0059) Therapeutic proteins corresponding to a Therapeutic protein portion of an albumin fusion protein of the invention, such as cell surface and secretory proteins, arc often modified by the attachment ofone 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 O-linked oligosaccharides, which are attached to serine or threonine residues; and glycosylation characterized by N-l:nked oligosaccharides, which arc attached to asparagine residues in an Asn-X-Scr or Asn-X-Thr sequence, where X can be any amino acid except proline. N-acetylneuramic acid (;ilso known as sialic acid) is usually the terminal residue of bath N-linked and O-linked oligosaccharides. Variables such as protein structure and
cell type influence the number and nature of the carbohydrate units within the chains at different glycosylalion sites. Glycosylalion isomers are also common at the same site within a given cell type.
10060] Therapeutic proteins corresponding to a Therapeutic protein portion uf in albumin fusion protein of the invention, a: -,vel! 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 rccombinant proteins may be produced by expressing the proteins in host cells that will not glycosyJate them, e.g. in E. culi or glycosylation- |0061] 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., Denvcnt). Exemplary nucleotide sequences of Therapeutic proteins which may be used to derive a polynucleotide of the invention are shown in column 7, "SEQ !D 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 pulymieleotide 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 (he sequence shown as SEQ ID NO:X to derive the construct described in the same row. For example, if SEQ JD 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.
(0062| Additional Therapeutic proteins corresponding to a Therapeutic prutein portion of an albumin fusion protein of Ihe invention include, but arc 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.
|0063| Table I 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 lo an individual Therapeutic protein molecule, or lo 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 lest the therapeutic and/or biological activity ofa Therapeutic protein:X or an albumin fusion protein comprising a Therapeutic protein X (or fragment thereof) portion. Each of the references cited in the "Exemplary Activity Assay" column are herein incorporated by reference in their entireties, particularly with respect to the description of the respective activity assay described in the reference (see Methods section therein, for example) for assaying the corresponding biological activity set forth in the "Biological Activity" column of Table 1. The fourth column, "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 albumm fusion protein comprising, or alternatively consisting of Ihe referenced Therapeutic I'rotein X (or fragment thereof) portion.
(Table Removed)
|0064| Table 2 provides a non-exhaustive list of polynucleotide 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;!,.; invention. The Construct IDs may be ujJ to identify polynucleotide which encode albumin fusion proteins comprising, or alternatively consisting of, a Therapeutic protein portion corresponding to a given Therapeutic ProteinrX 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.
|0065] 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 5. 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:IISA. 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.
|0066| 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 I ISA 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 ammo 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 NOY 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.
|0067) 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 polynucicotide 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 1 herapeutic protein.
|OOY>8| 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 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 uilo the appropriate vector.
|0069| 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-cstablishfd in tin- art Additional useful primer sequences cnuld readily be envisioned and utilized by
those of ordinary skill in the art.
|0070| 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.
|0071] As shown in Table 3, certain albumin fusion constructs disclosed in this application have been deposited with the ATCC®.
(Table Removed)
sible 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. [0073] In a further embodiment of the invention, an "expression cassette" comprising, or alternatively consisting of one or more of (1) a polynuclcotide 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 B1, and/or restriction enzyme digestion.
|0074| 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
[0075| 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.
(0076) 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.
|0077| 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 multimcrize, ability to bind a ligand) may still be retained. For example, the ability of polypeptides with N-tcrminal 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 polypcptide 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.
|0078| 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 polypcptide (i.e., a Therapeutic protein referred to in Table I, 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-temiinal 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 polypcptides are also encompassed by the invention.
100791 In addition, fragments of senim 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.
|OQ80| 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 arc also encompassed by the invention.
J0081J 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-tcrminal residues of a reference polypeptide retains Therapeutic activity can readily be determined by routine methods described herein and/or otherwise known in the art.
|()082] 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 arc also encompassed by the invention.
|0083| In addition, the present invention provides polypcptides 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 01 an albumin protein portion of an albumin fusion protein encoded by a polynucleotide or albumin fusion construct described in Table 2). In purticular, C-teiminal 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 I. Polynucleotides encoding these polypeptides are also encompassed by the invention. In particular, C-tcrminal 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. I'olynucleotides encoding these polypeptides are also encompassed by the invention.
|0084| 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.
[0085| In addition, any of the above described N- or C-tenninal 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 I, 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 (eg., SUQ ID NO:1), or an albumin protein partion 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 nnlynucleotide or albumin fusion construct of the invention) where n and m are integers as described above
Polynuclcotides encoding these polypeptides ate also encompassed by the invention.
10086] The present application is also directed lo proteins containing polypeptides at least 80%, 85%, 90%. 95%, 96%, 97%, 98% or 99%
identical to a reference polypeplide sequence (e.g., a Therapeutic protein referred to in Table I, 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.
[00871 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,
[0088J 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
(00891 "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. |0090| 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, idenlical to the amino acid sequence of the Therapeutic protein corresponding la aTherapcutic 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.
(0091 ] 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 ammo acid sequence of a Therapeutic protein portion of an albumin fusion protein encoded by a polynucleotide or albumin fusion construct described in Table ! 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 i 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 pulypep'.ides 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 oX 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 lo those of skill in the art (see, for example, Ausubel, P.M. et al., eds., 1989 Current protocol in Molecular Biology, (iieen 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 arc also encompassed by the invention.
|0092| By a polypeptide having an amino acid sequence at least, for example, 95% "identical" to a query amino acid sequence, it is intended thai the amir.o 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 ammo 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 anolhur 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.
[0093] 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 piotein of the invention or a fragment thereof (such as a Therapeutic protein portion of the albumin fusion protein or an albumin portion of the albumin fusion protein), can be determined conventionally using known computer programs. A preferred method for determining the best overall match between a query sequence (a sequence of the present invention) and a subject sequence, also referred to as a global sequence alignment, can be determined using the FASTDB computer program based on the algorithm of Brutlag et al. (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=l, Joining Penalty=20, Randomization Group Length=0, Cutoff Score=l, Window Size=sequence length, Gap Penalty=5, Gap Size Penalty=4).05, Window Size-500 or the length of the subject amino acid sequence, whichever is shorter.
[0094| 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-tcrmini 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.
I0095J 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 frsl 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 FASTDD 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 (if 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 arc nut matched/aligned with Ihe query sequence are manually corrected for. No other manual corrections are to made for the purposes of the present invention.
10096) 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, blasln, btastx, tblastn and tblastx (Karlin el al, Proc. Natl. Acad. Sci. USA 87: 2264-2268 (1990) and Altschul, J. Mol. Evol. 36: 290-300 (1993), fully incorporated by reference) which are tailored for sequence similarity searching.
100971 The approach used by the BLAST program is to fust consider similar segments between a query sequence and a database sequence, then It; 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 el al.. (Nature Genetics 6: 119-129 (1994)) which is fully incorporated by reference. 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^/ «/.. Proc. Natl. Acad. Sci. USA 89: 10915-10919 (1992), fully incorporated by reference). 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 penally); R=10 (gap extension penalty); wink=l (generates word hits at every wink1"1 position along Ihe ijueiy). and gapw-16 (sets the window width within which gapped alignments are generated). The equivalent Blastp parameter settings were Q=9; R=2; wink=l; and gapw=32. A Bestfit comparison between sequences, available in the GCG package version 10.0, uses DNA parameters GAP=50 (gap creation penally) and LEN=3 (gap extension penalty) and the equivalent settings in protein comparisons are GAP=8 and LEN=2. |0()98| The polynuclcotidc 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 polypeplidp Nucleoiide 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 ammo acids arc 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 codoni in llic human mRNA to those piefcrreJ by a bactuUI host, such as, yeast or E. coli).
[0099| 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. |OOH>()] In an alternative embodiment, a codon optimized polynucleotide which encodes a Therapeutic protein portion of an albumin fusion protein docs 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. [0100) 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.
(0101] 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 (1085)). 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 niutagenesis techniques or by direct synthesis.
|0102| Using known methods of protein engineering and recumbinant 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 ammo-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 ct al., J. Biotechnology 7:199-216 (1988).)
(0103) Moreover, ample evidence demonstrates that variants often retain a biological activity similar to that of the naturally occurring protein. For example, Gayle and eoworkers (J. Biol. Chem. 268:22105-22111 (1993)) conducted extensive mutational analysis of human cytokine IL-la. They used random niutagenesis 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 "[mjost of the molecule could be altered with little effect on either [binding or biological activity]." In fact, only 23 unique arnino acid sequences, out of more than 3,500 nucleotide sequences examined, produced a protein that significantly differed in activity from wild-type.
|0104| 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 hind antibodies which recognize the secreted form will likely he retained when less than the majority of the residues of the secreted form are removed from the N-lcrminus or C-termmus. Whether a particular polypeptide lacking N- or C-tcrminal residues of a protein retains such immunogenic activities can readily be determined by routine methods described herein and otherwise known in the art.
|OIU5| 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 I'herapeutic 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. Holynucleotides encoding such variants are also encompassed by the invention.
|0106| 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 He; replacement of the hydroxyl residues Ser and Thr; replacement of the acidic residues Asp and Cilu; replacement of the amide residues Asn and Gin, replacement of the basic residues Lys,
(0108| 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.
10109] The second strategy uses genetic engineering to introduce amino acid changes at specific positions of a cloned gene to identify regions critics.! for protein function. For example, site directed mutagenesis or ataninc-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 he tested for biological activity.
|0110] As the authors state, these two strategies have revealed that proteins are surprisingly tolerant of amino acid substitutions. The authors turthcr 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, Uu and lie; 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 amirio 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) polypepttdes 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.
|!>111| 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 immunogenic activity. See Pinckard et al., Clin. Exp. Immunol. 2:331-340 (1967), Robbins et al., Diabetes 36: 838-845 (1987); Cleland et al., Crit. Rev. Therapeutic Drug Carrier Systems 10:307-377 (1993).
[0112] 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, amina 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.
|UI 13] 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 polypepltdes may be modified by either natural processes, such as post-lranslational 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 peptiile 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 polypcptide 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 posttranstation natural processes or may be made by synthetic methods. Modifications include acetylation, acytation, ADP-nbosylation, amidation, covalent attachment of flavin, covalent attachment of a herne moiety, covalent attachment of a nucleotide or nucleotide derivative, covalent attachment of a lipid or lipid derivative, covalcnt attachment of phosphotidylinositol, cross-linking, cyclization, disulfide bond formation, dcmethylation, formation of covalent cross-links, formation of cysteine, formation of pyroglutamate, formylation, gamma-carboxylation, glycosylation, GP1 anchor formation, hydroxylation, iodination, melhylalion, myristylation, oxidation, pegylation, proteolytic processing, phosphorylation, prenylation, racemization, selenoylation, sulfation, iransfer-RKA mediated addition ofamino acids to proteins such as arginylatinn, and ubiquilinaticn. (See, for instance, PROTEINS - STRUCTURE AND MOLECULAR PROPERTIES, 2nd Ed., T. E. Creighton, W. 11. Freeman and Company, New York (1993); POST-TRANSLATION A L COVALENT MODIFICATION OF PROTEINS, B. C. Johnson. Ed., Academic Press. New York, pgs. 1-12 (1983); Seifleret al., Meth. Enzymo!
182:626-646(1990); Rattan et al, Arm. N.Y. Acad. Sci. 663:48-62 (1992)).
functional activity
|0!M] "A polypeptide having functional activity" refers to a polypcptide capable of displaying one or mere 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 polypcptides of the invention, and ability to bind to a receptor or ligand for a polypeptide.
|0115] "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).
(0116] 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.
|0117] In additional preferred embodiments, the albumin fusion protein of the invention has an increased plasma stability compared to the Therapeutic protein portion (or fragment or variant thereof) in an unfused state. Plasma stability of the albumin fusion protein of the invention or of the unfused Therapeutic protein portion (or fragment or variant thereof) can be assayed using or routinely modifying assays known in the art. [0118] 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. (0119) 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, immunodiiTusion 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.
|0120| 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 hlotting. 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 subslrate(s) of the Therapeutic polypeptide corresponding to the Therapeutic protein portion of the fusion can be routinely assayed using techniques known in the art.
|0121| 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.
|OI22| 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.
|OI23| 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 he 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.
(II174I Assays for the ability of the albumin fusion proteins (e.g.. comprising at least a fragment or variant of an antibody that hinds 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. Nail. 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)) (each of these references is incorporated herein in its entirety by reference). 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. |U125| 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.
|0126| Immunoassays which can be used to analyze (immunospccific) 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 irnmunosorbent assay), "sandwich" immunoassays, immunoprecipitation assays, prccipitin reactions, gel diffusion precipitin reactions, immunodiffusion assays, agglutination assays, complement-fixation assays, immunoradiometric assays, fluorescent immunoassays, and protein A immunoassays, to name but a few. Such assays are routine and well known in the art (see, eg., Ausubel et al, eds, 1994, Current Protocols in Molecular Biology, Vol. 1, John Wiley & Sons, Inc., New York, which is incorporated by reference herein in its entirety). Exemplary immunoassays are described briefly below (but are not intended by way of limitation).
|OI 27] 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 deoxycholatc, 0.1% SDS, 0.15 M NaCI, 0.01 M sodium phosphate at pH 7.2, 1% Trasylol) supplemented with protein phosphatase and/or protease inhibitors (e.g., EDTA, PMSF, aprotinin, sodium vanadate), adding the albumin fusion protein of the invention (e.g., comprising at least a fragment or variant of an antibody that binds a Therapeutic protein) to the cell lysate, incubating for a period of time (e.g., 1 to 4 hours) at 40 degrees C, adding sepharose beads coupled to an anti-albumin antibody, for example, to the cell lysate, incubating for about an hour 01 more at 40 degrees C, washing the beads in lysis buffer and rcstispending the beads in SDS/sample buffer. The ability of the albumin fusion piotein 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-clcaring 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.
|OI2Jt] Western blot analysis generally comprises preparing protein samples, electrophoresis of the protein samples in a polyacrylamide gel (e.g., 8%- 20% SDS-PAGE depending on the molecular weight of the antigen), transferring the protein sample from the polyacrylamide gel to a membrane such as nitrocellulose, PVDF or nylon, blocking the membrane in blocking solution (e.g., PBS with 3% BSA or non-fat milk), washing the membrane in washing buffer (e.g., PBS-Tween 20), applying the albumin fusion protein of the invention (diluted in blocking buffer) to the membrane, washing the membrane in washing buffer, applying a secondary antibody (which recognizes the albumin fusion protein, e.g., an anti-human serum albumin antibody) conjugated to an enzymatic substrate (e.g., horseradish peroxidase or alkaline phosphatase) or radioactive molecule (eg., "? or I2SI) 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.
101291 ELISAs comprise preparing antigen, coating the well of a 96-well microtitcr plate with the antigen, washing away antigen that did not hind the wells, adding the albumin fusion protein (e.g., comprising at least a fragment or variant of an antibody that binds a Therapeutic prolein) 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 docs not have to be conjugated to a delectable compound; instead, a second antibody (which recognizes albumin fusion protein) conjugated to a detectable compound may be added to the well. Further, instead of coaling 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 Dther variations of ELISAs known in the art. For further discussion regarding ELISAs see, eg., Ausubel et al, eds, 1994, Current Protocols in
Molecular Biology, Vol. 1, John Wiley & Sons, Inc., New York at 11.2.1.
|U130] The binding affinity of an albumin fusion protein to a protein, antigen, or epitope and the off-rate of an albumin fusion protcin-protcin/antigeii/epitopc 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 12il) 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., 5H or I2!f) 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.
[OI31| hi 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.
[0132| 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 IO"2 M, IO'2 M, 5 X 10° M, 10"3 M, 5 X IO"4 M, 10"1 M. More preferred binding affinities include those with a dissociation constant or Kd less than 5 X 10"5 M, 10s M, 5 X IO6 M, 10"'M, 5 X 10 ' M, 10' M, 5 X 10"' M or 10' M. Even more preferred binding affinities include those with a dissociation constant or ltd less than 5 X 10"' M, 10"' M, 5 X 10'° M, 10"'° M, 5 X 10" M, 10"" M, 5 X 10 l2 M, >c*" M, 5 X 10"" M, 10"'3 M, 5 X 10'" M, 10"" M, 5 X 10'" M, or IO"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 arc within the scope of the invention.
Albumin
|0133| 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.
|0134| An additional embodiment comprises al 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.
|013S| The terms, human serurn 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).
[0136| 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, El' 201 239, EP 322 094 WO 97/24445, WO95/23857) especially the mature form of human albumin as shown in Figure 1 and SEQ ID NO; I, or albumin from other vertebrates or fragments thereof, or analogs or variants of these molecules or fragments thereof. [0137| In preferred embodiments, the human serum albumin protein used in the albumin fusion proteins of the invention contains one or both of ihe following sets of point mutations with reference to SEQ ID NO: 1: Lcu^lOV to Ala, Leu-408 to Val, Val-409 to Ala, and Arg-410 to Ala; or Arg-410 to A, Lys^ll3 to Gin, and Lys-414 to Gin (see, e.g., International Publication No. WO95/23857, hereby incorporated in its entirety by reference herein). 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 recombinanl albumin fusion proteins expressed in yeast host cells.
|01381 As used herein, a portion of albumin sufficient to prolong the therapeutic activity or plasma stability or shelf-life of the Therapeutic protein refers to a portion of albumin sufficient in length or structure to stabilize or prolong the therapeutic activity or plasma stability of the protein so that the shelf life or plasma stability of the Therapeutic protein portion of the albumin fusion protein is prolonged or extended compared to the shelf-life or plasma stability in the non-fusion state. The albumin portion of the albumin fusion proteins may comprise the full length of the HA sequence as described above, or may include one or more fragments thereof that are capable of stabilizing or prolonging the therapeutic activity. Such fragments may be of 10 or more arnino 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. l-'or instance, one or more fragments of HA spanning the first two iinmunoglobulm-like domains may be used. In a preferred embodiment, the HA fragment is the mature form of HA.
[0139] The albumin portion of the albumin fusion proteins of the invention may be a variant of normal IIA. The ITieupeutic piotein 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-immunogcnic properties of albumin, or the active site, or active domain which confers the therapeutic activities of the Therapeutic proteins.
[0140| 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. [01411 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 N0:l), domains 1 and 2 (1-387 of SEQ ID N0:l), domains 2 and 3 (195-585 of SEQ ID NO: 1) or domains 1 and 3 (amino acids 1-194 of SEQ ID N0:l 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 mter-subdomain linker regions comprising residues Lysl06 to Glul 19, Glu292 to Val315 and Glu492 to AlaSl 1.
[0142| 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
|0143| 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
|0144| The basic antibody structural unit is known to comprise a tetramer. Each tetramer is composed of two identical pairs of polypeptide chains, each pair having one "light" (about 25 kDa) and one "heavy" chain (about 50-70 kDa). The amino-tenninal 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, IgG, IgA, and IgE, respectively. See generally. Fundamental Immunology Chapters 3-5 (Paul, W., ed., 4th ed. Raven Press, N.Y. (1998)) (incorporated by reference in its entirety for all purposes). The variable regions of each light/heavy chain pair form the antibody binding site.
[0145) Thus, an intact IgG antibody has two binding sites. Except in ^functional or bispecific antibodies, the two binding sites are the same. |OI46| 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 C'DRs. 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 epilope. From N-terminal to C-tcrminal, both light and heavy chains variable regions comprise the domains FR1, CDRI, FR2, CUK2, 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 Kahat Sequences of Proteins of Inimunological Interest (National Institutes of Health, Bethesda, Md. (1987 and 1991)), or Chothia & Lesk J Mol. Rial. 196.901-917 (1987); Chothia et al. Nature 342:878-883 (1989).
[0147| As used herein, "antibody" refers to imrmmoglobulin 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
[0148) 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
OM9| 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, mtirine (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 aminn 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.
|0150| The antibody molecules that bind to a Therapeutic protein and that may correspond to a Therapeutic protein portion of an albumin fusion protein of the invention can be of any type (e.g., IgG, IgE, IgM, IgD, IgA and IgY), class (e.g., IgGl, IgG2, IgG3, [gG4, IgAI 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 arc 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.
|01511 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, CHI, CI12, and CH3 domains.
[1)152) 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, tnspecific or of greater multispecificity. Multispecific antibodies may be specific for different epitopcs of a Therapeutic protein or may be specific for both a Therapeutic protein as well as for a heterologous epitope, such as a hcterologous polypeptide or solid support material. See, e.g., PCT publications WO 93/17715; WO 92/08802; WO 91/00360; WO 92/05793; Tun, 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). |01S3] 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 hybridornas or linking of Fab' fragments. See, e.g., Songsivilai & Lachmann C/i'n. Exp. Immunol: 79: 315-321 (1990), Kostelny et al. J Immunol. 148:1547 1553 (1992). In addition, bispecific antibodies may be formed as "diabodies" (Holhger et al. "'Diabodies': small bivalent and bispecific antibody fragments" PNAS USA 90:6444-6448 (1993)) or "Janusins" (Traunccker et ai. "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)).
|OI54| 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 nudeotidc 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, VI.CDR1, 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.
|0155] Antibodies that bind to a Therapeutic protein and that may correspond to a Therapeutic protein portion of an albumin fusion protein may lie 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 al 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.
|01S6| 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 polypeplides 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 embodiment, antibodies that bind to a Therapeutic protein and that may correspond to a Therapeulic 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
lhan 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 tne 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 immunopenic 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
[0157| Further included in the present invention are antibodies which bind polypeptides encoded by polynucleotides which hybridise to a polynuclcotide 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° M, 10' M, 5 X 10' 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' M, 10""M, 5 X 10' M, 10' M, 5 X 10"' M or 10'" M. Even more preferred binding affinities include those with a dissociation constant or Kd less than 5 X 10"' M, 10' M, 5 X lO'10 M, 10'° M, 5 X 10'" M, 10'" M, 5 X 10'n M, 10-'2 M, 5 X 10'13 M, 10"IJ M, 5 X 10"" M, 10'" M, 5 X 10'ls M, or I0"l! 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.
[0158] 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%.
|0159] 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/lhreonine) 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 prevent ing receptor activation compared to an un-fused fragment or variant of the antibody that binds the Therapeutic protein. |0160| 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. 'ITicsc antibodies may act as receptor agonists, i.e., potentiate or activate either all or a subset of the biological activities of the ligand-medialed receptor activation, for example, by inducing dimerization of the receptor. The antibodies may he 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. Immunoi. 161(4): 1786-1794 (1998); Zhu et al., Cancer Res. 58(I5):3209-32I4 (1998); Yoon et al., J. Immunol. 160(7):3170-3179 (1998); Prat et al., J. Cell. Sci. Ill (Pt2):237-247 (1998); Pitard et al., J. Immunol. Methods 205(2): 177-190 (1997); Liautard ct al., Cytokme 9(4):233-241 (1997); Carlson et al., J. Biol. Chem. 272(17):! 1295-11301 (1997); Taryman et al., Neuron 14(4):755-762 (1995); Mullcr et al.. Structure 6(9):1153-1167 (1998); Bartunek et al., Cytokine 8(1):14-20 (1996) (which are all incorporated by reference herein in their entireties). 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.
|016I] 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 immimoassays 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); incorporated by reference herein in its entirety. 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.
10162] 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 glycosylalion, acetylation, pcgylation, 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
|0163] 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.
|0164| Monoclonal antibodies can be prepared using a wide variety of techniques known in the art including the use of hybridoma, rccombinant, and phage display technologies, or a combination thereof. For example, monoclonal antibodies can be produced using hybndoma 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 cd 1988); Hammerling, et al, in: Monoclonal Antibodies and T-Cell Hybridomas 563-681 (Elscvicr, N.Y., 1981) (said references incorporated by reference in their entireties). The term "monoclonal antibody" as used herein is not limited to antibodies produced through hybridoma technology. The term "monoclonal antibody" refers to an antibody that is derived from a single clone, including any eukaryotic, prokaryotic, or phage clone, and not the method by which it is produced.
|0165] 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 polypcptide of the invention. Ascitcs fluid, which generally contains high levels of antibodies, can be generated by immunizing mice with positive hybridoma clones.
(0166) Accordingly, the present invention provides methods of generating monoclonal antibodies as well as antibodies produced by the method comprising culluring 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.
|OI67| Another well known method for producing both polyclonal and monoclonal human B cell lines is transformation using Epstein Barr Virus (F.QV). Protocols for generating EBV-transformed B cell lines are commonly known in the art, such as, for example, the protocol outlined in Chapter 7.22 of Current Protocols in Immunology, Coligan et al., Eds., 1994, John Wiley & Sons, NY, which is hereby incorporated in its entirety by reference. The source of B cells for transformation is commonly human peripheral blood, but B cells for transformation may also be derived from other sources including, but not limited to, lymph nodes, tonsil, spleen, tumor tissue, and infected tissues. Tissues are generally made into single cell suspensions prior to EBV transformation. Additionally, steps may be taken to either physically remove or inactivate T cells (e.g., by treatment with cyclosporin A) in B cell-containing samples, because T cells from individuals seropositivc for anti-EBV antibodies can suppress B cell immortalization by EBV.
|0168] 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 SVR-1492). Physical signs of EBV transformation can generally be seen towards the end of the 3-4 week culture period. By phase-coiiiraii microscopy, transformed cells may appear large, clear, hairy and tend tc 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.
|0169] Antibody fragments which recognize specific epitopes may be generated by known techniques. For example, Fab and F(abr)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 CHI domain of the heavy chain.
10170] 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 murme). 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 Ml3 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 184:177-186 (1995); Kettleborough et al., Eur. J. Immunol. 24:952-958 (1994); Persic ct 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; each of which is incorporated herein by reference in its entirety.
|0171| 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) (said references incorporated by reference in their entireties).
|0172| 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 al., BioTechniques 4:214 (1986); Gillies et al., (1989) J. Immunol. Methods 125:191-202; U.S. Patent Nos. 5,807,715; 4,816,567; and 4,816397, which are incorporated herein by reference in their entirety. Humanized antibodies are antibody molecules from non-human species antibody that binds the desired antigen having one or more complementarity determining regions (CDRs) from the non-human species and a framework regions from a human immunoglobulin molecule. 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 an, e.g., by modeling of the interactions of the CDR and framework residues to identify framework residues important for antigen binding and sequence comparison to identify unusual framework residues at particular positions. (See, e.g., Queen et al., U.S. Patent No. 5,585,089; Riechmann et al., Nature 332:323 (1988), which are incorporated herein by reference in their entireties.) Antibodies can be humanized using a variety of techniques known in the art including, for example, CDR-grafting (EP 239,400; PCT publication WO 91/09967; U.S. Patent Nos. 5,225,539; 5,530,101; and 5,585,089), veneering or resurfacing (EP 592,106; EP 519,596; Padlan, Molecular Immunology 28(4/5):489^98 (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).
|0173| 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 immunoglobulm sequences. See also, US. Patent Nos. 4,444,887 and 4,716,111; and PCT publications WO OX/46645, WO 98/50433 WO 98/24893, WO 98/16654, WO 96/34096, WO 96/33735, and WO 91/10741; each of which is incorporated herein by reference in its entirely. (0174| 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 arc then bred to produce homozygous offspring which express human antibodies. The transgenic mice arc 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 therapcutically 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 %/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, which are incorporated by reference herein in their entirety. 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.
|0175| 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 a!., Bio/technology 12:899-903 (1988)).
Polynucleotides Encoding Antibodies
[0176] 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.
|0177| 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 oligonuclcotides (e.g., as described in Kutmeier et a!., 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 ligatecl oligonuclcotidcs by PCR.
|()178| 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 Ihe 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).
|OI79] 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, which arc both incorporated by reference herein in their entireties ), to generate antibodies having a different amino acid sequence, for example to create amino acid substitutions, deletions, and/or insertions.
|018l)| In a specific embodiment, the amino acid sequence of the heavy and/or light chain variable domains maybe 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 rei-nmbinant ON A 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. Diol. 278: 457-479 (1998) for a listing of human framework regions). Preferably, the polynucleotide generated by (he 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 disulflde bond to generate antibody molecules lacking one or more intrachain disulfidc bonds. Other alterations to the polynucleotide are encompassed by the present invention and within the skill of the art. |0181) 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 ct 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 differenl 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.
|0182) Alternatively, techniques described for the production of single chain antibodies (U.S. Patent No. 4,946,778; Bird, Science 242:423- 42 11988), 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
|0183) 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 riolyniicleottde that encodes the antibody. Once a polynucleolide 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 rccombinant DNA technology using techniques well known in the art. Thus, methods for preparing a protein by expressing a polynucleotide containing an antibody encoding nudeotide 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. 'Itiese methods include, for example, in vitro reeombinanl DNA techniques, synthetic techniques, and in vivo genetic recombination. The invention, thus, provides replicable vectors comprising a nuclcotide 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.
|01K4| 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 hetcrologous 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 imniunogluhulin molecule, as detailed below.
(01K5| A variety of host-expression vector systems may be utilized to express the antibody molecules of Ihe invention. Such host-expression systems represent vehicles by which the coding sequences of inlerest 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. subtiiis) 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 nuisaic 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., melallothionein promoter) or from mammalian viruses (e.g., the aderiavirus late promoter; [he vaccinia virus 7.5K promoter). Preferably, bacterial cells such as Eschcrichia coli, and more preferably, eukaryotic cells, especially for the expression of whole recembinanl antibody molecule, arc 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 (Focckinget al., Gene 45:101 (1986); Cockett et al., Bio/Technology 8:2 (1990)).
|0186| In bacterial systems, a number of expression vectors may be advantageously selected depending upon the use intended for the aniibody 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 aniibody 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 (Rutheret al., EMBO J. 2:1791 (1983)), in which the antibody coding sequence may be ligaled individually into the vector in frame with the lac Z coding region so that a fusion protein is produced; plN veciors (Inouye & Inouye, Nucleic Acids Res. 13:3101-3109 (1985); Van Hecke & 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.
[0187] 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).
[0188| 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 Ihe 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 Bittneretal., Methods in Enzymol. 153:51-544(1987)).
[0189( '" addition, a host cell strain may be chosen which modulates the expression of the inserted sequences, or modifies nnd 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 ceil 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, CRI.7030 and Hs578Bst.
|0190| 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 :-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. |OI91| A number of selection systems may be used, including but not limited to the herpes simplex virus thymidine kinase (Wigler ct al., Cell 1 !:223 (1977)), hypoxanthine-guanine phosphoribosyltransferase (Szybatska & Szybalski, Proc. Natl. Acad. Sci. USA 48:202 (1992)), and adenine phosphuribnsyllransferase ([.owy 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'Harc eta!., Proc. Natl. Acad. Sci. USA 78:1527 (1981)); gpt, which confers resistance to mycophenoiic 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. Pharmacol. Toxicol. 32:573-596 (1993), Mulligan, Science 260:925-932 (1993); and Morgan and Anderson, Ann. Rev. Biochcm. 62:191-217 (1993); May, 1993, TIB TECH U(5):1S5-2I5 (!993)); and hygro, which confers returnee to hygromyrin (Sanlerre et a!, Dene 10-14"? (1984)) Maoris 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 jr. Chapters 12 and H Dracopoli et al. (eds), Current Protocols in Human Genetic';. John Wiley & Sons, NY (1994); Oolhen-r-fiarapin et al., J. Mol. Diol. 150:1 (1981), which arc incorporated by reference herein in their entireties.
|OI92| Hie 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 (Grouse et al., Mol. Cell. Biol. 3:257 (1983)).
[0193] Vectors which use giutamme 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 murinc myeloma cell line, NSO) which are glutamine synthase negative. Glutamine synthase expression systems can also function in glutamine synthase expressing cells (e.g. Chinese Hamster Ovary (CHO) cells) by providing additional inhibitor to prevent the functioning of the endogenous gene. A glutamine synthase expression system and components thereof are detailed in PCT publications: WO87/04462; WO86/05807; W089/01036; WO89/10404; and W091/06657 which are incorporated in their entireties by reference herein. Additionally, glutamine synthase expression vectors that may be used according to the present invention are commercially available from suppliers, including, for example Lonza Biologies, Inc. (Portsmouth, NH). Expression and production of monoclonal antibodies using a GS expression system in murine myeloma cells is described in Bebbington et al., Bio/technology 10:169(1992) and in Biblia and Robinson Biotechnol. Prog. 11:1 (1995) which are incorporated in their entireties by reference herein.
|0194| 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); Kohlcr, Proc. Natl. Acad. Sci. USA 77:2197 (1980)). The coding sequences for the heavy and light chains may comprise cDNA or genomic DNA.
|0195] 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), ccntrifugation, 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 hcterologous polypcptide sequences described herein or otherwise known in the art, to facilitate purification.
Modifications of Antibodies
|0196) 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 ammo acid sequence is a hexa-histidine peptide, such as the tag provided in a pQE vector (Q1AGEN, 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.
|0197| The present invention further encompasses antibodies or fragments thereof conjugated to a diagnostic or therapeutic agent. The anlibodies 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 pcroxidase, alkaline phosphatase, beta-galactosidase, or acctylcholinesterase; examples of suitable prosthetic group complexes include -sireplavidin/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, 11 lln or Wl'c. Other examples of detectable substances have been described elsewhere herein.
[0198] Further, an antiooay 01 me 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 melal 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, I-dehydrotestosterone, glucocorticoids, procaine, tetracaine, lidocaine, propranolol, and puromycin and analogs or homologs thereof. Therapeutic agents include, but are not limited to, antimetabolites (e.g., rnethotrexate, 6-mercaptopurine, 6-thioguanine, cytarabine, 5-fluorouracil decarbazine), alkylating agents (e.g., mcchlorcthamine, 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 (AMQ), and anti-mitotic agents (e.g., vincristine and vinblastine).
(0199) 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, fi-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., Int. Immuno!., 6:1567-1574 (1994)), VEGI (See, International Publication No. WO 99/23105), a thrornbotic agent or an anti- angiogenic agent, e.g., angiostatin or endostatin; or, biological response modifiers such as, for example, lymphokines, interlculcin-1 ("1L-1"), interleukin-2 ("1L-2"), interleukin-6 ("1L-6"), granulocyte macrophage colony stimulating factor ("GM-CSF"), granulocyte colony stimulating factor ("G-CSF"), or other growth factors.
[0200| 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. [02011 Techniques for conjugating such therapeutic moiety to antibodies are well known. See, for example, Amon 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, Pinchcra 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. (cds.), pp. 303-16 (Academic Press 1985), and Thorpe et al., "The Preparation And Cytotoxic Properties Of Antibody-Toxin Conjugates", Immuno!. Rev. 62:119-58 (1982).
|0202] 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, which is incorporated herein by reference in its entirety.
|0203| An antibody, with or without a therapeutic moiety conjugated to it, administered alone or in combination with cytotoxic factor(s) and/or cytokinc(s) can be used as a therapeutic.
Antibody-albumin fusion
[0204| 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.
|0205] 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 VII CDRI. 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. |0206] 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 CDRI. In other embodiments, the fragment or variant of an antibody that inmunospecifcally binds a Therapeutic protein and that corresponds to a Therapeutic protein portion of an albumin fusion protein comprises, or jlternatively consists of, the VL CDR2. In other embodiments, the fragment or variant of an antibody that immunnspedfrally 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. |0207] I" 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.
|0208] 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 !he Therapeutic antibody, linked to the VL domain of the therapeutic antibody by a peptide linker such as (Gly,Ser)3 (SEQ ID N0:4).
fmtnunophenotyping
[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 (or fragment or variant thereof) may be utilized for immunophenoryping of cell lines and biological samples. Therapeutic proteins of the present invenlion 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, 96:73149 (1999)),
(0210] These techniques allow for the screening of particular populations of cells, such as might be found with hematologica) malignancies (i.e. minimal residual disease (MRD) in acute Icukemic patients) and "non-self cells in transplantations to prevent Graft-versus-Kost Disease (GV'HD). Alternatively, these techniques allow for the screening of hematopoietic stem and progenitor cells capable of undergoing proliferation and/or differentiation, as mighl 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
|0211| 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 maybe 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. |0212| 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., Haughten, Bio/Techniques 13:412^121(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 phagc (e.g.. Scott and Smith, Science 249:386-390 (1990); Devlin, Science 249:404-406 (1990); Cwirla ct al., Proc. Nail. Acad. Sci. USA 87:6378-6382 (1990); and Felici, J. Mol. Biol. 222:301-310 (1991)) (each of these references is incorporated herein in its entirety by reference). The antibodies of the invention or albumin fusion proteins of the invention comprising at least a fragment or variant of an antibody (hat 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.
|0213| The albumin fusion proteins of (he invention comprising at least a fragmenl or variant of an antibody that binds a Therapeutic protein may be assayed for cross-reactivily with other antigens (e.g., molecules that have sequence/structure conservation with the moli;cule(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.
[0214| Immunoassays which can be used to analyze (immunospecificj binding and cross-reactivity include, bui are not limited to, competitive and non-competitive assay systems using techniques such as western blots, radioimmunoassays, EL1SA (enzyme linked immunosorbent assay), "sandwich" immunoassays, immunoprecipitation assays, precipitin reactions, gel diffusion precipitin reactions, immunodiffusion assays, agglutinalion assays, complement-fixation assays, immunoradiornetric assays, fluorescent immunoassays, and protein A immunoassays, lo name but a few. Such assays are routine and well known in the art (see, e.g., Ausubcl et al, eds, 1994, Current Protocols in Molecular Biology, Vol. I, John Wiley & Sons, Inc., New York, which is incorporated by icfercnce herein in its entirety). Exemplary immunoassays are described briefly below
(but are not intended by way oflimitation).
|0215] Immunoprecipitation protocols generally comprise lysing a population of cells in a lysis buffer such as RIPA buffer (1% NP-40 or Triton X-IOO, 1% sodium deoxycholate, 0.1% SDS, 0.15 M NaCI, 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.
[0216| Western blot analysis generally comprises preparing protein samples, elcctrophoresis 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, eg., an anti-human serum albumin antibody) conjugated to an enzymatic substrate (e.g., horseradish peroxidase or alkaline phosphatase) or radioactive molecule (e.g., "P or l!!l) 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.
|0217| ELlSAs 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. I.John Wiley & Sons, Inc., New York at 11.2.1. |02I8| The binding affinity of an albumin fusion protein to a protein, antigen, or epitope and the off-rate of an antibody- or albumin fusion protcin-protcin/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., 3I1 or I2il) 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 '"I) in the presence of increasing amounts of an unlabeled second protein that binds the same protein, antigen, or epitope as the albumin fusion protein of the invention
|0219| 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 polypeptidcs, antigens or epitopes from chips with immobilized specific polypeptides, antigens or epitopes, antibodies or albumin fusion proteins, respectively, on their surface.
Therapeutic Uses
|0220| 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
ot trie invention inciuae, out arc 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 aclivity of a Therapeutic protein includes, bul 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 pharmaceulically acceptable compositions as known in the art or as described herein. |0221| In a specific and preferred embodiment, Ihe 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 thai binds a Therapeutic protein may be provided in pharmaeeutically acceptable compositions as known in the art or as described herein.
|0222| 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.
|0223| 'ITie 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., 1L-2, IL-3 and IL-7), for example, which serve to increase the number or activity of effector cells which interact with the antibodies.
(0224) The antibodies of the invention or albumin fusion proteins of the invention comprising at least a fragment or variant of an antibody that hinds a Therapeutic protein rnay be administered alone or in combination with other types of treatments (e.g., radiation therapy, chemotherapy, hormonal therapy, immunothcrapy 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.
|0225| 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 coitelale of such an antibody) for both immunoassays directed to and therapy of disorders related to polynucleotidcs or polypeptides, including fragments thereof, of the present invention. Such antibodies, fragments, or regions, will preferably have an affinity for polynuclcotides or polypeptides of the invention, including fragments thereof. Preferred binding affinities include dissociation constants or Kd's less than 5 X 10 J M, 10'2 M, 5 X 10' M, 10° M, 5 X 104 M, XT' M. More preferred binding affinities include those with a dissociation constant or Kd less than 5 X 10'5 M, 105 M, 5 X 10s M, 10"6M, 5 X 10'1 M, 10' M, 5 X 10' M or 10'" M. Even more preferred binding affinities include those with a dissociation constant or Kd less than 5 X ICr5 M, 10' M, 5 X 10"'° M, 10"'° M, 5 X 10'" M, 10"" M, 5 X 10' 11 M, lO'12 M, 5 X 10 " M, 10'13 M, 5 X 10'14 M, 10 ' Gene Therapy
(0226] 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.
|0227] 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
(0228) 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
|0229| 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.
[0230] 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. |023I| 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, [n 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.
|0232] 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. [0233| 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 ct al., in Liposomes in the Therapy of Infectious Disease and Cancer, Lopcz-Bcrestein and Fidler (eds.), Liss, New York, pp. 353- 365 (1989); Lopez-Berestein, ibid., pp. 317-327; see generally ibid.)
|0234| In yet another embodiment, the compound or composition can be delivered in a controlled release system. In one embodiment, a pump may be used (sec Langer, supra; Sefton, CRC Crit. Ref. Biomed. Eng. 14:201 (1987); Buchwald et at, 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 (cds.), CRC Pres., Boca Raton, Florida (1974); Controlled Drug Bioavailability, Drug Product Design and Performance, Smolen and Ball (eds.), Wiley, New York (1984); Ranger and Peppas, J., Macromol. Sci. Rev. Macromol. Chem. 23:61 (1983); see also Levy et al., Science 228:190 (1985); During et al., Ann. Neurol. 25:351 (1989), Howard et al., J.Neurosurg. 71:105 (1989)). 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)). [0235] Other controlled release systems are discussed in the review by Langer (Science 249:1527-1533 (1990)).
|0236] 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 intraccllular, 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., .loliot et al., Proc. Natl. Acad. Sci. USA 88:1864-1868 (1991)),
etc. Alternatively, a nucleic acid can he introduced intracellularly and incorporated within host cell UNA for expression, by homologous recombination.
[0237] The present invention alsn 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, propylcne, 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.
|0238| 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.
|0239] 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. |0240] 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. |0241] 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 I 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
|0242| 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.
|0243] 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 prevcntative measures or aggressive treatment earlier thereby preventing the development or further progression of the cancer.
|0244] 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., seeJalkanen et al., J. Cell. Biol. 101:976-985 (1985); Jalkanen et al., J. Cell . Biol. 105:3087-3096(1987)). Other antibody-based methods useful for detecting protein gene expression include immunoassays, such as the enzyme linked immunosorbent assay (ELISA) and the radioimmunoassay (RIA). Suitable antibody assay labels 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.
|0245] 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.
|0246) 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 millicurics 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. Burchicl et al., "Imrhunopharmacokinctics of Radiolabcled 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)).
|0247j 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.
|0248] 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.
|0249| 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 lhat may be used in the diagnostic methods of the invention include, but arc not limited to, computed tomography (CT), whole body scan such as position emission tomography (PET), magnetic resonance imaging (MRI), and sonography.
|0250| In a specific embodiment, the molecule is labeled with a radioisotope and is detected in the patient using a radiation responsive surgical instrument (Thurslon 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 prolein 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
|0251| The present invention piovides 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).
[0252] 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 polypeptidc 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.
(0253) 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.
|0254| 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.
[0255] 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).
|0256| 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).
|0257| Thus, the indention 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
(0258) 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.
|0259| 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. [0260] Preferred albumin fusion proteins of the invention, include, but arc 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 polynuclcotide 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: (I) 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 rcgulatable or inducible promoter, a constitutive promoter), (3) a region
for termination of transcription, (4) a leader sequence, and (5) a selectable marker.
(02611 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 scrum albumin protein component of the albumin fusion protein is the mature portion of serum albumin.
|0262] 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.
[0263| 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 serurn albumin.
|0264| Preferably, the albumin fusion protein comprises HA as the N-terminal portion, and a Therapeutic protein as the C-tcrminal 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.
|0265] In other embodiments, the albumin fusion protein has a Therapeutic protein fused to both the N-terminus and the C-tenminus 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 proleins 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.
|II2661 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- lenninus 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 [hereof 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 anolhcr 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).
|0267| In one embodiment, one, two, three, or more taridemly 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. |0268| Albumin fusion proteins of the invention further encompass proteins containing one, two, three, four, or more molecules of a given Hierapculic 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-icrminus of albumin or variant thereof, wherein the molecules are joined through peptide linkers. Examples include those peptide linkers described in U.S. Pat. No. 5,073,627 (hereby incorporated by reference). Albumin fusion proteins comprising multiple Therapeutic protein X polypeptides separated by peptide linkers may he produced using conventional recombinant DNA technology. Linkers are particularly important when fusing a small peptide to the large HSA molecule. The peplide 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.
[0269] Further, albumin fusion proteins of the invention may also be produced by fusing a Therapeutic protein X or variants thereof to the N-termmal 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 monotneric 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.
[0270| In addition to albumin fusion protein in which the albumin portion is fused N- terminal and/or C-tcrminal 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 arc
stabilized by disulphide bonds. The loops, as determined from the crystal slmcture of HA (PDB identifiers 1A06, !BJ5, 1BK.E, 1BMO, 1E7E to
1E71 and 1UOR) 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, lo essentially generate an
albumin molecule with specific biological activity.
|02711 Loops in human albumin structure into which peptides or polypeptides may be inserted to generate albumin fusion proteins of the
invention include: Val54-Asn61, Thr76-Asp89, Ala92-GlulOO, Glnl70-Alal76, His 247 - Glu252, Glu 266 - Glu277, Glu 280-His288, Ala362-
Glu368, I.ys439-Pro447, Val462-Lys475, Thr478-Pro486, and Lys560-Thr566. In more preferred embodiments, peptides or polypeptides are
inserted into the Val54-Asn61, Glnl70-Alal76, and/or Lys560-Thr566 loops of mature human albumin (SEQ ID NO:1).
[0272] 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.
|0273) Such library(s) could be generated on HA or domain fragments of HA by one of the following methods:
randomized mutation of amino acids within one or more peptide loops of HA or HA domain fragments. Either one, more or all the residues within a loop could be mutated in this manner;
replacement of, or insertion into one or more loops of HA or HA domain fragments (i.e., internal fusion) of a randomized peptide(s) of length Xn (where X is an amino acid and n is the number of residues;
N-, C- or N- and C- terminal peptide/protein fusions in addition to (a) and/or (b).
|0274] The HA or HA domain fragment may also be made multifunctional by grading the peptides derived from different screens of different loops against different targets into the same HA or HA domain fragment.
[0275] 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 pcplide 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 ammo acids fused to the N-tcrminus 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 scrum albumin. For example, short peptides described in Table 1 and 2 (e.g.. Therapeutic Y) can be inserted into the albumin loops.
[0276| 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-tcrminal 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-IIA-Y or Y-HA-X.
[0277] For example, an anti-BI.yS™ scFv-HA-IFNa-2b fusion may be prepared lo modulate the immune response to IFNa-2b by anti-BLyS™ scFv. An alternative is making a bi (or even multi) functional dose of HA-fusions eg. HA-lFNa-2b fusion mixed with HA-anti-BLyS™ scFv fusion or other HA-fusions in various ratio's depending on function, half-life etc.
|0278| 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.
[0279] As an alternative to the fusion of known therapeutic molecules, the peptides could be obtained by screening libraries constructed as fusions lo the N-, C- or N- and C- termini of HA, or domain fragment of HA, of typically 6, 8, 12, 20 or 25 or Xr (where X is an amino acid (aa) and n equals the number of residues) randomized amino acids, arid 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 peplide derived hy any other method then being attached to HA.
[0280] 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 ofamino acids such that it is flexible or more rigid
[281] The linker sequence may he 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 S1. ccrevisiae protease kex2 or equivalent proteases.
[02X2] Thcicfore, as described above, the albumin fusion proteins of the invention may have the following formula R1-L-R2; R7.-L-R1; or Rl-
L-R2-L-R1, wherein Rl is at least one Therapeutic protein, peptide or polypeptide sequence, and not necessarily the same Therapeutic protein, Lisa
linker and R2 is a serum albumin sequence.
|0283] In preferred embodiments, albumin fusion proteins of the invention comprising a Therapeutic protein have a higher plasma stability
compared to the plasma stability of the same Therapeutic protein when not fused to albumin. Plasma stability typically refers to the time period
between when the Therapeutic protein is administered in vivo and carried into the bloodstream and when the therapeutic protein is degraded and
cleared from the bloodstream, into an organ, such as the kidney or liver, that ultimately clears the Therapeutic protein from the body. Plasma
stability is calculated in terms of the half-life of the Therapeutic protein in the bloodstream. The half-life of the Therapeutic protein in the
bloodstream can be readily determined by common assays known in the art.
[0284] 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.
|0285| 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.
|0286] 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
|0287] 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.
|0288] 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. |0289| The Saccharomyces cerevisiae invertase signal is a preferred example of a yeast-derived signal sequence.
|0290| Conjugates of the kind prepared by Poznarisky cl ai, (Fl-BS Lett. 239:18 (1988)), in which separately-prepared polypeptides are joined by chemical cross-linking, are not contemplated.
|02'>1] 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 arc known and may be used, including bacteria (for example E. coli and llacillus sublilis), yeasts (for example Saccharomyces cerevisiae, Kluyveramyces laclis and Pichia pastoris, filamentous fungi (for example Aspcrgillus), plant cells, animal cells and insect cells.
|0292] Preferred yeast strains to be used in the production of albumin fusion proteins are D88, DXY1 and BXPIO. D88 [Ieu2-3,leu2-122,canl, pral, tibc4] is a derivative of parent strain AH22A/S* (also known as DB1; see, e.g., Sleep el al. Biotechnology 8:42-46 (1990)). The strain contains a Iru2 mutation which allows for auxotropic selection of 2 rnicron-based plasmids that contain the LEU2 gene. D88 also exhibits a derepression of PRB1 in glucose excess. The PRO 1 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, hereby incorporated in its
entirety by reference herein).
[0293| DXY1, a derivative of U88, has the following genotype: [!eu2-3, Ieu2-122, canl.pral,ubc4. uri>3::yap3]. In addition to the mutations isolated in D88, this strain also has a knockou: of the YAP3 protease. This protease causes cleavage of mostly di-basic residues (RR RK KR, KK) but can also promote cleavage at single basic residues in proteins. Isolation of this yap3 mutation resulted in higher levels of full length HSA production (see, e.g., U.S. Patent No. 5,965,386 and Kerry-Williams et al., Yeast 14:161-169 (1998), hereby incorporated in their entireties by reference herein).
|0294| BXPIO has the following genotype: leu2-3, Ien2-l22, canl, prnl, ubc4, ura3, ynp3::URA3, Iys2, hsp!50::LYS2, pmll::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 evollltionarily conserved family of dolichyl-phosphate-D-mannose protein 0-mannosyltransferases (Pmts). The transmcmbrane 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 O-linked glycosylation of HSA fusions (see, e.g., International Publication No. WOOO/44772, hereby incorporated in its entirety by reference herein). Studies revealed that the HsplSO 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, hereby incorporated in its entirety by reference herein.
|0295) 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.
|0296| 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.
(0297) Useful yeast plasmid vectors include pRS403-406 and pRS4!3-416 and arc 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, 7RH1, LEU2 and URA3. Plasmids pRS413-416 are Yeast Centromere plasmids (Ycps).
|0298) Preferred vectors for making albumin fusion proteins for expression in yeast include pPPCOOOS, 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 PRHI S. cerevisiae promoter (PRBlp), a Fusion leader sequence (FL), DNA encoding HA (rHA) and an ADHI 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 arnino acids of the mating factor alpha 1 promoter (SLDK.R, see EP-A-387 319 which is hereby incorporated by reference in its entirety).
[0299| The plasmids, pPPCOOOS, 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 el ai, BioTechnology 8:42 (1999) which is hereby incorporated by reference in its entirety.
[0300] A 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 Mel25 promoter is 383 bases long (bases-382 to -1) and the genes expressed by this promoter are also known as Metis, Metl7, and YLR.303W. A preferred embodiment uses the sequence below, where, at the 5' end of the sequence below, Ihe Not 1 site used in the cloning is underlined and at the 3' end, the ATG start cod on is underlined:
|I)3U1| A variety ot memous nave been developed to operably link DNA to vectors via complementary cohesive termini. For instance, complementary homopolymcr 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 homopolynieric tails to form recombinant DNA molecules.
[03031 Synthetic linkers containing one or more restriction sites provide an alternative method of joining the DNA segment to vectors. The UNA segment, generated by endonuelease restriction digestion, is treated with bacteriophage T4 DNA polymerase or E. coli DNA polymerase i, enzymes that remove protruding, gamma-smgle-stranded termini with their 3' 5'-exorjuc!eolytic activities, and fill in recessed 3'-cnds with their polymerizing activities.
1U304| 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 [he presence of an enzyme that is able to catalyze the ligation of blunt-ended DNA molecules, such as hacteriophage 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 erwyrne that produces termini compatible with those of the DNA segment.
[0305] Synthetic linkers containing a variety of restriction endonuciense sites are commercially available from a number of sources including International Biotechnologies Inc, New Haven, CT, USA.
|0306) A desirable way to modify the DNA in accordance with the invention, if, for example, HA variants are to be prepared, is to use (he polymerase chain reaction as disclosed by Saiki el til. (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. |0307] 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 (Hanscnula), Sacckaromyces, Kluyveromyces, Candida, Torulopsis, Torulaspora, Schizosacchnrumyccs, Citeromyces, Puchysolun. Debaromyc.es, Metschunikowia, Rhodasporidium, Leucosporidium, Bolryoascus, Spvridioboliis, Endomycopsis, and the like. Preferred genera are those selected from the group consisting of Saccharomyces, Scfiaosaccharomyces, Klu)>veromyces, Pichia and Tomlnspora. Examples of Sticcftftromyces spp. are S. cercvisiue. S. italicus and S. toitxii.
|030SJ Examples of Kluyveromyces spp. are K. fragilis, K. lactis and K.. marxianus. A suitable Torulaspora species is T. delbrueckii. Examples of Pichia (Hanscnula) 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, EP258 067 and WO 90/01063, all of which are incorporated herein by reference. |0309| 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 8 Y4743 yap3 mutant (ATCC Accession No. 4022731); Saccharomyces cerevisiae Hansen, teleomorph strain BY4743 hsp)50 mulant (ATCC Accession No. 4021266); Saccharomyces cerevisiae Hansen, teleomorph strain BY4743 pmtl mutant (ATCC Accession No. 4B23792); Saccharomyces cerevisiae Hansen, telcomorph (ATCC Accession Nos. 20626; 44773; 44774; and 62995); Saccharomyces diastaticus Andrews et Gilliland ex van der Walt, teleomorph (ATCC Accession No. 62987); KJuyveromyces lactis (Dombrowski) van der Walt, teleomorph (ATCC Accession No. 76492); Pichia angusta (Teunisson et al.) KurUman, teleomorph deposited as Hansenula polymorpha dc 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 nijulam (Kidiim) Winter, anamorph (ATCC Accession No. 48756); and Yarrowia lipolytica (Wickerham et al.) van der Walt et von Arx, teleomorph (ATCC Accession No. 201847).
[0310J Suitable promoters for S. cerevisiae include Ihose associated with the PGK1 gene, GAL1 or GAUO genes, CYC1, PH05, TRIM, ADIII, ADH2. (lie genes for glyceraldehyde-3-phosphatc dehydrogenase, hexokmase, pyruvatc dccarboxylase, phosphofructokinasc. triosc phosphate isomcrasc, phosphoglucose isomcr.ise, glueokinase, alpha-mating factor phcromone, [a mating factor pheromone], the PRBI promoter, the GUT2 promoter, the GPD1 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 (eg. the promoter of EP-A-25S 067).
|03II| Convenient regulatablc promoters for use in Xchiiosaccharotnyces pambe are the thiarnine-reprcssible promoter from the nmt gene as described by Maundrell (1990J J. Biol. Chem. 265, 10857-10864 and the glucose repressible jbpl gene promoter as described by Hoffman & Winston (1990) Genetics 124. 807-816.
10312| Methods of transforming Pichia for expression of foreign genes are taught in, for example, Cregg ct al. (1993), and various Phillips palcnts (e.g. US 4 857 467, incorporated herein by reference), and Pichia expression kits are commercially available from Invitrogen BV, Leek, Netherlands, and Invitrogen Corp., San Diego, California. Suitable promoters include AOXI and AOX2. Gleeson et al. (1986) J. Gen. Microbiol.
132, 3459-3465 include mlormation on ilanscnula vectors and transformation, suitable promoters being MOX1 and FMD1; whilst EP 361 991,
Fleer et at. (1991) and other- publications from Rhone-Poulcnc Rorer teach how to express foreign proteins in Kluyveromyces spp., a suitable
promoter being PGKT
[03131 'Flic transcription termination signal is preferably the 3' flanking sequence of a eukaryotic gene which contains proper signals lor
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. cercvisiae ADHI gene is preferred.
[0314] 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:
theMPIF-l signal sequence (e.g., amino acids 1-21 of GenBank Accession number AAB51134) MKVSVAALSCLMLVTALGSQA
(SliQ ID N0:6)
the stanniocalcin signal sequence (MLQNSAVLLLLV1SASA, SEQ ID NO:7)
the pre-pro region of the USA signal sequence (e.g., MKWVTFISLLFLFSSAYSRGVFRR, SEQ ID NO:8)
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., MLLQAFLFLLAGFAAK1SA, SEQ ID NO:11)
I) the yeast mating factor alpha signal sequence (e.g.,
MRFPSlFTAVLAFAASSALAAPVNnTKDETAQ]PAEAVIGYSDLEGDFDVAVLPFSNSTNNGLLFINTTIASIAAK.EEGVSLEKR, SEQ ID NO: 12 or
MRFPSIFTAVLAFAASSALAAPVNTTTEDETAQIPAEAVIGYSDLEGDFDVAVLPFSNSTNNGLLFINTTIASIAAKF.EGVSLDKR, 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/MFa-l hybrid signal sequence (also known as HSA/kex2) (e.g., MK.WVSF1SLLFLFSSAYSRSLDKR, SEQ ID NO: 14) j) a K. lactis killer/ MFa-1 fusion leader sequence (e.g., MNIFYIFLFLLSFVQGSLDKR, SEQ ID NO: 15) k) the Immunoglobulin Ig signal sequence (e.g., MGWSCI1LFLVATATGVHS, 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, MK.WVSF1SLLFLFSSAYSRGVFRR (SEQ IDNO:20), MKWVTFISLLFLFAGVLG (SEQ ID NO:21), MK.WVTFISLLFLFSGVLG (SEQ ID NO:22), MKWVTF1SLLFLFGGVLG (SEQ ID NO:23),
Modified HSA leader USA #64- MKWVTFISLLFLFAGVSG (SEQ ID NO:24); Modified HSA leader HSA #66 - MKWVTFISLLFLKGGVSG (SEQ ID N0:25); Modified HSA (AI4) leader- MKWVTFISLLFLFAGVSG (SEQ ID NO:26);
Modified HSA (S14) leader (also known as modified HSA #65) - MKWVTF1SLLFLFSGVSG (SEQ ID NO:27), Modified HSA (G14) leader - MK.WVTF1SLLFLFGGVSG (SEQ ID NO:28), or MK.WVTFISLLFLFGGVLGDLHKS (SEQ ID N0:29) p) a consensus signal sequence (MPTWAWWLFLVLLLALWAPARG. SEQ ID NO:30) q) acid phosphatasc (PH05) leader (e.g., MFKSVVYSILAASLANA SEQ ID N0:31) r) the pre-scqucnce of MFoz-1 s) the pre-.sequence of 0 glucanase (BGL2) t) killer toxin leader u) the presequcnce of killer toxin v) k. lactis killer toxin prcpro (29 amino acids; 16 amino acids of pre and 13 amino acids of pro)
MNIFYIFLFLLSFVQGLI-HTHRRGSLDKR (SEQ ID NO:32) w) S. diaslnticus glucoamylase II secretion leader sequence x) S. carlsbergensis a-galactosidase (MELI) secretion leader sequence y) Candida glucoarnylasc leader sequence
2) The hybrid leaders disclosed in EP-A-387 319 (herin incorporated by reference)
aa) the gp67 signal sequence (in conjunction with baculoviral expression systems) (eg-, amino acids 1-19 of GenBank Accession Number
AAA72759) or
bb) the natural leader i'~the therapeutic protein X;
cc) S. ccrevisiae invertase (SUC2) leader, as disclosed in JP 62-096086 (granted as 911036516, herein incorporate by reference); or dd) Inulinase-MKLAYSLLLPLAGVSASVINYKR (SEQ ID N0:33). ee) A modified TA57 propeptide leader variant #1 -
MKLK'IA'RSAVLSSLI-'ASQVLGQPIDDTESQITSVNLMADDTESAFATQTNSGGLDVVGLISMAKR (SEQ ID N0:34) IT) A modified TA57 propeptide leader variant #2 -
MKLKTVRSAVI^SLFASQVLGQPIDDTESQTfSVNLMADDTESAFATQTNSGGLDVVGLlSMAEEGEPKR (SEQ ID NO:35) gg) A consensus signal peptide - MWWRLWWLLLLLLLLWPMVWA (SEQ ID N0:l 11) hh) A modified HSA/kex2 signal sequence- MKWVSFISLLFLFSSAYSGSLDKR (SEQ ID NO:112) n) A consensus signal peptide #2 - MRPTWAWWLFLVLLLALWAPARG (SEQ ID NO: 105) Additional Methods of Recombinant and Synthetic Production of Albumin Fusion Proteins
103151 'Hie present invention also relates to vectors containing a polynucleotidc 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.
|0316| 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. [0317) The polynucleotide insert should be opcratively linked to an appropriate promoter, such as the phage lambda PL promoter, the E. coli lac, Irp, 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 polypcptide to be translated.
[0318| As indicated, the expression vectors will preferably include at least one selectable marker. Such markers include dihydrofolate reductase, 0418, glutamine synthase, or neomycin resistance for eukaryotic cell culture, and tetracycline, kanamycin or ampicillin resistance genes for culturing in E. coli and other bacteria. Representative examples of appropriate hosts include, but are not limited to, bacterial cells, such as K. coli, Streptomyces and Salmonella typhimurium cells; fungal cells, such as yeast cells (e.g., Saccharomyces cerevisiue 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. [0319] Among vectors preferred for use in bacteria include pQE70, pQE60 and pQE-9, available from QIAGEN, Inc.; pDluescript vectors, Phagcscript vectors, pNHSA, pNH16a, pNHISA, pNH46A, available from Stratagene Cloning Systems, Inc.; and ptrc99a, pKX223-3, pK.(C233-3, pDR540, pRITS available from Pharmacia Biotech, Inc. Among preferred eukaryotic vectors are pWLNEO, pSV2CAT, pOG44, pXTl and pSG available from Stratagene; and pSVK.3, pBPV, pMSG and pSVL available from Pharmacia. Preferred expression vectors for use in yeast systems include, but are not limited to pYES2, pYDl, pTEFl/Zeo, pYES2/GS, pPICZ, pGAPZ, pGAPZalph, PP1C9, pP[C3.5, pHIL-D2, pHIL-SI, pPIC3.5K, pPIC9K, and PAO815 (all available from Invitrogen, Carlbad, CA). Other suitable vectors will be readily apparent to the skilled artisan. |0320| 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. co/i, 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 pelS 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 IhepelB pectate lyase signal sequence to increase the efficiency of expression and purification of such polypeptides in Gram-negative bacteria. Sec, U.S. Patent Nos. 5,576,195 and 5,846,818, the contents of which are herein incorporated by reference in their entireties.
(03211 Examples of signal peptides that may be fused to an albumin fusion protein of the invention in order to direct iis secretion in mammalian cells include, but are not limited to:
a) the MFlh-l signal sequence (e.g., amino acids 1-21 of GenBank Accession number AAB51134) MK.VSVAALSCLMLVTALGSQA
(SEQ ID NO:6) h) the stanniocalrin signal sequence (MI.QNSA VI.I LLVISASA. SFO ID N0:7)
the pre-pro region of the HSA signal sequence (e.g., MKWVTFISLLFLFSSAYSRGVFRR, SEQ ID NO:8)
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)
the invertase signal sequence (e.g., MLLQAFLFLLAGFAAKJSA, SEQ ID N0:l 1)
the yeast mating factor alpha signal sequence (e.g.,
MRFPSIFTAVLAFAASSALAAPVNTTTEDETAQIPAEAVIGYSDLEGDFDVAVLPFSNSTNNGLLFINTTIASIAAKEEGVSLEICR, SEQ ID NO:12 or
MRFPSlFTAVLAFAASSALAAPVNlTrEDETAQlPAEAVIGYSDLEGDFDVAVLPFSNSTNNGLLFINrflASIAAlCF.EGVSLDKR, SEQ ID NO: 12)
g) K. lactis killer toxin leader sequence
h) a hybrid signal sequence (e.g., MKWVSF1SLLFLFSSAYSRSLEKR, SEQ ID NO: 13)
i) an HSA/MFa-I hybrid signal sequence (also known as HSA/kex2) (e.g., MK.WVSFISLLFLFSSAYSRSLDKR, SEQ ID NO:14) j) a/C. lactis killer/ MFa-1 fusion leader sequence (e.g., MNIFYIFLFLLSFVQGSLDKR, SEQ ID NO:I5) k) the Immunoglobulin Ig signal sequence (e.g., MGWSCI1LFLVATATGVHS, 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, MKWVSF1SLLFLFSSAYSRGVFRR (SEQ ID N0:20), MKWVTFISLLFLFAGVLG (SEQ ID N0:21), MKWVTFISLLFLFSGVLG (SEQ ID NO:22), MKWVTFISLLFLFGGVLG (SEQ ID NO:23),
Modified HSA leader HSA #64 - MKWVTF1SLLFLFAGVSG (SEQ ID NO:24); Modified HSA leader HSA #66 - MKWVTFISLIJLFGGVSG (SEQ ID N0:25); Modified HSA (A14) leader- MKWVTFISLLFl.FAGVSG (SEQ ID NO:26);
Modified HSA (S14) leader (also known as modified HSA #65) - MKWVTFISLLFLFSGVSG (SEQ ID N0:27), Modified HSA (014) leader - MKWVTF1SLLFLFGGVSG (SEQ ID NO:28), or MKWVTFISLLFLFGGVLGDLHKS (SEQ ID N0:29) p) a consensus signal sequence (MPTWAWWLFLVLLLALWAPARG, SEQ ID N0:30) q) acid phosphatase (PH05) leader (e.gV, MFKSVVYS1LAASLANA SEQ ID NO:31) r) the pre-sequence of MFoz-1 s) the pre-scquencc of 0 glucanase (BGL2) t) killer toxin leader u) the prcscqucncc of killer tox in v) k. lactis killer toxin prepro (29 amino acids; 16 amino acids of pre and 13 amino acids of pro)
MNIFYIFLFLLSFVQGLEHTHRRGSLDKR (SEQ IDNO:32) w) S. diastaticus glucoarnylase II secretion leader sequence x) S. carlsbergensis a-galactosidase (MHL1) secretion leader sequence y) Candida glucoarnylase leader sequence
7.) The hybrid leaders disclosed in EP-A-387 319 (herin incorporated by reference) aa) the gp67 signal sequence (in conjunction with baculoviral expression systems) (e.g., amino acids 1-19 of GenBank Accession Number
AAA72759) or
bb) the natural leader of the therapeutic protein X;
cc) S. ccrevisiae-invertase (SUC2) leader, as disclosed in JP 62-096086 (granted as 911036516, herein incorporate by reference); or dd) Inulinase - MKLAYSLLLPLAGVSASVfNYKR (SEQ ID N0:33). ce) A modified TA57 propcptide leader variant #1 -
MKLKTVRSAVLSSLFASQVLGQPlDDTESQ'rrSVNLMADDTESAFATQTNSGGLDVVGLISMAKK (SEQ ID NO:34) ff) A modified TA57 propeptide leader variant #2 -
MKLKTVRSAVLSSLFASQVLGQP1DDTESQTTSVNLMADDTESAFATQTNSGGLDVVGLISMAEEGEPKR (SEQ ID N0:35)
gg) A consensus signal peptioc - MWWRLWWLLLLLLLLWPMVWA (SEQ ID NO: 111) jj) A modified HSA/kex2 signal sequence- MK.WVSFISLLFLFSSAYSGSLDICR (SEQ ID N0:l 12) kk) A consensus signal pcptidc #2 - MRPTWAWWLFLVLLLALWAPARG (SEQ ID NO: 105)
103221 In a preferred embodiment, the modified HSA/kex2 signal sequence (SEQ ID NO:112) is fused to the amino terminus of an albumin fusion protein, including fusion proteins comprising albumin and a therapeutic protein as described herein, as well as albumin fusion proteins disclosed in W093/15199; WO97/24445; WO03/60071; WO03/59934; and PCT/US04/01369, each of which are incorporated herein by reference in their entireties. The modified HSA/kex2 signal sequence is based on the HSA/kex2 signal sequence (SEQ ID NO: 14) disclosed, e.g., in Sleep et a)., Biotechnology 1990, vol. 8, pp. 42-46; and US Patent 5,302,697, both of which are incorporated herein by reference in their entireties. The modified HSA/kex2 leader sequence disclosed herein contains a non-conservative amino acid substitution (Arg to Gly) at residue 19 of the parent signal pcptide. The modified HSA/kex2 signal pcptide has been found to produce unexpectedly better expression yield and/or better cleavage efficiency of albumin fusion proteins when expressed in yeast than the unmodified HSA/kex2 signal sequence. Variants of the modified HSAAex2 signal peptide arc also encompassed by the invention. In particular the Gly residue at position 19 of SEQ ID NO:112 may be substituted with a Pro residue. Other conservative substitution variants of the modified HSA/kex2 signal sequence are also contemplated. Nucleic acids encoding the modified HSA/kcx2 signal sequence of SEQ ID NO:I12, as well as conservative substitution variants thereof, are also encompassed by the invention.
(0323) Vectors which use glutamine synthase (GS) or DHFR as the selectable markers can be amplified in the presence of the drugs methionine sulphoximine or methotrexate, respectively. An advantage of glutamine synthase based vectors are the availability of cell lines (e.g., the murine myeloma cell line, NSO) which are glutamine synthase negative. Glutamine synthase expression systems can also function in glutamine synthase expressing cells (e.g., Chinese Hamster Ovary (CHO) cells) by providing additional inhibitor to prevent the functioning of the endogenous gene. A glutamine synthase expression system and components thereof are detailed in PCT publications: W087/04462; W086/05807; WO89/Q1036; WO89/10404; and W091/06657, which are hereby incorporated in their entireties by reference herein. Additionally, glutamine synthase expression vectors can be obtained from Lonza Biologies, Inc. (Portsmouth, Nil). Expression and production of monoclonal antibodies using a GS expression system in murine myeloma cells is described in Bebbington el al, Bio/technology 10:169(1992) and in Biblia and Robinson Biolechnol. Prog. \ \'.\ (1995) which are herein incorporated by reference.
[0324] 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.
(0325) 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 polynucleotidc 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 pulynucleotide may activate, alter, and/or amplify endogenous polynucleotides.
[0326| 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) anoVor heterologous control regions (e.g., promoter and/or enhancer) with endogenous polynuclcntide 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 el al., Proc. Nail. Acad. Sci. USA 56:8932-8935 (1989); and Zijlstra cl al.. Nature 342:435-438 (1989), the disclosures of each of which are incorporated by reference in their entireties).
[0327] 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, hydroxylapawe cnroniaiogrdpny, nyuropnooic cnaigc hilcku.iiuii
chromatography and lectin chromatography. Most preferably, high performance liquid chromatography ("HPLC") is employed for purification.
10331] 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, Toyopcarl Q, Toyopearl QAE, Toyopearl DEAE,
Resource/Source Q and DEAE, Fraclogcl Q and DEAE columns.
[0332] 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, Fractoge] S and CM
columns and their equivalents and comparables.
[0333] 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.
[0334| In specific embodiments the albumin fusion proteins of the invention are purified using Size Exclusion Chromatography including, but
not limited to, sepharose SI 00, S200, S300, supcrdex resin columns and their equivalents and comparables.
|0335] In specific embodiments the albumin fusion proteins of the invention are purified using Affinity Chromatography including, but not
limited to. Mimetic Dye affinity, pcptide affinity and antibody affinity columns that are selective for cither the HSA or the "fusion target" molecules.
|0336| 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.
|0337| Additionally, albumin fusion proteins of the invention may be purified using the process described in PCT International Publication WO
00/44772 which is herein incorporated by reference in its entirety. One of skill in the art could easily modify the process described therein for use in
the purification of albumin fusion proteins of the invention.
|0338] Albumin fusion proteins of the present invention may be recovered from: products of chemical synthetic procedures; and products
produced by rccombinant techniques from a prokaryotic or cukaryotic host, including, for example, bacterial, yeast, higher plant, insect, and
mammalian cells. Depending upon the host employed in a recombinant production procedure, the polypcptides 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 mcthionine
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 prokaryotcs, 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.
|0339| In one embodiment, the yeast Pichia pastoris is used to express albumin fusion proteins of the invention in a eukaryotic system. Pichia
pasloris is a methylotrophic yeast which can metabolize methanol as its sole carbon source. A main step in the methano] metabolization pathway is
the oxidation of methanol to formaldehyde using Oj. This reaction is catalyzed by the enzyme alcohol oxidase. In order to metabolite 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
Oj. Consequently, in a growth medium depending on methanol as a main carbon source, the promoter region of one of the two alcohol oxidase
genes (AOX1) is highly active. In the presence of methanol, alcohol oxidase produced from the AOXI gene comprises up to approximately 30% of
the total soluble protein in Pichia pasloris. See Ellis, S.B., el at, Mol. Cell. Rial. 5:1111-21 (1985); Koutz, P.J, el al.. Yeast 5:167-77 (1989);
Tschopp, J.F., ct a!., Nucl. Acids Kcs. 15:3859-76 (1987). Thus, a heterologous coding sequence, such as, for example, a polynuclcotide of the
present invention, under the transcriptional regulation of all or part of the AOXI regulatory sequence is expressed at exceptionally high levels in
Pichia yeast grown in the presence of methanol.
[0340| In one example, the plasmid vector pP!C9K. is used to express DNA encoding an albumin fusion protein of the invention, as set forth
herein, in a Pichea yeasl 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
AOXI promoter linked to the Pichia pasloris alkaline phosphatase (PHO) secretory signal pcptide (i.e., leader) located upstream of a multiple
cloning site.
|0341] Many other yeast vectors could be used in place of pP!C9K, such as, pYES2, pYDl, pTEFl/Zco, pYES2/GS, pPICZ, pGAPZ,
pGAPZalpha, pPIC9, pPIC3.5, pHIL-D2, pHIL-Sl, PPIC3.5K, and PAO8I5, 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.
|0342| In another embodiment, high-level expression of a heterologous coding sequence, such as, for example, a polynucleotide encoding an
albumin fusion protein ot trie present invention, may be achieved by cloning the heterologous polynuclcotide of the invention into an expression vector such as, for example, pGAPZ or pGAPZalpha, and growing the yeast culture in the absence of methanol.
|0343] In addition, albumin fusion proteins of the invention can be chemically synthesized using techniques known in the art (eg., 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, neoclassical amino acids or chemical ammo acid analogs can be introduced as a substitution or addition into the polypeptide sequence. Non-classical ammo acids include, but are not limited to, to the D-isomcrs 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, omimine, norleucine, norvaline, hydroxyproline, sarcosine, citrulline, homocitrulline, cysteic acid, t-butylglycine, t-butylalanine, phenylglycme, 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).
|0344| The invention encompasses albumin fusion proteins of the present invention which are differentially modified during or after translation, e E , 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, NaBH,; acetylation, formylation, oxidation, reduction; metabolic synthesis in the presence of tunicamycin; etc,
(0345) Additional post-translational modifications encompassed by the invention include, for example, e.g., N-linked or 0-linkcd carbohydrate chains, processing of N-terminal or C-terminal ends), attachment of chemical moieties to the amino acid backbone, chemical modifications of N-linkcd or O-linked carbohydrate chains, and addition or deletion of an N-terminal methionine residue as a result of procaryotic host cell expression. The albumin fusion proteins may also be modified with a detectable label, such as an enzymatic, fluorescent, isotopic or affinity label to allow for detection and isolation of the protein.
|0346| 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 umhelliferone, fluorescein, fluorescein isothiocyanate, rhodamine, dichlorotriazinylamine fluorescein, dansyl chloride or phycoerythrin; an example of a luminescent material includes luminol; examples of biolumincscent materials include luciferase, lucifcrin, and aequorin; and examples of suitable radioactive material include iodine (UII, '"l, '"l, '"]), carbon (MC), sulfur ("S), tritium (3H), indium ("'In, "2ln, "3™ln, llimln), technetium (">Tc,"™Tc), thallium (JO'Ti), gallium ("Ga, 67Ga), palladium (103Pd), molybdenum ("Mo), xenon (n!Xe), fluorine ("F), '"Sm, 177Lu, '"Gd, "'Pm, 140U '"Yb, "6Ho, MY, "Sc, ""Re, mRe, '«Pr, "»Rh, and 97Ru.
[0347| In specific embodiments, albumin fusion proteins of the present invention or fragments or variants thereof are attached to macrocyclic dictators that associate with radiometal ions, including but not limited to, '17Lu, MY, "6Ho, and IS3Sm, to polypeptides. In a preferred embodiment, the radiometal ion associated with Ihe macrocyclic chelators is "'in. In another preferred embodiment, the radiometal ion associated with the macrocyclic chelator is WY. In specific embodiments, the macrocyclic chelator is l,4,7,10-tetraazacyclododecane-N,N',N",N"'-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(IO):2483-90 (1998); Peterson ct al., Bioconjug. Chcm. 10(4):553-7 (1999); and Zimmerman ct al, Nucl. Med. Biol. 26(8):943-50 (1999); which are hereby incorporated by reference in their entirety.
[0348| 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 ubiquitinalion, and they may be cyclic, with or without branching. Cyclic, branched, and branched cyclic polypeptides may result from posrtranslalion natural processes or may be made by synthetic methods. Modifications include acetylation, acylation, ADP-rihosylation, amidation, covalent attachment of flavin, covalent attachment of a heme moiety, covalent attachment of a nucleotide or nucleotidc 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, GP1 anchor formation, hydroxylatian, iodination, methylation, myristylation, oxidation, pegylation, proteolytic processing, phosphorylation, prenylation, racemization, selenoylation, sulfation, transfcr-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)).
|0349| 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 peplide, such as the tag provided in a pQE vector (QIAGEN, Inc., 9259 Eton Avenue, Chatsworth, CA, 91311), among others, many of which arc commercially available. As described in Gentz ct al., Proi.. 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. [0350| 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, vincrisline, vinblastine, colchicin, doxorubicin, daunorubicin, dihydroxy anthracin dione, mitoxantrone, mithramycin, actinomycin D, I -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, streplozotocin, 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).
[0351| 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 1 (See, International Publication No. WO 97/33899), AIM 11 (See, International Publication No. WO 97/34911), Fas Ligand (Takahashi el al.. Int. Immunol.. (5:1567-1574 (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, lymphokincs, intcrleiikin-1 ("IL-1"), interleukin-2 ("IL-2"), interleukin-6 ("1L-6"), granulocyte macrophage colony stimulating factor ("GM-CSF"), granulocyte colony stimulating factor ("G-CSF"), or other growlh factors. Techniques for conjugating such therapeutic moiety to proteins (e.g., albumin fusion proteins) are well known in the art.
|0352| 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.
10353) 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.
|03S4] 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. [0355| 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. [0356] 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 (VH/VL) pair of the original antibody that binds a Therapeutic protein.
[0357| 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, carboxymclhylcellulose, 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.
[0358J The polymer may be of any molecular weight, and may be branched or unbranched. For polyethylene glycol, the preferred molecular weight is between about IkDa and about 100 kDa (the term "about" indicating that in preparations of polyethylene glycol, some molecules will weigh more, v>me less, than 'he stateH 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 l.avt an average molecular weight of about 200, 500, 1000, 1500, 2000, 2500, 3000, 3500, 4000, 4500, 5000. S^OO, 6000, 6500, 7000, 7500 8000, 8500, 9000, 9500, 10,000, 10,500, 11,000, 11,500, 12,000, 12,500, 13,000, 13,500, 14,000, 14,500, 15,000, 15,500, 16,000, 16,500, 17,000, 17,500, 18,000, 18,500, 19,000, 19,500, 20,000, 25,000, 30,000, 35,000, 40,000, 45,000, 50,000, 55,000, 60,000, 65,000, 70,000, 75,000, 80,000, 85,000, 90,000, 95,000, or 100,000 kDa.
|0359| 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 ct al., Appl. Biochem. Biotechnol. 5(5:59-72 (1996); Vorobjcv et al.. Nucleosides Nucleotides /S:2745-2750 (1999); and Caliceti etai, Bioconjug. Chem. 70:638-646 (1999), the disclosures of each of which are incorporated herein by reference. [0360) 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), herein incorporated by reference; 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. Sulftiydryl 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.
|0361] 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.
[0362| 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 (polypcptide) molecules in the reaction mix, the type of pegylation reaction to be performed, and the method of obtaining the selected N-tenminally pegylaled protein. The method of obtaining the N-terminally pegylatcd preparation (i.e., separating this moiety from other monnpegylated 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 alkylation which exploits differential reactivity of different types of primary amino groups (lysine versus the N-terminal) available for derivatization in a particular protein. Under the appropriate reaction conditions, substantially selective derivatization of the protein at the N-terminus with a carbonyl group containing polymer is achieved.
[036.1) 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, the disclosures of each of which arc incorporated herein by reference.
[0364) One system for attaching polyethylene glyco] directly to amino acid residues of proteins without an intervening linker employs trcsylated
MPEG, which is produced by the modification of monmethoxy polyethylene glycol (MPEG) using tresylchloride (CISOjCHjCFj). 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.
[0365) Polyethylene glycol can also be attached to proteins using a number of different intervening linkers. For example, U.S. Patent No.
5,612,460, the entire disclosure of which is incorporated herein by reference, discloses urcthane 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 I,l'-carbonyldiimida7.ole, 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,
the entire disclosure of which is incorporated herein by reference. Pegylated protein products produced using the reaction chemistries set out herein
are included within the scope of the invention.
|03G6| 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-B, 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 Dclgado et a!., Crit. Rev. Thera. Drug Carrier Sys. 9:249-304 (1992).
|0367| The polypeptides of the invention can he recovered and purified from chemical synthesis and recombinant cell cultures by standard methods which include, but arc not limited to, ammonium sulfale or ethanol precipitation, acid extraction, anion or cation exchange chrumatography, 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.
|0368| 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 Polvnucleotides
|0369] 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.
[0370] 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.
(03711 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 trail in the host cell. Additional non-limiting examples of gene therapy methods encompassed by the present invention are more thoroughly described elsewhere herein (sec, e.g., the sections labeled "Gene Therapy", and Examples 61 and 62).
Uses of the Polypeytides
[0372] Each of the polypeptides identified herein can be used in numerous ways. The following description should be considered exemplary and utilizes known techniques.
|0373| Albumin fusion proteins of the invention are useful to provide immunological probes for differential identification of the tissue(s) (e.g., imiminohistochemistry assays such as, for example, ABC immunoperoxidase (Hsu et al., J. Histochem. Cytochem. 29:577-580 (1981)) or cell lype(i) (e.g , immunocytochemistry assays).
[0374| Albumin fusion proteins can be used to assay levels of polypeptides in a biological sample using classical immunohistological methods known to those of skill in the art (e.g., see Jalkanen, et al., J. Cell. Biol. 101:976-985 (1985); Jalkanen, et al., J. Cell. Biol. 105:3087-3096 (1987)). Other methods useful for detecting protein gene expression include immunoassays, such as the enzyme linked immunosorbent assay (ELISA) and the radioimmunoassay (R1A). Suitable assay labels are known in the art and include enzyme labels, such as, glucose oxidase; radioisotopes, such as iodine (I3'l, '"I, ml, 12II), carbon ("C), sulfur ("S), tritium (3H), indium ("imln, "3mln, mln, "'In), and technetium ("Tc, "Tc), thallium (""Ti), gallium ("Ga, "Ga), palladium (1MPd), molybdenum (wMo), xenon (IIJXe), fluorine ("F), '"Sm, "7Lu, '"Gd, '"Pm, "°La, 175Yb, "6Ho, '°Y, "Sc, "'Re, "*Re, H2Pr, '°sRh, 97Ru; luminescent labels, such as luminol; and fluorescent labels, such as fluorescein and rhodamine, and biotin. [0375] 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.
[0376| An albumin fusion protein which has been labeled with an appropriate detectable imaging moiety, such as a radioisotope (for example, '"I, "V "To, ('"I, '"1, '"I, '"}), carbon (14C), sulfur (3!S), tritium (!H), indium ("imln, ":"ln, "!ln, '"in), and technetium ("Tc, Tc), thallium (M'Ti), gallium ("Ga, "Ga), palladium (lo3Pd), molybdenum (MMo), xenon (l33Xe), fluorine (18F, l!3Sm, n?Lu, '"Gd, '"Pm, "°La, "sYb, '"Ho, WY, 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 rase of a radioisotope moiety, for a human subject the quantity of radioactivity injected will normally range from about 5 to 20 millicuries of ""Tc. The labeled albumin fusion protein will then preferentially accumulate at locations in the body (e.g., organs, cells, extracellular spaces or matrices) where one or more receptors, ligands or substrates (corresponding to that of the Therapeutic protein used to make the albumin fusion protein of the invention) are located. 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 polypeptidcs/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 el al., "Immunopharmacokinetics of Radiolabclcd Antibodies and Their Fragments" (Chapter 13 in Tumor Imaging: The Radiochcmical Detection of Cancer, S.W. Burchiel and B. A. Khodcs, 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.
[0377] 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 ribozymcs) 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.
J0378] 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.
(0379| By "toxin" is meant one or more compounds that bind and activate endogenous cytotoxic effector systems, radioisotopcs, 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 kinasc, cndonuclease, 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, 2l3Bi, or other radioisotopes such as, for example, '03Pd, '33Xe, 13'1, "Ge, "Co, "Zn, '5Sr, 32P, 35S, *>Y, '"Sm, ISJGd, "5Yb, s'Cr, s |03X2| 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.
|0383| 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, proliferate 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). [0384] 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).
|0385| 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.
Diasnostic Assays
[0386| 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-Angiogcnesis 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.
|0387] 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.
[0388| 'Hie present invention is also useful as a prognostic indicator, whereby patients exhibiting enhanced or depressed gene expression will experience a worse clinical outcome.
|0389| 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.
[0390] By "biological sample" is intended any biological sample obtained from an individual, cell line, tissue culture, or other source containing polypeplides 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.
[03911 Total cellular RNA can be isolated from a biological sample using any suitable technique such as Ihe single-step guanidimum-thiocyanate-phenol-chloroform method described in Chomczynski and Sacchi, Anal. Biochcm. 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, SI 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).
|0392| 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 witn 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 polypcptides 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 radioimmunoassavs, competitive-binding assays. Western Blot analysis and EUSA assays. Assaying polypeptide levels in a biological sample can occur using any art-known method.
[0393| 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., 1. 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 immunosorbcnt assay (EL1SA) and the radioimmunoassay (RLA). Suitable antibody assay labels are known in the art and include enzyme labels, such as, glucose oxidase, and radioisotopcs, such as iodine ('2!l, 'J1I), carbon (MC), sulfur (35S), tritium (5H), indium (llzln), and technetium (w°Tc), and fluorescent labels, such as fluorescein and rhodarnine, and biotin
[0394] The tissue or cell type to be analyzed will generally include those which are known, or suspected, to express the gene of interest (such as, for example, cancer). The protein isolation methods employed herein may, for example, be such as those described in Harlow and Lane (Harlow, E. and Lane, D., 1988, "Antibodies: A Laboratory Manual", Cold Spring Harbor Laboratory Press, Cold Spring Harbor, New York), which is incorporated herein by reference in its entirety. The isolated cells can be derived 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.
[0395[ 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 immunofluoreseence techniques employing a fluorescently labeled albumin fusion protein coupled with light microscopic, flow cytometric, or fluorimetric detection. [0396] 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 peptidc fragments thereof. This can be accomplished, for example, by immunofluoreseence techniques employing a fluorescently labeled antibody coupled with light microscopic, flow cytometric, or fluorimetric detection.
[0397] The albumin fusion proteins of the present invention may, additionally, be employed histologically, as in immunofluoreseence, immunoclcctron microscopy or non-immunological assays, for in situ detection of polypcptides 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.
|0398| Immunoassays and non-immunoassays that detect polypcptides 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.
[0399] 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 delectably 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.
[0400] By "solid phase support or carrier" is intended any support capable of binding a polypeplide (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, porvacrylamides, gabbros, and magnetite. The nature of the carrier can be cither 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.
|0401] The binding activity of a given lot of albumin fusion protein maybe determined according to well known methods. Those skilled in the ar1 will be ahle to determine operative and optimal assay conditions for each determination by employing routine experimentation. [0402] 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, (04031 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 BSR 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. |0404] 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.
10405) A polypeptide-specific antibody or antibody fragment which has been labeled with an appropriate detectable imaging moiety, such as a radioisotope (for example, ml, "7In, "Tc), a radio-opaque substance, or a material detectable by nuclear magnetic resonance, is introduced (for example, parenterally, subcutaneously or intraperitoncally) 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 millicunes of "™Tc. 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 Radiolabcled 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)).
|0406] 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, Walkcrsville, MD); Voller et al., J. 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 Shoiri, Tokyo). The reporter enzyme which is bound to the antibody will react with an appropriate substrate, preferably a chromogcnic 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 dchydrogenase, alpha-glycerophosphate, dehydrogenasc, triose phosphate isomerase, horseradish peroxidase, alkaline phosphatase, asparaginase, glucose oxidase, beta-galactosidase, ribonudease, urease, catalase, glucose-6-phosphate dehydrogenase, glucoamylase and acetylcholinesterase. Additionally, the detection can be accomplished by colorimetric methods which employ a chromogenic substrate for the reporter enzyme. Detection may also be accomplished by visual comparison of the extent of enzymatic reaction of a substrate in comparison with similarly prepared standards.
Albumin fusion proteins may also be radiolabelled and used in any of a variety of other immunoassays. For example, by radioactively
labeling the albumin fusion proteins, it is possible to the use the albumin fusion proteins in a radioimmunoassay (RIA) (see, for example,
Weintrauh, B., Principles of Radioimmunoassays, Seventh Training Course on Radioligand Assay Techniques, The Endocrine Society, March,
1986, which is incorporated by reference herein). The radioactive isotope can be detected by means including, but not limited to, a gamma counter,
a scintillation counter, or autoradiography.
Additionally, chelator molecules, are blown 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 Mearcs, C.F., "Bifunctional Chelating Agents for Radiomctal-labeled monoclonal Antibodies," in Cancer
Imaging with Radiolabeled Antibodies (D. M. Goldcnberg, Ed.) Kluwer Academic Publications, Boston; Saji, H., "Targeted delivery of radiolabeled
imaging and therapeutic agents: bifunctional radiopharmaceuticals." Crit. Rev. Ther. Drug Carrier Syst. /rJ:209-244 (1999); Srivastava S.C. and
Mease R.C., "Progress in research on ligands, nuclides and techniques for labeling monoclonal antibodies." Int. J. Rad, Appl. Inslrum. B /<589-> 603 (1991); and Liu, S. and Edwards, D.S., "Bifunctional chelators for therapeutic lanthanide radiopharmaceuticals." Bioconjug. Chern. /2:7-34
(2001). Any chelator which can be covalcntly 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.
|0409] 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-
isothiocyanatobenzyl)-6- methyldiethylenetriaminepcntaacctic acid (1B4M-DPTA, also known as MX-DTPA), 2-melhyl-6-(rho-nitrobenzyl)-l,4,7-
triazaheptane-N,N,N',N",N"-pcntaacetic acid (nitro-lB4M-DTPA or nitro-MX-DTPA); 2-(p-isothiocyanatobenzyl)-
cyclohexyldiethylenetriaminepentaacetic acid (CHX-DTPA), or N-[2-amino-3-(rho-nitrophenyl)propyl]-trans-cyclohexane-l,2-diamine-N,N',N"-
pentaacetic acid (nitro-CHX-A-DTPA).
|0410) In another embodiment, the Albumin fusion protein of the invention arc attached to an acyclic terpyridine chelator such as 6,6"-
bis[[N,N,N",N"- tctra(carboxymelhyl)amino]methyl]-4l-(3-amino-4-methoxyphenyl)-2,2l:6',2 "- terpyridine (TMT-amine).
(0411] 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",N"'-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 al., Bioconjug. Chem. 70(4):553-7, 1999; and Zimmerman
el al., Nucl. Med. Biol. 2 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.
|0412] 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. Sac. 49:2639 (1989) (2-p-nitrobenzyl-l,4,7,10-
tetraazacyclododecane-N,N',N",N'"-tetraacctic acid): S. V. Deshpande ct al., J. Nucl. Mcd. 31:473 (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.,J. Nucl. Med. .35:850 (1995).
|0413] 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.
|0414] In one embodiment, suitable chelator molecules that may be attached to the the Albumin fusion protein of the invention include DOXA
(l-oxa-4,7,IO-triazacyclododecanetriacetic acid), NOTA (1,4,7-tria2acyclononanetriacetic acid), TETA (1,4,8,11-tetraazacyclotetradecanetetraacetic
acid), and TUT (4'-(3-aminoj}-methoxy-phenyl)-6,6"-bis(N',N'-dicarboxymethyl-W-methylhydra zino)-2,2':6',2"-terpyridine), and analogs and
derivatives thereof. See, e.g., Ohmono ct al., J. Med. Chem. 35: 157-162 (1992); ICung 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; WO89/00557; and EP-A-232751.
|0415| In another embodiment, suitable macrocyclic carboxylic acid chelators which can be used in the present invention include 1,4,7,10-
tetraazacyclododecane-A',W^V'^v7"-tetraacetic acid (DOTA); l,4,8,12-tetraazacyclopentadecane-W,A^'>A'"-tetTaacetic acid (15N4); 1,4,7-
triazacyclononane-N,N',N"-triacetic acid (9N3); l,5,9-tnazacyclododecane-N,N',N"-triacctic acid (12N3); and 6-bromoacetamido-benzyl-l,4,8,11-
telraazacyclotetradccane- A'^V^V",W"-tetraacetic acid (BAT).
|0416| A preferred chelator that can be attached to the Albumin Fusion protein of the invention is a-(5-isothiocyanato- 2-methoxyphenyl)-
1,4,7,10-tetraazacyclododecane-l,4,7,10-tetraacetic acid, which is also known as MeO-DOTA-NCS. A salt or ester of a-(5-isothiocyanato- 2-
mcthoxyphenyl)- l,4,7,10-tetraazacyclododecane-l,4,7,10-tetraacctic acid may also be used.
[0417] 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, '"In, 67Ga, or "Ga. In another embodiment, the radionuclide used for diagnostic purposes is
'"In, or "Ga. Examples of the radionuclide used for therapeutic purposes are '"Ho, '"Dy, ""Y, "!"ln, S2Fe, or 72Ga. In one embodiment, the
radionuclide used for diagnostic purposes is "*TIa or MY. Examples of the radionuclides used for both therapeutic and diagnostic purposes include
"'Sm, "7Lu, '"Gd, 17iYb, or 47Sc. In one embodiment, the radionuclide is '"Sm, "7Lu, n!Yb, or "'Gd.
|0418| Preferred metal radionuclides include 90Y, "Tc, '"In, '77"Sn, 67Cu, ""Tm, "Ru, "8Re, '77Lu, "9Au, mPb and "'Ce.
|0419| 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 consisting of "°Y, '"in, '77Lu, "6Ho, 2"Bi, and "5Ac.
(0420] Moreover, y-emitting radionuclides, such as ""Tc, '"In, *7Ga, and "'Yb have been approved or under investigation for diagnostic
imaging, while p-cmilters, such as 61Cu, "'Ag, '"Re, and "V are useful for the applications in tumor therapy. Also other useful radionuclides
include y-emitters, such as Tc, '"in, "Ga, and "'Yb, and p-emitters, such as interest such as 2"At, 2'2Bi, 177Lu, "Rb , loiRh, ISISm, '"Au, "'Pm, "Sr. H2Pr, 2"Pb, 10'Pd, '"Ho, 2 invention to which chelators are covalently attached maybe labeled with the radionuclides described above.
[0421] 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.
[0422] In another embodiment, Albumin fusion proteins of the invention to which chelators are covalently attached may be labeled with
fliinrf-Trnt metal ions including lanthanides, in particular I.a, O, Pr, Nd, Pm. Sm, Hu (eg., IMEuV Gd. Tb, Oy, Ho, Er, Tm. Yb, and 1 u.
[0423J 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.
|0424| 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, rhodaminc, phycoerythrin, phycocyanin, allophycocyanin, ophthaldehyde and fluorescamine.
|0425| The albumin fusion protein can also be dctcctably labeled using fluorescence emitting metals such as 1S2Eu, 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)
[0426| The albumin fusion proteins can also can be detectably labeled by coupling it to a chemiluminescent compound. The presence of the
chcmiluminesccnt-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.
[0427| 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 lucifcrin, luciferase and aequorin.
TranSKenic Organisms
[0428| 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 recornbinant, 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. |0429| 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.
[0430] 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 recornbinant viral and retroviral infection (see. e.g., U.S. Patent No. 4,736,866; U.S. Patent No. 5,602,307; Mullins et at. (1993) Hypertension 22(4):630-633; Brenin el ai (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.
[0431] A number of recornbinant 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 (1RF-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).
[0432] 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, eg., Kim et al. (1997) Mol, Reprod. Dev.
46(4):515-526; Houdebme (,iy«j Keprod. Nulr. Dev. 35(6):609-617; Petters (1994) Reprod. Fertil. Dev. 6(5):643-645; Schnieke cl al. (1997) Science 278(5346):2130-2133; and Amoah (1997) J. Animal Science 75(2):578-585).
[0433] To direct the secretion of the transgene-encoded protein of tlie 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 el al. (1989) BioTechnology 7:487^192; Gorton el at. (1987) BioTechnology 5:1183-1187; and Soulier et at. (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.
[0434] 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 ("Rccombinant 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, all of which are hereby incorporated by reference.
Pharmaceutical or Therapeutic Compositions
[0435] 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.
|0436| 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. |0437] 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.
[0438] 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-bome 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. [0439] 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 epitopcs appear to the human immune system to be human in nature rather than foreign. |0440| 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 bolh, and then, if necessary, shaping the product.
|0441| Formulations suitable for parenteral administration include aqueous and non-aqueous sterile injection solutions which may contain anli-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.
[0442] As an example, when an albumin fusion protein of the invention comprises one of the proteins listed in the "Therapeutic Protcin:X" column of Table I 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 tne potency 01 the therapeutic protein alone, while taking into account the prolonged serum half-life and shelf-life of the albumin fusion proteins compared to that of native therapeutic protein. For example, if the therapeutic protein is typically administered at 0.3 to 30.0 IU/kg/week, or 0.9 to 12.0 IU/kg/wcek, 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.
[0443] 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.
|0444] 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.
|0445| 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. |0446| 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.
|0447] 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 I -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.
[0448] As noted above, the albumin fusion protein of the invention has a higher plasma stablity compared to the Therapeutic protein portion (or fragment or variant thereof) alone. This increase in plasma stability should be taken into account when determining the effective amount of the albumin fusion protein to be administered per dose and the dosing administration schedule. In particular, higher plasma stability may allow the albumin fusion protein to be administered at a lower dose at the same frequency of administrations, or alternatively, may allow the albumin fusion protein to be administered in fewer dosings. Preferably, the higher stability allows the albumin fusion protein of the invention to be administered less oflen in fewer dosings. More preferably, the albumin fusion protein can be administered once every two weeks. Still more preferably, the albumin fusion protein can be administered once every three, four, five, or more weeks depending on the pharmacokinetics of the albumin fusion piotein. For example, as discussed above, the pharmacokinetics of an IFN-alpha-HSA fusion protein supports a dosing regimen of once every 1-4 weeks or more, and even dosing at intervals of 4 weeks or more than every 4 weeks.
|0449| Albumin fusion proteins and/or polynucleotides can be are administered orally, rectally, parenterally, intracistemally, intravaginally, intraperitoneally, topically (as by powders, ointments, gels, drops or transdcrmal patch), bucally, or as an oral or nasal spray. "Pharmaceutically acceptable carrier" refers to a non-toxic solid, scmisolid or liquid filler, diluent, encapsulating material or formulation auxiliary of any. The term "parenleral" as used herein refers to modes of administration which include intravenous, intramuscular, intraperitoneal, intrastemal, subcutaneous and mtraarticular injection and infusion.
|0450| Albumin fusion proteins and/or polynucleotides of the invention are also suitably administered by sustained-release systems. Examples of suslained-release albumin fusion proteins and/or polynucleotides are administered orally, rectally, parenterally, intracistemally, intravaginally, mtraperitoncally, 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 lerm "parcntcral" as used herein refers to modes of administration which include intravenous, intramuscular, intraperitoneal, intrastemal, subcutaneous and inlraarticular 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 mirocapsulcs), suitable hydrophobic materials (for example as an emulsion in an acceptable oil) or ion exchange resins, and sparingly soluble derivatives (such ns, for exampK a sparingly soluble salt)
|0451) Sustained-release matrices include polylactides (U.S. Pat. No. 3,773,919, EP 58,481), copolymcrs of L-glutamic acid and gamma-ethyl-L-glutamate (Sidman el 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-hydroxyburyric acid (EP 133,988).
|0452) 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 ct 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.
(0453) In yet an additional embodiment, the albumin fusion proteins and/or polynucleotides of the invention are delivered by way of a pump (sec 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)).
|0454] Other controlled release systems are discussed in the review by Langer (Science 249:1527-1533 (1990)).
|0455] 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. J04S6) Generally, the formulations are prepared by contacting the albumin fusion protein and/or polynucleotide uniformly and intimately with liquid earners 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 arc also useful herein, as well as liposomes.
|0457] 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., polyargininc or tripeptides; proteins, such as scrum albumin, gelatin, or immunoglobulins; hydrophilic polymers such as polyvinylpyrrolidonc; amino acids, such as glycme, glulamic acid, aspartic acid, or argininc; monosaccharides, disacchandcs, and other carbohydrates including cellulose or its derivatives, glucose, manose, or dextrins; dictating agents such as EDTA; sugar alcohols such as mannitol or sorbitol; counterions such as sodium; and/or nonionic surfactants such as poiysorbates (including, for example, Tween-20), poloxamers, or PEG.
[0458] 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.
[0459| 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. [0460| 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. [04611 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/milligTarn 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/niilliliter polysorbate 80, pH 7.2. The pH and buffer are chosen to match physiological conditions and the salt is added as a tonicifier. Sodium octanoatc 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 01 me ainumin fusion protein to the container closure system.
|04621 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 polynuclcotides 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.
|0463| 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.), QS21 (Genentech, Inc.), BCG (e.g., THERACYS®), MPL and nonviable preparations of Corynebcicterium parwm. 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 lOOa, 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, tctanus/diptheria, hepatitis A, hepatitis B, Haemophilus influenzae B, whooping cough, pneumonia, influenza, Lyme's Disease, rolavirus, 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.
|0464) 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, chemotherapcutic 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.
|0465| 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., COUMADIN®), dicumarol, 4-hydroxycoumarin, anisindione (e.g., MIRADON™), accnocournarol (e.g., nicoumalone, SINTHROME™), indan-l,3-dione, phenprocoumon (e.g., MARCUMAR™), ethyl biscoumacetate (e.g., TROMEXAN™), 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.
|()466| In another embodiment, the albumin fusion proteins and/or polynucleotides of the invention are administered in combination with ihrornbolytic 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., KABIK1NASE™), anliresplace (e.g., EMINASE™), tissue plasminogen activator (t-PA, altevase, ACTIVASE™), urokinase (e.g., ABBOKINASE™), sauruplasc, (I'rourokinase, single chain urokinasc), and aminocaproic acid (e.g., AM1CAR™). In a specific embodiment, compositions of the invention are administered in combination with tissue plasminogen activator and aspirin.
|0467| In another embodiment, the albumin fusion proteins and/or polynucleotides of the invention are administered in combination with antiplatelct drugs. Antiplatelet drugs that may be administered with the compositions of the invention include, but are not limited to, aspirin, dipyridamole (e.g., PERSANTINE™), and ticlopidine (e.g., T1CL1D™).
|0468) 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
polynuclcotides ot 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 hcmodialysis patients, hemodialysis machines, and cardiopulmonary bypass machines).
(04691 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, RETROV1R™ (zidovudine/AZT), VIDEX™ (didanosine/ddl), H1VID™ (zalcitabine/ddC), ZER1T™ (stavudineAMT), EPIVIR™ (lamivudine/3TC), and COMBIVIR™ (zidovudine/lamivudine). NNRTIs that may be administered in combination with the albumin fusion proteins and/or polynucleotides of the invention, include, but arc not limited to, VIRAMUNE™ (nevirapine), RESCR1PTOR™ (delavirdine), and SUSTIVA™ (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, CRIXIVAN™ (indinavir), NORVIR™ (ritonavir), INVIRASE™ (saquinavir), and VIRACEPT™ (nelfinavir). In a specific embodiment, antiretroviral agents, nucleoside reverse transcriptase inhibitors, non-nuclcoside 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.
10470] Additional NRTIs include LODENOSINE™ (F-ddA; an acid-stable adenosine NRTI; Triangle/Abbott; COV1RACIL™ (cmtricitabinc/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); Adcfovir (refused approval for anti-HPV therapy by FDA; Gilead Sciences); PREVEON® (Adefovir Dipivoxil, the active prodrug of adefovir; its active form is PMEA-pp); TENOFOVIR™ (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); ZIAGEN™ (abacavir/159U89; Glaxo Wellcome Inc.); CS-87 (3'azido-2',3'-dideoxyuridine; WO 99/66936); and S-acyl-2-thiocthyl (SATE)-bcaring prodrug forms of P-L-FD4C and p-L-FddC (WO 98/17281).
10471] Additional NNRTIs include COACTINON1" (Emivirinc/MKC-442, potent NNRTI of the HEPT class; Triangle/Abbott); CAPKAVIR1NE™ (AG-I549/S-1153, a next generation NNRTI with activity against viruses containing the K.1G3N 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 K.103N mutation; DuPont); GW-420867X (has 25-fold greater activity than HBY097 and is active against K.103N 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). (04721 Additional protease inhibitors include LOPINAV1R™ (ABT378/r; Abbott Laboratories); BMS-232632 (an azapeptide; Bristol-Myres Squibb); TIPRANAVIR™ (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-I776 (a peptidomimetic with in vitro activity against protease inhibitor-resistant viruses; Agouron); VX-175/CW-433908 (phosphate prodrug of amprenavir; Vertex & Glaxo Welcome); CGP61755 (Ciba); and AGENERASE™ (amprenavir; Glaxo Wellcome Inc.). |04731 Additional antiretroviral agents include fusion inhibitors/gp41 binders. Fusion inhibitors/gp41 binders include T-20 (a peptidc from residues 643-678 of the HIV gp4l transmenibrane protein ectodomain which binds to gp41 in its resting state and prevents transformation to the fusogcnic state; Trimeris) and T-1249 (a second-generation fusion inhibitor; Trimeris).
(04741 Additional antiretroviral agents include fusion inhibitors/chemokine receptor antagonists. Fusion inhibitors/chemokme 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 TI40; CCR5 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, MlP-la, MlP-lp, etc., may also inhibit fusion.
|047S] Additional antirctroviral agents include integrase inhibitors. Integrase inhibitors include dicaffeoylquinic (DFQA) acids; L-chicoric acid (a dicaffeoyltartaric (DCTA) acid), quinalizarin (QLC) and related anlhraquinones; ZINTEVIR™ (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.
[0476] Additional antiretroviral agents include hydroxyurea-like compunds such as BCX-34 (a purine nucleoside phosphorylase inhibitor; Biocryst), ribonuclcotide reductase inhibitors such as Dll)OXTU (Molecules for Health); inosine monophosphate dehydrogenase (IMPDH) inhibitors sucha as VX-497 (Vertex); and mycopholic acids such as CellCcpt (mycophenolate mofetil; Roche).
|0477| Additional antiretrovirai 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 phannacoenhancers such as ABT-378.
I0478J Other antiretrovirai therapies and adjunct therapies include cytokines and lymphokines such as MiP-la, MDP-lfi, SDF-la, 1L-2, PROLEUKIN™ (aldesleukin/U-7001; Chiron), IL-4, IL-IO, IL-12, and IL-13; interferons such as IFN-alpha2a, IFN-aIpha2b, or IFN-beta; antagonists of TNFs, NFKB, GM-CSF, M-CSF, and IL-10; agents that modulate immune activation such as1 cyclosporin and prednisone; vaccines such as Remune™ (HIV Immunogen), APL 400-003 (Apollon), recombinant gp!20 and fragments, bivalent (B/E) recombinant envelope glycoprotein, rgp!20CM235, MN rgp!20, SF-2 rgp!20, gp!20/soluble CD4 complex, Delta JR-FL protein, branched synthetic peptide derived from discontinuous gp!20 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 ai. PNAS 94:\ 1567-72 (1997); Chen el at, Nat. Med. 3:1110-16 (1997)); antibodies such as the anti-CXCR4 antibody 12G5, the anti-CCR5 antibodies 2D7, 5C7, PA8, PA9, PAIO, PAl 1, PA12, and PA14, the anti-CD4 antibodies Q4120 and RPA-T4, the anti-CCR3 antibody 7B11, the anti-gp!20 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 (v-GCE; WO 99/56764). [0479] In a further embodiment, the albumin fusion proteins and/or polynucleotldes 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, rernantidine, 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.
|U480| In a most preferred embodiment, interferon albumin fusion protein is administered in combination with ribavirin. In a further preferred emhodiment, 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.
[0481] 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, TR1METHOPRIM-SULFAMETHOXAZOLE™, DAPSONE™, PENTAMIDFNE™, ATOVAQUONE™, ISONIA/.IO™, RIFAMPIN™, PYRAZINAM1DE™, ETHAMBUTOL™, RIFABUTIN™, CLARITHROMYCIN™, AZITHROMYCIN™, GANCICLOVIR™, FOSCARNET™, CfDOFOVIR™, FLUCONAZOLE™, ITRACONAZOLE™, KETOCONAZOLK™, ACYCI.OVIR™, FAMCICOLVIR™, PYR1METHAMINETU, LEUCOVORINTU, NEUPOGEN™ (filgrastinVG-CSF), and LEUKINE™ (sargramostim/GM-CSF). In a specific embodiment, albumin fusion proteins and/or polynucleotides of the invention are used in any combination with TRIMETHOPRIM-SULFAMETHOXAZOLE™. DAPSONE™, PENTAMIDINE™, and/or ATOVAQUONE™ 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 ISONIAZID™, RfFAMPIN™, PYRAZINAMIDE™, and/or ETHAMBUTOL™ to prophylactically treat or prevent an opportunistic Mycobuctcrium avium complex infection. In another specific embodiment, albumin fusion proteins and/or polynucleotides of the invention are used in any combination with RIFABUTFN™, CLARITHROMYCIN™, and/or AZ1THROMYCINP" 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 GANCICLOVIR™, FOSCARNET™, and/or CIDOFOVIR™ to prophylactically treat or prevent an opportunistic cytomcgalovirus infection. In another specific embodiment, albumin fusion pioteins and/or polynucleotides of the invention are used in any combination with FLUCONAZOLE™, 1TRACONAZOLE™, and/or K.ETOCONAZOLE™ 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 ACYCLOVIR™ and/or FAMCICOLVIR™ 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 PYRIMETHAMINE™ and/or LEUCOVORIN™ 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 LEUCOVOR1N™ and/or NEUPOGEN™ to prophylactically treat or prevent an opportunistic bacterial infection.
10482] 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, bela-lactamases, aminoglycosides, beta-lactam (glycopeptide), beta-lactamases, Clindamycin, chloramphenicol, cephalosporins, ciprofloxacin, erythromycin, fluoroquinolones, macrolides, melronidazole, penicillins, quinolones, rapamycin, rifampin, streptomycin, sulfonamide, tetracyclines, trimethoprim, trimethoprim-sulfamethoxazole, and vancomycin.
|0483| 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., ERGAMISOL™), isoprinosine (e.g. 1NOS1PLEX™), interferons (e.g. interferon alpha), and interleukins (e.g., IL-2).
|0484] In other embodiments, albumin fusion proteins anoVor 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 (BREDININ™), brequinar, deoxyspergualin, and azaspirane (SKF 105685), ORTHOCLONE OK.T® 3 (muromonab-CD3), SANDIMMUNE™, NEORAL™, SANGDYA™ (cyclosporine), PROGRAF® (FKS06, tacrolimus), CELLCEPT® (mycophenolate motefil, of which the active metabolite is mycophenolic acid), IMURAN™ (azathioprine), glucocorticosteroids, adrenocortical steroids such as DELTASONE™ (prednisone) and HYDELTRASOL™ (prednisolone), FOLEX™ and MEXATE™ (methotrxate), OXSORALEN-ULTRA™ (methoxsalen) and RAPAMUNE'" (sirolimus). In a specific embodiment, immunosuppressants may be used to prevent rejection of organ or bone marrow transplantation. (0485) 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, GAMMAR™, 1VEEGAM™, SANDOGLOBULIN™, GAMMAGARD S/D™, ATGAM™ (antithymocyte glubulin), and GAM1MUNE™. 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).
|0486| 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 lL-2-albumin fusions, are administered repeatedly during passive immunotherapy for cancer, such as adoptive cell transfer therapy for metastatic melanoma as described in Dudley el at. (Science Express, 19 September 2002.. at www.scienceexpress.org, hereby incorporated by reference in its entirety).
|0487| 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, meclofenamale, 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, pyrazolcs, pyrazoloncs, salicylic acid derivatives, thiazmccarboxamides, e-acetamidocaproic acid, S-adenosylmethionine, 3-amino-4-hydroxybutyric acid, amixetrine, bcndazac, benzydamine, bucolome, difenpiramide, ditazol, emorfazone, guaiazulene, nabumetone, nimesulide, orgotein, oxaceprol, paranyline, perisoxal, pifoxime, proquazone, proxazolc, and tenidap.
[0488| 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, Roclcville, MD), Troponin-1 (Boston Life Sciences, Boston, MA), anti-Invasive Factor, retinoic acid and derivatives thereof, paclitaxel (Taxol), Suramin, Tissue Inhibitor of Mctalloproteinase-l, Tissue Inhibitor of Metalloproteinase-2, VEGI, Plasminogen Activator Inhibitor-1, Plasminogen Activator lnhibitor-2, and various forms of the lighter "d group" transition rnetals.
[0489] 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.
[0490] Representative examples of vanadium complexes include oxo vanadium complexes such as vanadate and vanadyl complexes. Suitable vanad.itc complexes include metavanadate and orthovanadate comnlexes 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 trihydratcs.
[0491] Representative examples of tungsten and molybdenum complexes also include oxo complexes. Suitable oxo tungsten complexes include tungstale 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, molybdeny] acetylacetonate. Other suitable tungsten and molybdenum complexes include hydroxo derivatives derived from, for example, glycerol, tartaric acid, and sugars.
[0492] 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-dehydropro!ine, Thiaproline, alpha,alpha-dipyridyl, aminopropionitrile fumarate; 4-propyl-5-(4-pyridinyl)-2(3H)-oxazolone; Methotrexate; Mitoxantrone; Heparin; Interferons; 2 Macroglobulin-serum; ChIMP-3 (Pavloff et al., J. Bio. Chem. 267:17321-17326, (1992)); Chymostatin (Tomkinson et al., Biochem J. 286:475^180, (1992)); Cyclodextrin Tetradecasulfate; Eponcmycin; 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-antiptasmin (Holmes et al., J. Biol. Chem. 262(4): 1659-1664, (1987)); Bisantrene (National Cancer Institute); Lobenzarit disodium (N-(2)-carboxyphcnyl-4- chloroanthronilic acid disodium or "CCA"; (Takeuchi et al.. Agents Actions 36:312-316, (1992)); and metalloproteinase inhibitors such as BB94.
[0493] 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. Hrem and J. Folkman JPediatr. 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); Conbrctastatin A^l (CA4P) (OXiGENE, Boston, MA); Squalamine (Magainin Pharmaceuticals, Plymouth Meeting, PA); TNP470, (Tap Pharmaceuticals, Deerfield, IL); ZD-OI01 AstraZeneca (London, UK); APRA (CT2584); Benefin, Byrostatin-l (SC339555); CGP-41251 (PKC 412); CM101; Dexrazoxane (ICRP187); DMXAA; Endostatin; Flavopridiol; Gcncstein; GTE; ImmTher; Iressa (ZD1839); Octreotide (Somatostatin); Panretin, Penacillamine; Photopoint; PI-88; Prinomastat (AG-3340) Purlytin; Suradista (FCE26644); Tamoxifen (Nolvadcx); Tazarotene; Tetrathiomolybdate; Xeloda (Capccitabine); and 5-Fluorouracil.
[0494| 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 ofangiogenesis induccrs 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 arc 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 ftiotcch, Oxford, UK), and Metastat (Aetema, 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, Gaithcrsburg, 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), Anli-VEGF antibody (Genentech, S. San Francisco, CA), PTK.-787/ZK.-225846 (Novartis, Basel, Switzerland), SU-101 (Sugen, S. San Francisco, CA), SU-5416 (Sugcn/ Pharmacia Upjohn, Bridgewater, NJ), and SU-6668 (Sugcn). 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), Interfcron-alpha, IL-12 (Roche, Nulley, NJ), and Pcntosan polysulfate (Georgetown University, Washington, DC). |0495| 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. |0496| In a particular embodiment, the use of compositions of the invenlion in combination with anti-angiogenic agents is contemplated for the treatment, prevenlion, 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.
10497] In another embodiment, tne polynucleotides encoding a polypcptide of the present invention are administered in combination with an angiogenic protein, or polynucleotides encoding an angiogenic protein. Examples of angiogenic proteins thai may be administered with the compositions of the invention include, but are not limited to, acidic and basic fibroblast growth factors, VEGF-I, 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.
10498) 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, Carmustinc (BCNU), Lomustine (CCNU), Semustine (methyl-CCNU), and Streptozocin (strcptozotocin)), triazenes (for example, Dacarbazine (DTIC; dimethyltriazenoimidazolecarboxamidc)), folic acid analogs (for example, Methotrexale (amethopterin)), pyrimidine analogs (for example, Fluorouacil (5-fluorouracil; 5-FU), Floxuridine (fluorodeoxyundine; I-udR), 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 intcrfcron-alpha-2b), platinum coordination compounds (for example, Cisplatin (cis-DDP) and Carboplatin), anthracenedionc (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 Fluoxyrnesterone), antiandrogens (for example, Flutamide), gonadotropin-releasing horotnone 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).
|()499| In one embodiment, the compositions of the invention are administered in combination with one or more of the following drugs: infliximab (also known as Remicade™ Centocor, Inc.), Trocade (Roche, RO-32-3555), Lcflunomide (also known as Arava™ from Hoechst Marion Roussel), Kineret™ (an IL-1 Receptor antagonist also known as Anakinra from Amgen, Inc.)
|0500| 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 Riluximah 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 tositurnomah 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.
|050I | In another specific embodiment, the compositions of the invention are administered in combination Zevalin"1. In a further embodiment, compositions of the invention are administered with Zevalin™ and CHOP, or Zevalin™ and any combination of one or more of (he components of CHOP, particularly cyclophosphamide and/or prednisone. Zevalin™ may be associated with one or more radisotopes. Particularly preferred isotopes are ""Y and '"in.
|0502| In an additional embodiment, the albumin fusion proteins and/or polynucleotides of the invention are administered in combination with cytokines. Cytokines that may he administered with the albumin fusion proteins and/or polynucleotides of the invention include, but are not limited to, 11.2, 1L3, !L4, IL5, IL6, IL7, IL10, IL12, ILI3, IL15, anti-CD40, CD40L, IFN-gamma and TNF-alpha. In another embodiment, albumin fusion proieins and/or polynucleotides of the invention may be administered with any interleukin, including, but not limited to, IL-lalpha, IL-lbeta, 1L-2, I1.-3, IL-4, IL-5, 1L-6, IL-7, IL-8,1L-9, 11.-10, IL-I I, IL-12,1L-13, IL-14, IL-15. IL-16, IL-17, IL-18, IL-19, IL-20, and IL-21.
|0503| In one embodiment, the albumin fusion proteins and/or polynucleotides of the invention arc 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-alpba, also known as TNF-beta), LT-beta (found in
complex heterotrimcr LT-alpha2-beta), UI'GL, FasL, CD27L, CD30L, CD40L, 4-1BBL, DcR3, OX40L, TNF-gamma (International Publication No. WO 96/14328), AIM-1 (International Publication No. WO 97/33899), endokine-alpha (International Publication No. WO 98/07880), OPG, and neutrokine-alpha (International Publication No. WO 98/18921, OX40 and nerve growth factor fNGF), and soluble forms of Fas, CD30, CD27, CD40 and 4-IBD, 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 CDI54, CD70, andCD153.
|0504] 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-D (PDGF-B), as disclosed in European Patent Number EP-282317; Placental Growth Factor (P1GF), as disclosed in International Publication Number WO 92/06194; Placental Growth Factor-2 (P1GF-2), as disclosed in Mauser ct 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. The above mentioned references are herein incorporated by reference in their entireties.
[0505] In an additional embodiment, the albumin fusion proteins and/or polynucleotides of the invention are administered in combination with Fihroblast Growth Factors. Fibroblast Growth Factors that may be administered with the albumin fusion proteins and/or polynucleotides of the invention include, but are not limited to, FGF-I, FGF-2, FGF-3, FGF-4, FGF-5, FGF-6, FGF-7, FGF-8, FGF-9, FGF-10, FGF-11, FGF-12, FGF-13, FGF-14, andFGF-15.
|0506] In an additional embodiment, the albumin fusion proteins and/or polynucleotides of the invention are administered in combination with hcmatopoietic 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, LEUKINE™, 1'ROK.INE™), granulocyte colony stimulating factor (G-CSF) (filgrastim, NEUPOGEN™), macrophage colony stimulating factor (M-CSF, CSF-1) erythropoietin (epoetin alfa, EPOGEN™, PROCRIT™), 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, intcrferon-gamma, or thrombopoictin. |0507] 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.
[0508] In another embodiment, the albumin fusion proteins and/or polynucleotides of the invention are administered in combination with an antiarrhythmic drug (e.g., adenosine, amidoarone, bretylium, digitalis, digoxin, digitoxin, diliazem, disopyramide, esmolol, flecainide, lidocainc, mexiletine, rnoricizine, phenytoin, procainamide, N-acetyl procainamide, propafcnone, propranolol, quinidine, sotalol, tocainide, and verapamil). |0509] In another embodiment, the albumin fusion proteins and/or polynucleotides of the invention are administered in combination with diuretic agents, such as carbonic anhydrasc-inhibiting agents (e.g., acetazolamide, dichlorphenamide, and methazolamide), osmotic diuretics (e.g., glycerin, isosorbide, mannitol, and urea), diuretics that inhibit Na'-IC*-2Cl' symport (e.g., furosemide, bumetanide, azosemide, piretanide, tripamidc, ethacrynic acid, muzolimine, and torsemidc), thiazide and thiazide-like diuretics (e.g., bendroflumethiazide, benzthiazide, chlorothiazide, hydrochlorothiaiide, hydrotlumcthiazide, methyclothiazide, polythiazide, trichormethiazide, chlorthalidone, indapamide, metolazone, and quinetharone), potassium sparing diuretics (e.g., amiloride and triamterene), and mineralcorticoid receptor antagonists (e.g., spironolactone, canrcnone, and potassium canrenoate).
|0510| In one embodiment, ihe 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, '"], radioactive isotopes of iodine such as '"I and '"I; recombinant growth hormone, such as HUMATROPE™ (recombinant somatropin); growth hormone analogs such as PROTROPIN™ (somatrem); dopamine agonists such as PARLODEL™ (bromocriptine); somatostatin analogs such as SANDOSTATIN™ (octreotide); gonadotropin preparations such as PREGNYLTL SUPPRF.IJN™ (histrelin acetate), SYNARHL™ (nafarelin acetate), and ZOLADEX™ (goserelin acetate); synthetic preparations of thyrotropin-releasmg hormone such as RELEFACT TRH™ and THYPINONE™ (protirclin); recombinant human TSH such as THYROGEN™; synthetic preparations of the sodium salts of the natural isorners of thyroid hormones such as L-T4™, SYNTHROID™ and LEVOT! IROID™ (Icvothyroxinc sodium), L-Tj™, CYTOMEL™ and TRIOSTAT™ (liothyroine sodium), and THYROLAR™ (liotrix); antithyroid compounds such as 6-n-propylthiouracil (propylthiouracil), l-methyl-2-mercaptoimidazo!e and TAPAZOLE™ (methimazole), NEO-MERCAZOLE™ (carbimazole); beta-adrenergic receptor antagonists such as propranolol and esmolol; Ca" channel blockers; dcxamethasonc and iodinated radiological contrast agents such as TELEPAQUE™ (iopanoic acid) and ORAGRAFIN™ (sodium ipodate).
|0511) Additional treatments for endocrine and/or hormone imbalance disorders include, but are not limited to, estrogens or congugated estrogens such as ESTRACE™ (estradiol), ESTINYL™ (ethinyl estradiol), PREMARIN™, ESTRATAB™, ORTHO-EST™, OGEN™ and cstropipate (estrone), ESTROVIS™ (quinestrol), ESTRADERM™ (estradiol), DELESTROGEN™ and VALERGEN™ (estradiol valerate), DEPO-KSTRAD10L CYPIONATE™ and ESTROJECT LA™ (estradiol cypionate); antiestrogens such as NOLVADEX™ (tamoxifen), SEROPHENE™ and CLOMID™ (clomiphene); progcstins such as DURALUTIN™ (hydroxyprogesterone caproate), MPA™ and DEPO-PROVERA™ (medroxyprogesterone acetate), PROVERA™ and CYCR1N™ (MPA), MEGACE™ (megestrol acetate), NORLUTIN™ (norethindrone), and NORLUTATE™ and AYGEST1N™ (norethindrone acetate); progesterone implants such as NORPLANT SYSTEM™ (subdermal implants of norgestrel); antiprogestins such as RU 486TU (mifepristone); hormonal contraceptives such as ENOVID™ (norethynodrel plus mestranol), PROGESTASERT™ (intrauterine device that releases progesterone), LOESTRIN™, BREVICON™, MODICON™, GENORA™, NELONA™, NORINYL™, OVACON-35™ and OVACON-50™ (ethinyl estradiol/norethindrone), LEV1.EN™, NORDETTE™ TRI-LEVLEN™ and TRIPHASII.-21™ (ethinyl estradiol/levonorgestrel) LO/OVRAL™ and OVRAL™ (ethinyl estradiol/norgestrel), DEMULEN™ (ethinyl estradiol/ethynodiol diacetate), NORINYLTU, ORTHO-NOVUM™, NORETHIN™, GENORA™, and NELOVATU (norcthindrone/mestranol), DESOGEN™ and ORTHO-CEPT™ (ethinyl cstradiol/desogestrel), ORTHO-CYCLEN™ and ORTHO-TRICYCLEN™ (ethinyl ostradiol/norgestimatc), M1CRONOR™ and NOR-QD™ (norethindrone), and OVRETTE™ (norgestrel).
|0512| 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-50™ (testosterone), TESTEX™ (testosterone propionate), DELATESTRYL™ (testosterone enanthate), DEPO-TESTOSTERONE™ (testosterone cypionate), DANOCRINE™ (danazol), HALOTESTIN™ (fluoxymesterone), ORETON METHYL™, TESTRED™ and VlRILON™ (methyltestosterone), and OXANDRIN™ (oxandrolone); testosterone transdermal systems such as TESTODERM™; androgen receptor antagonist and 5-alpha-reductase inhibitors such as ANDROCUR™ (cyproterone acetate), EULEXIN™ (flutamide), and PROSCAR™ (finasteride), adrenocorticotropic hormone preparations such as CORTROSYN™ (cosyntropin); adrenocortical steroids and their synthetic analogs such as ACLOVATE™ (alclometasone dipropionate), CYCLOCORT™ (amcmonide), BECLOVENT™ and VANCER1L™ (beclomethasone dipropionate), CELESTONE™ (betamethasone), BENISONE™ and UTICORT™ (betamethasone benzoatc), DIPROSONE™ (betamethasone dipropionate), CELESTONE PHOSPHATE™ (betamethasone sodium phosphate), CELESTONE SOLUSPAN™ (betamethasone sodium phosphate and acetate), BETA-VAL™ and VALISONE™ (betamethasone valerate), TEMOVATE™ (clobetasol propionate), CLODERM™ (clocortolone pivalate), CORTEF™ and HYDROCORTONE™ (cortisol (hydrocortisone)), HYDROCORTONE ACETATE™ (cortisol (hydrocortisone) acetate), LOCOID™ (cortisol (hydrocortisone) butyrate), HYDROCORTONE PHOSPHATE™ (cortisol (hydrocortisone) sodium phosphate), A-HYDROCORT™ and SOLU CORTEF™ (cortisol (hydrocortisone) sodium succinate), WESTCORT™ (cortisol (hydrocortisone) valerate), CORTISONE ACETATE™ (cortisone acetate), DESOWEN™ and TRIDESILON™ (desonide), TOP1CORT™ (desoximetasone), DECADRON™ (dexamelhasone), DECADRON LA™ (dexamethasone acetate), DECADRON PHOSPHATE™ and HEXADROL PHOSPHATE™ (dexamethasone sodium phosphate), FLORONE™ and MAX1FLOR™ (diflorasone diacetate), FLORINEF ACETATE™ (fludrocortisone acetate), AEROBID™ and NASAL1DE™ (flunisolide), FLUON1D™ and SYNALAR™ (fluocinolone acetonidc), L1DEX™ (fluocinonide), FLUOR-OP™ and FML™ (nuorometholone), CORDRAN™ (flurandrenolide), HALOG™ (halcinonide), HMS LIZUIFILM™ (medrysone), MEDROL™ (methylprednisolone), DEPO-MEDROL™ and MEDROL ACETATE™ (mcthylprednisonc acetate), A-METHAPRED™ and SOLUMEDROL™ (methylprednisolone sodium succinate), ELOCON™ (mometasone furoate), HALDRONE™ (paramethasone acetate), DELTA-CORTEF™ (prcdnisolonc), ECONOPRED™ (prednisolone acetate), HYDELTRASOL™ (prednisolone sodium phosphate), HYDELTRA-T.B.A™ (prednisolone tebutate), DELTASONE™ (prednisone), ARISTOCORT™ and JCENACORT™ (triamcinolonc), KENALOG™ (triamcinolone acetonide), ARISTOCORT™ and K.ENACORT DIACETATE™ (triamcinolone diacetate), and ARISTOSPAN™ (triamcinolone hexacetonide); inhibitors of biosynthesis and action of adrenocortical steroids such as CYTADREN™ (aminoglutethimide), NIZORAL™ (kctoconazole), MODRASTANE™ (trilostane), and METOPIRONE™ (mctyrapone); bovine, porcine or human insulin or mixtures thereof; insulin analogs; recombinant human insulin such as HUMULIN™ and NOVOLIN™; oral hypoglycemic agents such as ORAMIDE™ and ORINASE™ (tolbutamide), DIAB1NESE™ (chlorpropamide), TOLAMIDE™ and TOUNASE™ (tolazamidc), DYMELOR™ (acetohexamidc), glibenclamide, M1CRONASE™, DIBETA™ and GLYNASE™
(glyburide), GLUCOTROL1" (glipizide), and DIAMICRON™ (gliclazidc), GLUCOPHAGE™ (metformin), ciglitazone, pioglitazone, and alpha-glucosidase inhibitors; bovine or porcine glucagon; somatostatins such as SANDOSTATIN™ (octreotide); and diazoxides such as PROGLYCEM™ (dia?oxide).
[0513] 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/medroxyprogestcrone (e.g., PREMPRO™ and PRKMPHASE*) and norethindronc acetatc/ethinyl estsradiol (e.g., FEMHRT™).
|0514| 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, FEOSOL™), ferrous fumarate (e.g., FEOSTAT™), ferrous gluconate (e.g., FERGON™), polysaccharide-iron complex (e.g., NIFEREX™), iron dextran injection (e.g., INFED™), cupric sulfate, pyroxidine, riboflavin, Vitamin Bn, cyancobalamin injection (e.g., REDISOL™, RUBRAMIN PC™), hydroxocobalamin, folic acid (e.g., FOLVITE™), leucovorin (folinic acid, 5-CHOH4PleG!u, citrovorum factor) or WELLCOVORIN (Calcium salt of leucovorin), transfer™ or ferritin.
10515] 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, molindonc, olanzapine, perphenazine, pimozide, quetiapine, rispcridone, thioridazine, thiothixene, trifluoperazine, and triflupromazine), antimanic agents (e.g., carbamazepine, divalproex sodium, lithium carbonate, and lithium citrate), antidepressants (e.g., arnitriptyline, amoxapine, bupropion, citalopram, clomiprarnine, desipramine, doxepin, fluvoxamine, fluoxetine, imipramine, isocarboxazid, maprotiline, mirtazapine, nefazodone, nortriptyline, paroxetine, phenelzine, protriptyline, sertraline, tranylcypromine, trazodone, trimipramine, and vcnlafaxinc), antianxiety agents (e.g., alprazolam, buspirone, chlordiazepoxide, clorazepate, diazepam, halazepam, lorazepam, oxazepam, and prazepam), and stimulants (e.g., d-amphetamine, methylphenidate, and pemoline).
|0516] Iii 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, phenoharhital, phenytoin, primidone, valprotc acid, divalproex sodium, felbamate, gabapentin, lamotrigine, levetiracetam, oxcarbazepine, tiagabme, topiramate, zoni.samide, diazepam, lora/.eparn, and clonazepam), antiparkinsonian agents (e.g., levodopa/carbidopa, selegiline, amantidine, bromocriptinc, pcrgolidc, ropinirolc, pramipcxole, bcnztropine; biperiden; ethopropazine; procyclidine; trihexyphenidyl, tolcapone), and ALS therapeutics (e.g. riluzole). |0517| 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 pnlynuclcotides of the invention include, but are not limited to, Angiotensin Converting Enzyme (ACE) inhibitors (e.g., papaverine, isoxsuprine, benazepril, captopril, cilazapril, enalapnl, enalaprilat, fosinopril, lisinopril, moexipril, pcrindopril, 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, diltiazcm, felodipine, flunarizine, isradipine, nicardipine, nifedipine, nimodipine, and verapamil.
(0518) 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, Hj histarnme receptor antagonists (e.g., TAGAMET™ (cimetidine), ZANTAC™ (ranitidinc), PEPCID™ (famotidine), and AXID™ (nizatidinc)); inhibitors of H\ K" ATPase (e.g., PREVACID™ (lansoprazole) and PRILOSEC™ (omeprazole)); Dismuth compounds (e.g., PEPTO-B1SMOL™ (bismuth subsalicylate) and DE-NOL™ (bismuth subcitrate)); various antacids; sucraltate; prostaglandin analogs (e.g. CYTOTEC™ (misoprostol)); muscarinic cholinergic antagonists; laxatives (e.g., surfactant laxatives, stimulant laxatives, saline and osmotic laxatives); antidiarrheal agents (e.g., LOMOT1L™ (diphenoxylate), MOTOFEN™ (diphcnoxin), and 1MODIUM™ (loperarnide hydrochloride)), synthetic analogs of somatostatin such as SANDOSTATIN™ (octreotide), antiemetic agents (e.g., ZOI-'RAN™ (ondansetron), KYTRIL™ (granisetron hydrochloride), tropisetron, dolasetron, metoclopramide, chlorpromazine, perphenazine, prochlorperazine, promethazinc, thiethylperazine, triflupromazine, dompcridone, haloperidol, droperidol, trimethobenzamide, dcxamcthasonc, mcthylpredmsolone, 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. |0519] 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.
[0520] 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
|05211 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, arc expressed efficiently in cells which have taken up viral vector nucleic acid.
|0522| Retrovirus vectors and adcno-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) Shod 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, P.M. et ai, (eds.) Greene Publishing Associates, (1989), Sections 9.10-9.14 and other standard laboratory manuals. [0523) 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 ai. BioTechniques 6:616 (1988); Rosenfeld et ai, Science 252:431-434 (1991); and Roscnfeld el ai. Cull 68:143-155 (1992). Suitable adenoviral vectors derived from the adenovirus strain Ad type 5 d!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 ai, (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 inscrtional 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 ai, cited supra; Haj-Ahmand et ai, J. Virol, 57:267 (1986)).
|0524| In another embodiment, non-viral gene delivery systems of the present invention rely on endocytic pathways for the uptake of the subject nuclcotidc 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., lipofcctins) and (optionally) which are tagged with antibodies against cell surface antigens of the target tissue (Mizuno et ai (1992) No Shinkei Geka 20:547-5 5 1; PCT publication W091/06309; Japanese patent application 1047381; and European patent publication EP-A^t3075).
[0525| 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 locali/.ed. 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
[0526] 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, which is herein incorporated by reference.
[0527] 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)), which are herein incorporated by reference. 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.
[0528] 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.
[0529] 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, which are herein incorporated by reference.
[0530] The polynucleotide vector constructs used in the gene therapy method are preferably constructs that will not integrate into the host
genome nor will they contain sequences that allow for replication. Appropriate vectors include pWLNEO, pSV2CAT, pOG44, pXTl and pSG
available from Stratagene; pSVK.3, pBPV, pMSG and pSVL available from Pharmacia; and pEFl/V5, pcDNA3.1, and pRc/CMV2 available from
Invitrogen. Other suitable vectors will be readily apparent to the skilled artisan.
(05311 Any strong promoter known to those skilled in the art can be used for driving the expression of the polynucleotide sequence. Suitable
promoters include adcnoviral 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 ApoAl 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.
[l)532| 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.
|0533] 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,
mucopolysacchande matrix among the reticular fibers of organ tissues, clastic fibers in the walls of vessels or chambers, collagen fibers of fibrous
tissues, or that same matrix within connective tissue enshcathing 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.
|0534] 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.
[0535] 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 Ihc
procedure.
|0536| 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.
[0537] 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.
|OS38| 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, which is herein incorporated by reference); mRNA (Malone et al., Proc. Natl. Acad. Sci. USA (1989) 86:6077-6081, which is herein incorporated by reference); and purified transcription factors (Debs et al., J. Biol. Chem. (1990) 265:10189-10192, which is herein incorporated by reference), in functional form.
[0539] Cationic liposomes are readily available. For example, N[l-2,3-dioleyloxy)propyl]-N,N,N-triethylammonium (DOTMA) liposomes are particularly useful and are available under the trademark Lipofectin, from GIBCO BRL, Grand Island, N.Y. (See, also, Feigner et al., Proc. Natl Acad. Sci. USA (1987) 84:7413-7416, which is herein incorporated by reference). Other commercially available liposomes include transfectace (DDAB/DOPE) and DOTAP/DOPE (Boehrmger).
|0540] Other cationic liposomes can be prepared from readily available materials using techniques well known in the art. See, e.g. PCT Publication No. WO 90/11092 (which is herein incorporated by reference) for a description of the synthesis of DOTAP (l,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, which is herein incorporated by reference. Similar methods can be used to prepare liposomes from other cationic lipid materials.
|OS41| 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.
|0542| 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. 'ITius, 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.
|0543| 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., Straubingcr et al., Methods of Immunology (1983), 101:512-527, which is herein incorporated by reference. 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 he entrapped is added to a suspension of preformed MLVs and then sonicated. When using liposomes containing cationic lipids, the dried lipid film is rcsuspcnded in an appropriate solution such as sterile water or an isotonic buffer solution such as 10 mM Tris/NaCI, 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., Ceil 17:77 (1979)); ether injection (Dearner, D. and Bangham, A., Biochim. Biophys. Acta 443:629 (1976); Oslro 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-Riddcr et al., Science 215:166 (1982)), which are herein incorporated by reference.
[0544J 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.
|0545| U.S. Patent No. 5,b/b,y54 (which is herein incorporated by reference) reports on the injection of genetic material, completed with catiomc 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 (which are herein incorporated by reference) 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.
|OS46| 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 arc not limited to, Moloney Murine Leukemia Virus, spleen necrosis virus, Rous sarcoma Vims, Harvey Sarcoma Virus, avian leukosis virus, gibbon ape leukemia virus, human immunodeficiency virus, Myeloproliferative Sarcoma Virus, and mammary tumor virus.
|0547| The retroviral plastnid vector is employed to transduce packaging cell lines to form producer cell lines. Examples of packaging cells which maybe 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+envAml2, and DAN cell lines as described in Miller, Human Gene Therapy 1:5-14 (1990), which is incorporated herein by reference in its entirety. 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 CaPO« precipitation. In one alternative, the retroviral plasmid vector may be encapsulated into a hposome, or coupled to a lipid, and then administered to a host.
(0548] The producer cell line generates infectious retroviral vector particles which include polynuclcotide 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 eukaryotic cells will express a fusion protin of the present invention.
|0549] 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. Aricnovinis expression is achieved without integration of the viral DNA into the host cell chromosome, thereby alleviating concerns about insertional mutagenesis. Furthermore, adenoviruscs 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).
|0550| 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 etal., Cell 68:143-155 (1992); Engclhardt et al., Human Genet. Ther. 4:759-769 (1993); Yang ct al., Nature Genet. 7:362-369 (1994); Wilson etal., Nature 365:691-692 (1993); and U.S. Patent No. 5,652,224, which are herein incorporated by reference. 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) arc also useful in the present invention.
|0551| 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, Elb, E3, E4, E2a, or LI through L5.
[0552] In certain other embodiments, the cells arc 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. |0553| 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 etal., Molecular Cloning: A Laboratory Manual, Cold Spring Harbor Press (1989). The rccombinant 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, elc. 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 arc 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.
[0554| Another method of gene therapy involves operably associating hetcrologous control regions and endogenous polynuclcotide 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;
Keller et a!., 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.
[0555] 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.
[0556) 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.
|0557| 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.
|0558| 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.
[0559) 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.
[0560] 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)).
[0561| 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.
[0562) 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.
|05631 Therapeutic compositions useful in systemic administration, include fusion proteins of the present invention cornplexed 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.
[0564| 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, which is incorporated herein by reference). 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.
|0565| 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 polynudeoiide constructs dministered per aose, as wcu a.s me 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.
[05661 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
|U567| 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.
10568) In preferred embodiments, the present invention encompasses a method of treating a disease or disorder listed in the "Preferred Indication Y" column of Table I 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.
[0569| 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 I 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 foi which the indications in the Examples are related in an amount effective to treat, prevent or ameliorate the disease or disorder. [0570J 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 arc 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 lndication:Y" column of Table 1.
[0571] In preferred embodiments, fusion proteins of the present invention maybe 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 (sec, 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). |0572] 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.
[0573) 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.
[0574) 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
[0575) 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 (chemolaxis) of immune cells. Immune cells develop through a process called
hcmatopoiesis, producing mycloid (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.
[0576] 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 inhibitor enhance an immune response generated by cells associated with the
tissue(s) in which the polypeptide of the invention is expressed.
[0577] 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-linkcd 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, dysgammaglobulincmia, 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), tg 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.
[0578] 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.
[0579] 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
SCID, adenosine deaminase deficiency, purine nucleoside phosphorylase (PNP) deficiency, Class 11 MHC deficiency (Bare lymphocyte syndrome),
Wiskott-Aldrich syndrome, and ataxia telangiectasia), thymic hypoplasia, third and fourth pharyngeal pouch syndrome, 22qll,2 deletion, chronic
mucocutaneous candidiasis, natural killer cell deficiency (NIC), idiopathic CD4+ T-lymphocytopenia, immunodeficiency with predominant T cell
defect (unspecified), and unspecified immunodeficiency of cell mediated immunity.
|0580| 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.
[0581] 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, myclopcroxidase 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 andVor Cy deficiencies), reticular
dysgenesis, thymic alymphoplasia-aplasia, immunodeficiency with thymoma, severe congenital leukopenia, dysplasia with immunodeficiency,
neonatal ncutropcnia, short limbed dwarfism, and Nezelof syndrome-combined immunodeficiency with Igs.
|0582| 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 andVor polynucleotides encoding albumin fusion proteins of the
invention
[0583] 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 immunorespnnsivencss
among B cell and/or T cell immunodeiicient individuals.
[0584] 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 chcrnotaxis of T-cells, may be an effective therapy in preventing
autoimmune disorders.
[0585) Autoimmune diseases or disorders that may be treated, prevented, diagnosed and/or prognosed by fusion proleins 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
erythcniatosus, rheumatoid arthritis, ankylosing spondylitis, multiple sclerosis, autoimmune thyroiditis, Hashimoto's thyroiditis, autoimmune
hemolytic anemia, hemolytic anemia, thrombocytopenia, autoimmune thrombocytopenia purpura, autoimmune neonatal thrombocytopcnia,
idiopathic thrombocytopenia purpura, purpura (e.g., Henloch-Scoenlein purpura), autoimmunocytopcnia, Goodpasture's syndrome, Pemphigus
vulgaris, myasthenia gravis, Grave's disease (hyperthyroidism), and insulin-resistant diabetes mellitus.
[0586) 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, ncuntis, uveins opninaiima, inilyendocrinopathies, Relief's Disease, Stiff-Man Syndrome, autoimmune pulmonary inflammation, autism, Guillain-Barre Syndrome, insulin dependent diabetes mellitus, and autoimmune inflammatory eye disorders.
10587] 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 extractablc 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 antispemialozoal antibodies), glomerulonephritis (often characterized, e.g., by glomerular basement membrane antibodies or immune complexes), bullous pcmphiguid (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).
|()588| 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 characteri'zed, 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 rnyocardial 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. |0589| 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.
|0590) 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.
(05911 'n 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.
|0592| 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.
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).
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.
[0595| 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, hypcrsensitivity to an anligenic molecule, or blood group incompatibility.
[0596] 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-mediatcd 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 vnro or in vivo.
|0597| 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 polynuclcotidcs 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-rcperfusion injury, endotoxin lethality, complement-mediated hyperacute rejection, nephritis, cytokine or chemokine induced lung injury, inflammatory bowel disease, Crohn's disease, over production of cytokincs (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).
|0598| 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, angiocholccystitis, 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, mctritis, mucitis, myocarditis, myosititis, myringitis, nephritis, neuritis, orchitis, osteochondritis, otitis, pericarditis, peritcndonitis, peritonitis, pharyngitis, phlebitis, poliomyelitis, prostatitis, pulpitis, retinitis, rhinitis, salpingitis, scleritis, sclcrochoroiditis, scrotitis, sinusitis, spondylitis, steatitis, stomatitis, synovitis, syringitis, tendonitis, tonsillitis, urethritis, and vaginitis.
|0599| 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. I'olypeptides, 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.
[0600| 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.
(0601) 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.
[0602] 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.
|0603| 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, Junm, UnKungunya, Rift Valley Fever, herpes simplex, and yellow fever.
[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-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.
|0605| 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: Yibrio cholerae. Mycobacterium leprae. Salmonella typhi. Salmonella paratyphi, Meisseria meningilidis. Streptococcus pneumoniae, Group B streptococcus, Shigella spp., Enterotoxigenic Escherichia coti, Etiterohernorrhagic E. coli, and liorre/ia burgdorferi.
[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 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.
[0607] 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.
|0608| In another specific embodiment, albumin fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention arc used as an antigen for the generation of antibodies to inhibit or enhance immune mediated responses against polypeptides of the invention.
[0609| 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.
|OfilO| 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.
|06111 ln 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.
[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 that elevates the immune status of an individual prior to their receipt of immunosuppressive therapies. [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 induce higher affinity antibodies.
10614] 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.
[061S| 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 immunocompromiscd individuals.
[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 aged populations and/or neonates.
|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 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.
[0618] 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 polynuclcotides encoding aiDumin fusion proteins of the invention, include, but are not limited to, HIV Infection, AIDS, bone marrow transplant, and D cell chronic lyrnphocytic leukemia (CLL).
[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 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.
[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 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 maybe useful as an anti-tumor treatment or to modulate the immune system.
106211 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 THl cellular response.
[0622] In another specific embodiment, albumin fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention arc 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.
(0623| In another specific embodiment, albumin fusion proteins ofthe 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 Immimodificiency.
[0624] In another specific embodiment, albumin fusion proteins ofthe invention and/or polynucleotides encoding albumin fusion proteins ofthe 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 ofthe invention and/or polynuclcotides encoding albumin fusion proteins ofthe invention are used in the pretreatment of bone marrow samples prior to transplant.
|0625] In another specific embodiment, albumin fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins ofthe invention are used as a gene-based therapy for genetically inherited disorders resulting in immuno-incompetence/immunodeficiency such as observed among SCtD patients.
|0626| In another specific embodiment, albumin fusion proteins ofthe invention and/or polynucleotides encoding albumin fusion proteins ofthe invention are used as a means of activating monocytes/macrophages to defend against parasitic diseases that effect monocytes such as Leishmania. 10627| In another specific embodiment, albumin fusion proteins ofthe invention and/or polynucleotides encoding albumin fusion proteins ofthe invention are used as a means of regulating secreted cytokines that are elicited by polypeptidcs ofthe invention.
|0628| 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 dccribed herein, as they may apply to veterinary medicine.
|062'J| In another specific embodiment, albumin fusion proteins ofthe invention and/or polynucleotides encoding albumin fusion proteins ofthe 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. [0630] In another specific embodiment, albumin fusion proteins ofthe invention and/or polynucleotides encoding albumin fusion proteins ofthe invention are used as a therapy for preventing the 8 cell proliferation and Ig secretion associated with autoimmune diseases such as idiopathic thrornbocytopcnic purpura, systemic lupus erythematosus and multiple sclerosis.
[0631] In another specific embodiment, polypeptides, antibodies, polynuclcotides and/or agonists or antagonists ofthe present fusion proteins of the invention and/or polynucleotidcs encoding albumin fusion proteins ofthe 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.
[0632) In another specific cmbodimenl, albumin fusion proteins ofthe invention and/or polynucleotides encoding albumin fusion proteins ofthe invemion 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.
|0633| In another specific embodiment, albumin fusion proteins ofthe invention and/or polynucleotides encoding albumin fusion proteins ofthe
invention may be employed tor 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 CDS 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.
(0634) 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.
|063S1 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.
106361 In another specific embodiment, albumin fusion proteins of the invention anoVor polynucleotides encoding albumin fusion proteins of
the invention are used to enhance or inhibit antibody dependent cellular cytotoxicity.
[06371 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.
[0638| 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).
[0639] 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
he used to stimulate the regeneration of mucosal surfaces.
[0640] 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
irnmunoglobulin 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 hcrnoglobinuria.
[06411 'n 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 hypogammaglobulincmia") or a subset of this disease.
[0642] 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 myclogcnous leukemia, chronic
myelogcnous leukemia, llodgkm's disease, non-Hodgkjn'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
"I lyperproliferative Disorders" elsewhere herein.
|0643] 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.
|06441 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.
|0645| In specific embodiments, the compositions of the invention are used as an agent to boost immunoresponsiveness among B cell
immunodcficient individuals, such as, for example, an individual who has undergone a partial or complete splenectomy.
Blood-Related Disorders
[0646] The albumin fusion proteins of the invention anoVor 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 aciivity, 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 polynucleotiQes encoaing 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
[0647| 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).
|0648| 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.
|0649| The fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention maybe 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 maybe useful in the prevention, detection, diagnosis and/or treatment of leukocytoses, such as, for example eosinophilia. |0650| "I'he 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.
|0651| 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 (hcmolysis). 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, gIucose-6-phosphatc dchydrogenasc deficiency, and sickle cell anemia.
|()652| 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.
|0653| 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 (eg, idiopathic thrombocytopenic purpura, and thrombotic thrombocytopenic purpura), Von Willebrand's disease, hereditary platelet disorders (e.g., storage pool disease such as Chediak-Higashi and Hennansky-Pudlak syndromes, thromboxane A2 dysfunction, thromboasthenia, and Bemard-Soulier syndrome), hemolytic-uremic syndrome, hemophelias such as hcmophelia A or Factor VII deficiency and Christmas disease or
l-actor IX deficiency, Hereditary iiemorhhagic Telangiectsia, also known as Rendu-Oslcr-Weber syndrome, allergic purpura (Henoch Schonlein purpura) and disseminated intravascular coagulation.
|0654] The effect of the albumin fusion proteins of the invention and/or polynucleotidcs 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.
|0655| 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.
|0656| 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 leukocylosis. 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.
[0657] Leukopenia may be a generalized decreased in all types of white blood cells, or may be a specific depiction 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 polynucleotidcs encoding albumin fusion proteins of the invention include, but arc not limited to, infantile genetic agranulocytosis, familial neutropenia, cyclic neutropenia, ncutropenias 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., Helty syndrome, malaria and sarcoidosis), and some drug treatment regimens.
(0658| 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 lymphocytopcnias (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., DiGcorge syndrome, Wiskott-Aldrich Syndome, severe combined immunodeficiency, ataxia tclangiectsia).
[0659] 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 macrophagc function including, but not limited to, Gaucher's disease, Niemann-l'ick disease, Letterer-Siwe disease and Hand-Schuller-Christian disease. |0660] 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 cosinophil function including, but not limited to, idiopathic hypereosinophilic syndrome, eosinophilia-myalgia syndrome, and Hand-Schullcr-Christian disease.
[0661] 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 Icukemias 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 Icukemias, 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. [0662] 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, monociunai gammaopathies, monoclonal gammopathies of undetermined significance, multiple myeloma,
macroglobulinemia, Waldenstrom's macroglobulinemia, cryoglobulinemia, and Raynaud's phenomenon.
|0663] 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 scconday thrombocythemia) and chronic myelocytic leukemia.
[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 a treatment prior to surgery, to increase blood cell production.
[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 enhance the migration, phagocytosis, superoxide production, antibody dependent cellular cytotoxicity of
neutrophils, eosionophils and macrophages.
(0666] 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 maybe useful as an agent to
increase the number of stem cells in circulation prior to platelet pheresis.
[0667] 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.
|0668| 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
|0669] 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.
[0670] For example, by increasing an immune response, particularly increasing antigenic qualities of the hyperproliferative disorder or by proliferating, differentiating, or mobilizing T-cclls, hyperprolifcrative 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.
|0671] 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.
[0672] 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 Lyinphocytic Leukemia, Adult Acute Myeloid Leukemia, Adult Hodgkin's Disease, Adult Hodgkin's Lymphoma, Adult Lyniphocylic Leukemia, Adult Non-Hodgkin's Lymphoma, Adult Primary Liver Cancer, Adult Soft Tissue Sarcoma, AIDS-Rclated Lymphoma, AIDS-Rclated Malignancies, Anal Cancer, Astrocytoma, Bile Duct Cancer, Bladder Cancer, Bone Cancer, Brain Stem Gliorna, 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 Aslrocytoma, 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 Ncuroectodermal 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, Swing'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, Gallbladdei Cancer, Gastric Cancer, Gastrointestinal Carcinoid Tumor, Gastrointestinal Tumors, Germ Cell Tumors, Gestational Trophohlastic Tumor, Hairy Cell Leukemia, Head and Neck Cancer, Hepatocellular Cancer, Hodgkin's Disease, Hodgkin's
Lyiiiphoma, Hypcrgammaglobulmemia, Hypopharyngeal Cancer, Intestinal Cancers, Intraocular Melanoma, Islet Cell Carcinoma, Islet Cell Pancreatic Cancer, Kaposi's Sarcoma, Kidney Cancer, Laryngca] Cancer, Lip and Oral Cavity Cancer, Liver Cancer, Lung Cancer, I vrnphnproliferativc 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 I.yrnphoma During Pregnancy, Nonmclanoma 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 Ilistiocytoma of Bone, Ovarian Epithelial Cancer, Ovarian Germ Cell Tumor, Ovarian Low Malignant Potential Tumor, Pancreatic Cancer, Paraproteinemias, Purpura, Parathyroid Cancer, Penile Cancer, Phcochrornocytoma, 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, Urcthral 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.
(0673) 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-ncoplastic 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 fid., W. B. Saunders Co., Philadelphia, pp. 68-79.)
|0674| 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. Hypcrplastic disorders which can be diagnosed, prognosed, prevented, and7or 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 rnelanocytic 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, pseudoepitheliomalous hyperplasia, senile sebaceous hyperplasia, and verrucous hyperplasia.
|0675| 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, rnetaplastjc anemia, metaplastic ossification, metaplastic polyps, myeloid metaplasia, primary myeloid metaplasia, secondary myeloid metaplasia, squamous metaplasia, squamous metaplasia of amnion, and symptomatic myeloid metaplasia.
|0676| Dysplasia is frequently a forerunner of cancer, and is found mainly in the epithelia; it is the most disorderly form of non-ncoplastic 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 anoVor polynucleotides encoding albumin fusion proteins of the invention include, but are not limited to, anhidrotic ectodermal dysplasia, antcrofacial dysplasia, asphyxiating thoracic dysplasia, atriodigital dysplasia, bronchopulmonary dysplasia, cerebral dysplasia, cervical dysplasia, chondroectodermal dysplasia, clcidocranial 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 cpiphysialis 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 eclodermal dysplasia, hypohidrotic ectodemial dysplasia, lymphopenic thymic dysplasia, mammary dysplasia, mandibulofacial dysplasia, rnetaphysial dysplasia, Mondini dysplasia, monostotic fibrous dysplasia, mucoepithelial dysplasia, multiple epiphysial dysplasia, oculoauriculovertebral dysplasia, oculodentodigital dysplasia, oculovertebral dysplasia, odontogcnic dysplasia, ophthalmomandibulomelic dysplasia,
penapical cemcntal dysplasia, poiyusiotic fibrous dysplasia, pseudoachondroplastic spondyloepiphysial dyspiasia, retinal dysplasia, septo-optic dysplasia, spondyloepiphysial dysplasia, and ventriculoradial dysplasia.
[0677J Additional pre-neoplastic disorders which can be diagnosed, prognoscd, prevented, and/or treated with fusion proteins of the invention and/or polynucleotides encoding a.bumin 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, keraloses, Bo\vcn's disease, Farmer's Skin, solar cheilitis, and solar keratosis.
|067fi| 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. [0679| 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. [0680| Additionally, fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention may affect apoplosis, 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, retmoblastoma, 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, Sjogrcn'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 adenovtruses), inflammation, graft v. host disease, acute graft rejection, and chronic graft rejection. [0681] 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.
[0681| 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 metaslases of malignancies and related disorders such as leukemia (including acute leukemias (e.g., acute lyrnphocytic leukemia, acute myelocytic leukemia (including myeloblastic, promyelocytic, myelomonocytic, monocytic, and crythroleukemia)) and chronic leukemias (e.g., chronic myelocytic (granulocytic) leukemia and chronic lyrnphocytic 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, osteogcnic sarcoma, chordoma, angidsarcoma, cndotheliosarcoma, lymphangiosarcoma, lympnangioendotncliasaneoma, 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, cystadcnocarcinoma, medullary carcinoma, bronchogcnic carcinoma, renal cell carcinoma, hepatoma, bile duct carcinoma, choriocarcinoma, semmoma, embryonal carcinoma, Wilrn's tumor, cervical cancer, testicular tumor, lung carcinoma, small cell lung carcinoma, bladder carcinoma, epithelial carcinoma, gliorna, astrocytoma, medulloblastuma, craniopharyngioma, ependymoma, pinealoma, emangioblastoma, acoustic neuroma, oligodendroglioma, menangioma, melanoma, neuroblasloma, and rctinoblastoma.
|0683| 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, rctmitis pigmentosa, cerebellar degeneration and brain tumor or prior associated disease); autoimmune disorders (such as, multiple sclerosis, Sjogren's syndrome, Hashimoto's Ihyroiditis, biliary cirrhosis, Bchcet'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 reperfjsion injury), liver injury (e.g., hepatitis related liver injury, ischemia/repcrfusion injury, cholestosis (bile duct injury) and liver cancer); toxin-induced liver disease (such as that caused by alcohol), septic shock, cachexia and anorexia.
[0684| Hyperproliferative diseases and/or disorders that could be diagnosed, prognosed, prevented, and/or treated by fusion proteins of the invention and/or polynudeotides encoding albumin fusion proteins of the invention, include, but are not limited to, neoplasms located in the liver, abdomen, bone, breast, digeslive 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 urogcnital tract. |06SS] Similarly, olher 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: hypergammaglobulinemia, lymphoproliferative disorders, paraproteinemias, purpura, sarcoidosis, Sezary Syndrome, Waldenstron's macroplobulinemia, Gaucher's Disease, histiocytosis, and any other hyperproliferative disease, besides neoplasia, located in an organ system listed above.
(0686) 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.
|0687| 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. 10688) 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 rccombinant 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 retrovims, or more preferably an adcnoviral 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,
[0689] 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.
|0690) 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 including, but not limited to transfefction, elcctroporation, microinjection of cells, or in vehicles such as liposomes, lipofectin, or as naked polynucleotides, or any other method described throughout the specification. The polynucleotide of the present invention may be delivered by known gene delivery systems such as, but not limited to, retroviral vectors (Gilboa, J. Virology 44:845 (1982); Hocke, Nature 320:275 (1986); Wilson, et al., Proc. Natl. Acad. Sci. U.S.A. 85:3014), vaccinia virus system (Chakrabarty et al., Mol. Cell Biol. 5:3403 (1985) or other efficient DNA delivery systems (Yates et al.. Nature 313:812 (1985)) known to those skilled in the art. These references are exemplary only and are hereby incorporated by reterencc. In order to specifically deliver or transfect cells which are abnormally proliferating and spare non-dividing cells, it is preferable to utilize a retrovirus, or adenoviral (as described in the art and elsewhere herein) delivery system known to those of skill in the art. Since host DNA replication is required for retroviral DNA to integrate and the retrovirus will be unable to self replicate due to the lack of the retrovirus genes needed for its life cycle. Utilizing such a retroviral delivery system for polynucleotides of the present invention will target said gene and constructs to abnormally proliferating cells and will spare the non-dividing normal cells.
[06911 The polynucleotides of the present invention may be delivered directly to cell proliferative disorder/disease sites in internal organs, body cavities and the like by use of imaging devices used to guide an injecting needle directly to the disease site. The polynucleotides of the present invention may also be administered to disease sites at the time of surgical intervention.
|0692] By "cell proliferative disease" is meant any human or animal disease or disorder, affecting any one or any combination of organs, cavities, or body parts, which is characterized by single or multiple local abnormal proliferations of cells, groups of cells, or tissues, whether benign or malignant.
|0693| Any amount of the polynucleotides of the present invention may be administered as long as it has a biologically inhibiting effect on the proliferation of the treated cells. Moreover, it is possible to administer more than one of the polynucleotide of the present invention simultaneously to the same site. By "biologically inhibiting" is meant partial or total growth inhibition as well as decreases in the rate of proliferation or growth of the cells. The biologically inhibitory dose may be determined by assessing the effects of the polynucleotides of the present invention on target malignant or abnormally proliferating cell growth in tissue culture, tumor growth in animals and cell cultures, or any other method known to one of ordinary skill in the art.
[0694] Moreover, fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention of the present invention are useful in inhibiting the angiogcnesis of proliferative cells or tissues, either alone, as a protein fusion, or in combination with other polypcptides directly or indirectly, as described elsewhere herein. In a most preferred embodiment, said anti-angiogenesis effect may be achieved indirectly, for example, through the inhibition of hernatopoietic, tumor-specific cells, such as tumor-associated macrophages (See Joseph IB, et al. J Natl Cancer Inst, 90(21):l648-53 (1998), which is hereby incorporated by reference).
[0695] Albumin fusion proteins ol the invention and/or polynucleotides encoding albumin fusion proteins of the invention may be useful in inhibiting proliferativc cells or tissues through the induction of apoptosis. These fusion protieins and/or polynucleotides may act either directly, or indirectly to induce apoptosis of proliferative cells and tissues, for example in the activation of a death-domain receptor, such as tumor necrosis factor (TNF) receptor-1, CD95 (Fa.s/APO-1), TNF-rcceptor-related apoptosis-mediated protein (TRAMP) and TNF-related apoptosis-inducing hgand (TRAIL) receptor-1 and -2 (See Schulze-Osthoff K, et.al., Eur J Biochem 254(3):439-59 (1998), which is hereby incorporated by reference). Moreover, in another preferred embodiment of the present invention, these fusion proteins and/or polynucleotides may induce apoptosis through other mechanisms, such as in the activation of other proteins which will activate apoptosis, or through stimulating the expression of these proteins, either alone or in combination with small molecule drugs or adjuviants, such as apoptonin, galectins, thioredoxins, anti-inflammatory proteins (See for example, Mutat Res 400(1-2):447-55 (1998), Med Hypotheses.50(5):423-33 (1998), Chem Biol Interact. Apr 24; 111-112:23-34 (1998), J Mol Med.76(6):402-12 (1998), Int J Tissue React;20(l):3-15 (1998), which are all hereby incorporated by reference).
|0696| Albumin fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention arc useful in inhibiting the metastasis of proliferative cells or tissues. Inhibition may occur as a direct result of administering these albumin fusion proteins and/or polynucleotides, or indirectly, such as activating the expression of proteins known to inhibit metastasis, for example alpha 4 integrins, (See, e.g., Curr Top Microbiol Immunol 1998;231:125^)1, which is hereby incorporated by reference). Such thereapeutic affects of the present invention may be achieved either alone, or in combination with small molecule drugs or adjuvants.
|0697] In another embodiment, the invention provides a method of delivering compositions containing the albumin fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention to targeted cells expressing the a polypeplide bound by, that binds to, or associates with an albumin fuison protein of the invention. Albumin fusion proteins of the invention may be associated with with hctcrologous polypeptides, heterologous nucleic acids, toxins, or prodrugs via hydrophobia, hydrophilic, ionic and/or covalent interactions. [0698] Albumin fusion proteins of the invention are useful in enhancing the immunogenicity and/or antigenicity of proliferating cells or tissues, either directly, such as would occur if the albumin fusion proteins of the invention 'vaccinated' the immune response to respond to proliferative antigens and irnmunogens, or indirectly, such as in activating the expression of proteins known to enhance the immune response (e.g. chemokines), to said antigens and irnmunogens.
Renal Disorders
|0699] Albumin 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 disorders of the renal system. Renal disorders which can be diagnosed, prognosed, prevented, and/or treated with compositions of the invention include, but are not limited to, kidney failure, nephritis, blood vessel disorders of kidney, metabolic and congenital kidney disorders, urinary disorders of the kidney, autoimmune disorders, sclerosis and necrosis, electrolyte imbalance, and kidney cancers.
[0700| Kidney diseases which can he diagnosed, prognosed, prevented, and/or treated with compositions of the invention include, but are not limited to, acute kidney failure, chronic kidney failure, athcrocmbolic renal failure, end-stage renal disease, inflammatory diseases of the kidney (e g , acute glomerulonephritis, postinfectious glomerulonephritis, rapidly progressive glomerulonephritis, nephrotic syndrome, membranous glonierulonephntis, familial nephrotic syndrome, membranoproliferative glomerulonephritis I and II, mesangial proliferative glomerulonephritis, chronic glomerulonephritis, acute tubulointerstilial nephritis, chronic tubulointerstitial nephritis, acute post-streptococcal glomerulonephritis (PSGN), pyelonephritis, lupus nephritis, chronic nephritis, interstitial nephritis, and post-strcptococcal glomerulonephritis), blood vessel disorders of the kidneys (e.g., kidney infarction, athcrocmbolic kidney disease, cortical necrosis, malignant nephrosclerosis, renal vein thrombosis, renal underperfusion, renal retinopathy, renal ischemia-reperfusion, renal artery embolism, and renal artery stenosis), and kidney disorders resulting form urinary tract disease (e.g., pyelonephritis, hydroncphrosis, urolithiasis (renal lithiasis, nephrolithiasis), reflux nephropathy, urinary tract infections, urinary retention, and acute or chronic unilateral obstructive uropathy.)
|0701| In addition, compositions of the invention can be used to diagnose, prognose, prevent, and/or treat metabolic and congenital disorders of ihe kidney (e.g., uremia, renal amyloidosis, renal osteodystrophy, renal tubular acidosis, renal glycosuria, nephrogenic diabetes insipidus, cystinuria, Fanconi's syndrome, renal fibrocystic osteosis (renal rickets), Hartnup disease, Banter's syndrome, Liddle's syndrome, polycystic kidney disease, medullary cystic disease, medullary sponge kidney, Alport's syndrome, nail-patella syndrome, congenital nephrotic syndrome, CRUSH syndrome, horseshoe kidney, diabetic nephropathy, nephrogenic diabetes insipidus, analgesic nephropathy, kidney stones, and membranous nephropathy), and autoimmune disorders of the kidney (e.g., systemic lupus erythemalosus (SUE), Goodpasture syndrome, IgA nephropathy, and IgM mesangial proliferalive glomerulonephritis).
[0702] Compositions of the invention can also be used to diagnose, prognose, prevent, and/or treat sclerotic or necrotic disorders of the kidney (eg., glomerulosclcrosis, diabetic nephropathy, focal segmental glomcrulosclerosis (FSGS), nccrotizing glomerulonephritis, and renal papillary necrosis), cancers of ihe kidney (e.g., nephroma, hypernephroma, ncphroblastoma, renal cell cancer, transitional cell cancer, renal adenocarcinoma, squamous cell cancer, and Wilm's tumor), and electrolyte imbalances (e.g., nephrocalcinosis, pyuria, edema, hydronephritis, proleinuria, hyponatremia, hypematremia, hypokalcmia, hyperkalemia, hypocalccmia, hypercalcemia, hypophosphatemia, and hypcrphosphatcmia).
|U703| Compositions ol me invention may be administered using any method known in the art, including, but not limited to, direct needle injection at the delivery site, intravenous injection, topical administration, catheter infusion, biolistic injectors, particle accelerators, gelfoam sponge depots, other commercially available depot materials, osmotic pumps, oral or suppositorial solid pharmaceutical formulations, decanting or topical applications during surgery, aerosol delivery. Such methods are known in the art. Compositions of the invention may be administered as part of a Therapeutic, described in more detail below. Methods of delivering polynucleotides of the invention are described in more detail herein.
Cardiovascular Disorders
[0704] Albumin 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 cardiovascular disorders, including, but not limited to, peripheral artery disease, such as limb ischemia. |0705| Cardiovascular disorders include, but are not limited to, cardiovascular abnormalities, such as arterio-artcrial fistula, arteriovenous fistula, cerebral arteriovenous malformations, congenital heart defects, pulmonary atresia, and Scimitar Syndrome. Congenital heart defects include, but are not limited to, aortic coarctation, cor triatriatum, coronary vessel anomalies, crisscross heart, dextrocardia, patent ductus arteriosus, Ebstein's anomaly, Eisenmenger complex, hypoplastic left heart syndrome, levocardia, tetralogy of fallot, transposition of great vessels, double outlet right ventricle, tricuspid atresia, persistent truncus arteriosus, and heart septal defects, such as aortopulmonary septa! defect, endocardia! cushion defects, Lutembacher's Syndrome, trilogy of Fallot, ventricular heart septal defects.
|0706] Cardiovascular disorders also include, but are not limited to, heart disease, such as arrhythmias, carcinoid heart disease, high cardiac output, low cardiac output, cardiac tamponade, endocarditis (including bacterial), heart aneurysm, cardiac arrest, congestive heart failure, congestive cardinmyopathy, paroxysmal dyspnea, cardiac edema, heart hypertrophy, congestive cardiomyopathy, left ventricular hypertrophy, right ventricular hypertrophy, post-infarction heart rupture, ventricular septal rupture, heart valve diseases, myocardial diseases, myocardial ischemia, pericardia! effusion, pericarditis (including constrictive and tuberculous), pneumopericardium, postpericardiotomy syndrome, pulmonary heart disease, rheumatic heart disease, ventricular dysfunction, hyperemia, cardiovascular pregnancy complications, Scimitar Syndrome, cardiovascular syphilis, and cardiovascular tuberculosis.
[0707] Arrhythmias include, but are not limited to, sinus arrhythmia, atrial fibrillation, atrial flutter, bradycardia, extrasystole, Adams-Stokes Syndrome, bundle-branch block, sinoatrial block, long QT syndrome, parasystole, Lown-Ganong-Levine Syndrome, Mahaim-type pre-excitation syndrome, Wolff-Parkinson-Whitc syndrome, sick sinus syndrome, tachycardias, and ventricular fibrillation. Tachycardias include paroxysmal tachycardia, supraventricular tachycardia, accelerated idioventricular rhythm, atrioventricular nodal reentry tachycardia, ectopic atrial tachycardia, ectopic junctional tachycardia, sinoatrial nodal reentry tachycardia, sinus tachycardia, Torsades de Pointes, and ventricular tachycardia. |0708| Heart valve diseases include, but are not limited to, aortic valve insufficiency, aortic valve stenosis, hear murmurs, aortic valve prolapse, mitral valve prolapse, tricuspid valve prolapse, mitral valve insufficiency, mitral valve stenosis, pulmonary atresia, pulmonary valve insufficiency, pulmonary valve stenosis, tricuspid atresia, tricuspid valve insufficiency, and tricuspid valve stenosis.
(0709] Myocardial diseases include, but are not limited to. alcoholic cardiomyopathy, congestive cardiomyopathy, hypcrtrophic cardiomyopathy, aortic subvalvular stenosis, pulmonary subvalvular stenosis, restrictive cardiomyopathy, Chagas cardiomyopathy, endocardial fibroelastosis, endotnyocardial fibrosis, Kearns Syndrome, myocardial reperfusion injury, and myocarditis.
|07IO] Myocardial ischemias include, but are not limited to, coronary disease, such as angina pcctoris, coronary aneurysm, coronary arteriosclerosis, coronary thrombosis, coronary vasospasm, myocardial infarction and myocardial stunning.
|07111 Cardiovascular diseases also include vascular diseases such as aneurysms, angiodysplasia, angiomatosis, bacillary angiomatosis, Hippel-Lindau Disease, Klippel-Trcnaunay-Weber Syndrome, Sturge-Weber Syndrome, angioneurotic edema, aortic diseases, Takayasu's Arteritis, aortitis, Lenchc's Syndrome, arterial occlusive diseases, arteritis, enarteritis, polyartentis nodosa, cerebrovascular disorders, diabetic angiopathies, diabetic retinopathy, embolisms, thrombosis, crythromelalgia, hemorrhoids, hepatic veno-occlusive disease, hypertension, hypotension, ischemia, peripheral vascular diseases, phlebitis, pulmonary veno-occlusive disease, Raynatid's disease, CREST syndrome, retinal vein occlusion, Scimitar syndrome, superior vena cava syndrome, telangiectasia, atacia telangiectasia, hereditary hemorrhagic telangiectasia, varicocele, varicose veins, varicose ulcer, vasculitis, and venous insufficiency.
|0712| Aneurysms include, but arc not limited to, dissecting aneurysms, false aneurysms, infected aneurysms, ruptured aneurysms, aortic aneurysms, cerebral aneurysms, coronary aneurysms, heart aneurysms, and iliac aneurysms.
|07I3| Arterial occlusive diseases include, but are not limited to, arteriosclerosis, intermittent claudication, carotid stenosis, fibromuscular dysplasias, mesenteric vascular occlusion, Moyamoya disease, renal artery obstruction, retinal artery occlusion, and thromboangiitis obliterans. |071 |0715| Embolisms include, but are not limited to, air embolisms, amniotic fluid embolisms, cholesterol embolisms, blue toe syndrome, fat
embolisms, pulmonary eniDOlisms, ana thromoboembolisms. Thrombosis include, but are not limited to, coronary thrombosis, hepatic vein
thrombosis, retinal vein occlusion, carotid artery thrombosis, sinus thrombosis, Wallenberg's syndrome, and thrombophlebitis.
[0716| Ischemic disorders include, but are not limited to, cerebral ischemia, ischernic colitis, compartment syndromes, anterior compartment
syndrome, myocardial ischemia, rcperfusion injuries, and peripheral limb ischemia. Vasculitis includes, but is not limited to, aortitis, arteritis,
Behcet's Syndrome, Churg-Strauss Syndrome, mucocutaneous lymph node syndrome, thromboangiitis obliterans, hypersensitivity vasculitis,
Schoenlein-Henoch purpura, allergic cutaneous vasculitis, and Wegener's granulomatosis.
(0717] Albumin fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention may be administered
using any method known in the art, including, but not limited to, direct needle injection at the delivery site, intravenous injection, topical
administration, catheter infusion, biolistic injectors, particle accelerators, gelfoam sponge depots, other commercially available depot materials,
osmotic pumps, oral or suppositorial solid pharmaceutical formulations, decanting or topical applications during surgery, aerosol delivery. Such
methods are known in the art. Methods of delivering polynucleotides are described in more detail herein.
Respiratory Disorders
10718] Albumin 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 diseases and/or disorders of the respiratory system.
|0719| Diseases and disorders of the respiratory system include, but are not limited to, nasal vestibulitis, nonallergic rhinitis (e.g., acute rhinitis, chronic rhinitis, atrophic rhinitis, vasomotor rhinitis), nasal polyps, and sinusitis, juvenile angiofibromas, cancer of the nose and juvenile papillomas, vocal cord polyps, nodules (singer's nodules), contact ulcers, vocal cord paralysis, laryngocclcs, pharyngitis (e.g., viral and bacterial), tonsillitis, tonsillar cellulitis, parapharyngeal abscess, laryngitis, laryngoceles, and throat cancers (e.g., cancer of the nasopharynx, tonsil cancer, larynx cancer), lung cancer (e.g., squamous cell carcinoma, small cell (oat cell) carcinoma, large cell carcinoma, and adenocarcinoma), allergic disorders (eosinophilic pneumonia, hypersensitivity pneumonitis (e.g., extrinsic allergic alveolitis, allergic interstitial pneumonitis, organic dust pneumoconiosis, allergic bronchopulmonary aspcrgillosis, asthma, Wegener's granulomatosis (granulomatous vasculitis), Goodpasture's syndrome)), pneumonia (e.g., bacterial pneumonia (e.g., Streptococcus pneumoniae (pneumoncoccal pneumonia), Staphylococcus aiireus (staphylococcal pneumonia), Gram-negative bacterial pneumonia (caused by, e.g., Klebsiella and Pseudomas spp.), Mycoplasmti pneumoniae pneumonia, Hemophilus influenzae pneumonia, Legionella pneumophUa (Legionnaires' disease), and Chlamydia psittaci (Psittacosis)), and viral pneumonia (e.g., influenza, chickenpox (varicella).
|0720] Additional diseases and disorders of the respiratory system include, but are not limited to bronchiolitis, polio (poliomyelitis), croup, respiratory syncytial viral infection, mumps, erythema infectiosum (fifth disease), roseola infantum, progressive rubella panencephalitis, german measles, and subacutc sclerosing panencephalitis), fungal pneumonia (e.g., Histoplasmosis, Coccidioidomycosis, Blastomycosis, fungal infections in people with severely suppressed immune systems (e.g., cryptococcosis, caused by Cryptococcus neoformans; aspergillosis, caused by Aspergillus spp.; candidiasis, caused by Candida; and mucormycosis)), Pneumocyslis carinii (pneumocystis pneumonia), atypical pneumonias (e.g., Mycoplasma and Chlamydia spp.), opportunistic infection pneumonia, nosocomial pneumonia, chemical pneumonitis, and aspiration pneumonia, pleural disorders (eg., pleurisy, pleural effusion, and pneumothorax (e.g., simple spontaneous pneumothorax, complicated spontaneous pneumothorax, tension pneumothorax)), obstructive airway diseases (e.g., asthma, chronic obstructive pulmonary disease (COPD), emphysema, chronic or acute bronchitis), occupational lung diseases (e.g., silicosis, black lung (coal workers' pneumoconiosis), asbestosis, berylliosis, occupational asthsma, byssinosis, and benign pneumoconioscs), Infiltrative Lung Disease (e.g., pulmonary fibrosis (e.g., fibrosing alveolitis, usual interstitial pneumonia), idiopathic pulmonary fibrosis, desquamative interstitial pneumonia, lymphoid interstitial pneumonia, histiocytosis X (e.g., Lclterer-Siwe disease, Hand-Schuller-Christian disease, eosinophilic granuloma), idiopathic pulmonary hemosiderosis, sarcoidosis and pulmonary alveolar proteinosis), Acute respiratory distress syndrome (also called, e.g., adult respiratory distress syndrome), edema, pulmonary embolism, bronchitis (e.g., viral, bacterial), bronchiectasis, atelectasis, lung abscess (caused by, e.g., Staphylococcus aureus or Legionella pncumophila), and cystic fibrosis.
Anli-Angiogcnesis Aclivity
|0721| The naturally occurring balance between endogenous stimulators and inhibitors of angiogenesis is one in which inhibitory influences predominate. Rastmejad el a!.. Cell 5(5:345-355 (1989). In those rare instances in which neovascularization occurs under normal physiological conditions, such as wound healing, organ regeneration, embryonic development, and female reproductive processes, angiogenesis is stringently regulated and spatially and temporally delimited. Under conditions of pathological angiogenesis such as that characterizing solid tumor growth, these regulatory controls fail. Unregulated angiogenesis becomes pathologic and sustains progression of many neoplastic and non-neoplastic diseases. A number of serious diseases are dominated by abnormal neovascularization including solid tumor growth and mctastases, arthritis, some types of eye disorders, and psoriasis. See, e.g., reviews by Moses el a/.. Biotech. 9:630-634 (1991); Folkman el at., N. Engl. J. Med., 335:1757-1763 (1995); Aucrbach el ai. J. Microvasc. Res. 29:40Ml I (1985); Folkman, Advances in Cancer Research, eds. Klein and Weinhouse, Academic Press, New York, pp. 175-203 (1985); Patz. Am. J. Opthalmol. 94:715-743 (1982); and Folkman et /., Science 221:1 \9-125 (1983). In a number of pathological conditions, the process of angiogenesis contributes to the disease state. For example, significant data have accumulated which suggest
that the growth of solid tumors is dependent on angiogcnesis. Folkman and Klagsbrun, Science 235:442-447 (1987).
|0722| The present invention provides for treatment of diseases or disorders associated with neovascularization by administration of fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention. Malignant and metastatic conditions which can be treated with the polynucleotides and polypeptides, or agonists or antagonists of the invention include, but are not limited to, malignancies, solid tumors, and cancers described herein and otherwise known in the art (for a review of such disorders, see Fishman et ni. Medicine, 2d Ed., J. B. Lippincott Co., Philadelphia (1985)).Thus, the present invention provides a method of treating an angiogenesis-related disease and/or disorder, comprising administering to an individual in need thereof a therapeutically effective amount of an albumin fusion protein of the invention and/or polynucleotides encoding an albumin fusion protein of the invention. For example, fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention may be utilized in a variety of additional methods in order to therapeutically treat a cancer or tumor. Cancers which may be treated with fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention include, but are not limited to solid tumors, including prostate, lung, breast, ovarian, stomach, pancreas, larynx, esophagus, testes, liver, parotid, biliary tract, colon, rectum, cervix, uterus, endometrium, kidney, bladder, thyroid cancer; primary tumors and mctastases; melanomas; gliohlastoma; Kaposi's sarcoma; leiomyosarcoma; non- small cell lung cancer; colorectal cancer; advanced malignancies; and blood born tumors such as leukemias. For example, fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention may be delivered topically, in order to treat cancers such as skin cancer, head and neck tumors, breast tumors, and Kaposi's sarcoma.
[0723] Within yet other aspects, fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention may be utilized to treat superficial forms of bladder cancer by, for example, intravesical administration. Albumin fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention may be delivered directly into the tumor, or near the tumor site, via injection or a catheter. Of course, as the artisan of ordinary skill will appreciate, the appropriate mode of administration will vary according to the cancer to be treated. Other modes of delivery are discussed herein.
[07241 Albumin fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention may be useful in treating other disorders, besides cancers, which involve angiogenesis. These disorders include, but are not limited to: benign tumors, for example hemangiomas, acoustic neuromas, neurofibromas, trachomas, and pyogenic granulomas; artheroscleric plaques; ocular angiogenic diseases, for example, diabetic retinopathy, retinopathy of prematurity, macular degeneration, comeal graft rejection, neovascular glaucoma, retrnlental fibroplasia, rubeosis, retinoblastoma, uvietis and Plerygia (abnormal blood vessel growth) of the eye; rheumatoid arthritis; psoriasis; delayed wound healing, endometriosis; vasculogcnesis; granulations; hypcrtrophic scars (keloids); nonunion fractures; sclcroderma; trachoma; vascular adhesions; myocardial angiogenesis; coronary collaterals; cerebral collaterals; arteriovenous malformations; ischemic limb angiogenesis; Osier-Webber Syndrome; plaque neovascularization; tclangiectasia; hemophiliac joints; angiofibroma; fibromuscular dysplasia; wound granulation; Crohn's disease; and atherosclerosis.
[0725| For example, within one aspect of (he present invention methods are provided for treating hypertrophic scars and keloids, comprising the step of administering albumin fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention to a hypcrtrophic scar or keloid.
|0726| Within one embodiment of the present invention fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention are directly injected into a hypertrophic scar or keloid, in order to prevent the progression of these lesions. This therapy is of particular value in the prophylactic treatment of conditions which are known to result in the development of hypertrophic scars and keloids (e.g., bums), and is preferably initiated after the proliferative phase has had time to progress (approximately 14 days after the initial injury), but before hypertrophic scar or keloid development. As noted above, the present invention also provides methods for treating neovascular diseases of the eye, including for example, comeal neovascularization, neovascular glaucoma, proliferative diabetic retinopathy, retrolental fibroplasia and macular degeneration.
|0727| Moreover, Ocular disorders associated with neovascularization which can be treated with the albumin fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention include, but are not limited to: neovascular glaucoma, diabetic retinopathy, retinoblastoma, retrolental fibroplasia, tiveitis, retinopathy of prematurity macular degeneration, comeal graft ncovascularizalion, as well as other eye inflammatory diseases, ocular tumors and diseases associated with choroidal or iris neovascularization. See, e.g., reviews by Waltman el til.. Am. J. Ophlhal <704-710 and gartner el at.. surv. ophlhal.> |0728| Thus, within one aspect of the present invention methods are provided for treating neovascular diseases of the eye such as comeal neovascularization (including comeal graft neovascularization), comprising the step of administering to a patient a therapeutically effective amount of a compound (e.g., fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention) to the comca, such that the formation of blood vessels is inhibited. Briefly, the cornea is a tissue which normally lacks blood vessels. In certain pathological conditions however, capillaries may extend inlo the cornea from the pericomeal vascular plexus of the limbus. When the cornea becomes vascularizcd, it also becomes clouded, resulting in a decline in the patient's visual acuity. Visual loss may become complete if the cornea completely opacitates. A wide variety of disorders can result in corneal neovascularization, including for example, comeal infections (e.g., trachoma, herpes simplex keratitis,
leishmaniasis and onchocerciasis), immunological processes (e.g., graft rejection and Stevens-Johnson's syndrome), alkali burns, trauma, inflammation (of any cause), toxic and nutritional deficiency states, and as a complication of wearing contact lenses.
|0729| Within particularly preferred embodiments of the invention, may be prepared for topical administration in saline (combined with any of the preservatives and antimicrobial agents commonly used in ocular preparations), and administered in eyedrop form. The solution or suspension may be prepared in its pure form and administered several times daily. Alternatively, anti-angiogenic compositions, prepared as described above, may also be administered directly to the cornea. Within preferred embodiments, the anti-angiogenic composition is prepared with a muco-adhesive polymer which binds to cornea. Within further embodiments, the anti-angiogenic factors or anti-angiogenic compositions may be utilized as an adjunct to conventional steroid therapy. Topical therapy may also be useful prophylactically in corneal lesions which are known to have a high probability of inducing an angiogenic response (such as chemical burns). In these instances the treatment, likely in combination with steroids, may be instituted immediately to help prevent subsequent complications.
|0730| Within other embodiments, the compounds described above may be injected directly into the comeal stroma by an ophthalmologist under microscopic guidance. The preferred site of injection may vary with the morphology of the individual lesion, but the goal of the administration would he to place the composition at the advancing front of the vasculature (i.e., interspersed between the blood vessels and the normal cornea). In most cases this would involve perilimbic comeal injection to "protect" the cornea from the advancing blood vessels. This method may also be utilized shortly after a corneal insult in order to prophylactically prevent comeal neovascularization. In this situation the material could be injected in the perilimbic cornea interspersed between the corneal lesion and its undesired potential limbic blood supply. Such methods may also be utilized in a similar fashion to prevent capillary invasion of transplanted corneas. In a sustained-release form injections might only be required 2-3 times per year. A steroid could also be added to the injection solution to reduce inflammation resulting from the injection itself.
[0731] Within another aspect of the present invention, methods are provided for treating ncovascular glaucoma, comprising the step of administering to a patient a therapeutically effective amount of an albumin fusion protein of the invention and/or polynucleotldes encoding an albumin fusion protein of the invention to the eye, such that the formation of blood vessels is inhibited. In one embodiment, the compound may be administered topically to the eye in order to treat early forms of neovascular glaucoma. Within other embodiments, the compound may be implanted by injection into the region of the anterior chamber angle. Within other embodiments, the compound may also be placed in any location such that the compound is continuously released into the aqueous humor. Within another aspect of the present invention, methods are provided for treating proliferate diabetic retinopathy, comprising the step of administering to a patient a therapeutically effective amount of an albumin fusion protein of the invention and/or polynucleotides encoding an albumin fusion protein of the invention to the eyes, such that the formation of blood vessels is inhibited.
|0732| Within particularly preferred embodiments of the invention, proliferative diabetic retinopathy may be treated by injection into the aqueous humor or the vitreous, in order to increase the local concentration of the polynucleotide, polypeptide, antagonist and/or agonist in the retina. Preferably, this treatment should be initiated prior to the acquisition of severe disease requiring photocoagulation.
|0733| Within another aspect of the present invention, methods are provided for treating retrolental fibroplasia, comprising the step of administering to a patient a therapeutically effective amount of an albumin fusion protein of the invention and/or polynucleotides encoding an albumin fusion protein of the invention to the eye, such that the formation of blood vessels is inhibited. The compound may be administered topically, via intravitreous injection and/or via intraocular implants.
|0734| Additionally, disorders which can be treated with fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention include, but are not limited to, hemangioma, arthritis, psoriasis, angiofibroma, atherosclerotic plaques, delayed wound healing, granulations, hemophilic joints, hypertrophic scars, nonunion fractures, Osier-Weber syndrome, pyogemc granuloma, scleroderma, trachoma, and vascular adhesions.
[0735] Moreover, disorders and/or states, which can be treated, prevented, diagnosed, and/or prognosed with the the albumin fusion proteins of [he invention and/or polynucleotides encoding albumin fusion proteins of the invention of the invention include, but are not limited to, solid tumors, blood born tumors such as leukemias, tumor metastasis, Kaposi's sarcoma, benign tumors, for example hemangiomas, acoustic neuromas, ncurofibromas, trachomas, and pyogenic granulomas, rheumatoid arthritis, psoriasis, ocular angiogenic diseases, for example, diabetic retinopalhy, lelinopathy of prematurity, macular degeneration, comeal graft rejection, ncovascular glaucoma, retrolental fibroplasia, rubeosis, retinoblastoma, and uvietis, delayed wound healing, endometriosis, vascluogencsis, granulations, hypertrophic scars (keloids), nonunion fractures, scleroderma, trachoma, vascular adhesions, myocardial angiogenesis, coronary collaterals, cerebral collaterals, arteriovenous malformations, ischemic limb angiogencsis, Osier-Webber Syndrome, plaque neovascularization, telangiectasia, hemophiliac joints, angiofibroma fibromuscular dysplasia, wound granulation, C'rohn's disease, atherosclerosis, birth control agent by preventing vascularization required for embryo implantation controlling menstruation, diseases that have angiogenesis as a pathologic consequence such as cat scratch disease (Rochele minalia quintosa), ulcers (Helicnbacter pylon), Bartoncllosis and bacillary angiomatosis.
|0736| In one aspect of the birth control method, an amount of the compound sufficient to block embryo implantation is administered before or after intercourse and fertilization have occurred, thus providing an effective method of birth control, possibly a "morning after" method. Albumin
fusion proteins of the invention ana/or polynucleolides encoding albumin fusion proteins of the invention may also be used in controlling menstruation or administered as either a peritoneal lavage fluid or for peritoneal implantation in the treatment of endometriosis. |0737| Albumin fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention may be incorporated into surgical sutures in order to prevent stitch granulomas.
|0738| Albumin fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention may be utilized in a wide variety of surgical procedures. For example, within one aspect of the present invention a compositions (in the form of, for example, a spray or film) may be utilized to coat or spray an area prior to removal of a tumor, in order to isolate normal surrounding tissues from malignant tissue, and/or to prevent the spread of disease to surrounding tissues. Within other aspects of the present invention, compositions (e.g., in the form of a spray) may be delivered via endoscopic procedures in order to coat tumors, or inhibit angiogcnesis in a desired locale. Within yet other aspects of the present invention, surgical meshes which have been coated with anti- angiogenic compositions of the present invention may be utilized in any procedure wherein a surgical mesh might be utilized. For example, within one embodiment of the invention a surgical mesh laden with an anti-angiogenic composition may be utilized during abdominal cancer resection surgery (e.g., subsequent to colon resection) in order to provide support to the structure, and to release an amount of the anti-angiogenic factor.
|0739| Within further aspects of the present invention, methods are provided for treating tumor excision sites, comprising administering albumin fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention to the resection margins of a tumor subsequent to excision, such that the local recurrence of cancer and the formation of new blood vessels at the site is inhibited. Within one embodiment of the invention, the anti-angiogenic compound is administered directly to the tumor excision site (e.g., applied by swabbing, brushing or otherwise coating the resection margins of the tumor with the anti-angiogenic compound). Alternatively, the anti-angiogenic compounds may be incorporated into known surgical pastes prior to administration. Within particularly preferred embodiments of the invention, the anti-angiogenic compounds are applied after hepatic resections for malignancy, and after neurosurgical operations.
[0740| Within one aspect of the present invention, fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention may be administered to the resection margin of a wide variety of tumors, including for example, breast, colon, brain and hepatic tumors. For example, within one embodiment of the invention, anti-angiogenic compounds may be administered to the site of a neurological tumor subsequent to excision, such that the formation of new blood vessels at the site are inhibited.
[0741] The albumin fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention may also be administered along with other anti-angiogenic factors. Representative examples of other anti-angiogenic factors include: Anti-Invasive Factor, retinoic acid and derivatives thereof, paclitaxcl, Suramin, Tissue Inhibitor of Metalloproteinase-1, Tissue Inhibitor of Metalloproteinase-2, Plasminogen Activator lnhibitor-1, Plasminogen Activator Inhibitor-2, and various forms of the lighter "d group" transition metals. |0742| 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.
|0743| 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 sulPate hydrates such as vanadyl sulfate mono- and trihydrates.
|0744] Representative examples of tungsten and molybdenum complexes also include oxo complexes. Suitable oxo tungsten complexes include tungstate and tungsten oxide complexes. Suitable tungslate 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 niolybdate, molybdenum oxide, and molybdenyl complexes. Suitable molybdate complexes include ammonium molybdate and its hydrates, sodium molybdale 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, glyccrol, tartaric acid, and sugars.
|0745] A wide variety of other anti-angiogenic factors may also be utilized within the context of the present invention. Representative examples include platelet factor 4; protamine sulphate; sulphated chitin derivatives (prepared from queen crab shells), (Murata et al., Cancer Res. 51:22-26, 1991); Sulphaled Polysaccharide Peptidoglycan Complex (SP- PG) (the function of this compound may be enhanced by the presence of steroids such as estrogen, and tamoxifen citrate); Staurosporine; modulators of matrix metabolism, including for example, proline analogs, cishydroxyproline, d,L-3,4-dehydroproline, Thiaproline, alpha,alpha-dipyridyl, aminopropionitrile fumarate; 4-propyl-5-(4-pyridinyl)-2(3H)-oxazolone; Methotrexate; Mitoxantrone; Heparin; Interferons; 2 Macroglobulin-serum; ChIMP-3 (Pavloff et al., J. 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)); anticollagcnase-serum; alpha2-antiplasmin (Holmes ct al., J. Biol. Chem. 262(4):1659-1664, (1987)); Bisantrcne (National Cancer Institute);
Lobenzarit disodium (N-(2)-carboxyphcnyl-4- chloroanthronilic acid disodiurn or "CCA"; Takeuchi et al., Agents Actions 36:312-316, (1992)); Thalidomide; Angostatic steroid; AGM-1470; carboxynaminolmidazole; and mctalloproteinase inhibitors such as BB94.
Diseases at the Cellular Level
[0746| Diseases associated with increased cell survival or the inhibition of apoptosis that could be treated, prevented, diagnosed, and/or prognosed using fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins 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, retmoblastoma, 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 ovanan 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 glomeruloncphritis 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.
|0747| 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 metasis of cancers, in particular those listed above.
[0748| Additional diseases or conditions associated with increased cell survival that could 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, 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 erythroleukemia)) 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, angiosarcoma, endotheliosarcoma, lymphangiosarcoma, 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, hepatorna, bile duct carcinoma, choriocarcinoma, seminorna, embryonal carcinoma, Wilm's tumor, cervical cancer, tcsticular tumor, lung carcinoma, small cell lung carcinoma, bladder carcinoma, epithelial carcinoma, gliorna, astrocytoma, medulloblastoma, craniopharyngioma, epcndymoma, pinealoma, hemangioblastorna, acoustic neuroma, oligodcndroglioma, menangioma, melanoma, neuroblastoma, and retinoblastorna.
|0749) Diseases associated with increased apoptosis that could be treated, prevented, diagnosed, and/or prognesed using fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention, include, but are not limited to, 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 rcpcrfusion injury), liver injury (e.g., hepatitis related liver injury, ischcmia/repcrfusion injury, cholestosis (bile duct injury) and liver cancer); toxin-induced liver disease (such as that caused by alcohol), septic shock, cachexia and anorexia.
Wound Healing and Epithelial Cell Proliferation
|0750] In accordance with yet a further aspect of the present invention, there is provided a process for utilizing fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention, for therapeutic purposes, for example, to stimulate epithelial cell proliferation and basal keratinocytes for the purpose of wound healing, and to stimulate hair follicle production and healing of dermal wounds. Albumin fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention, may be clinically useful in stimulating wound healing including surgical wounds, excisional wounds, deep wounds involving damage of the dermis and epidermis, eye tissue wounds, dental tissue wounds, oral cavity wounds, diabetic ulcers, dermal ulcers, cubitus ulcers, arterial ulcers, venous stasis ulcers, burns resulting Irani heat exposure or chemicals, and other abnormal wound healing conditions such as uremia, malnutrition, vitamin deficiencies and complications associated with systemic treatment with sleroids, radiation therapy and antineoplastic drugs and antimctabolites. Albumin fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention, could be used to promote dermal reestablishment subsequent to derma! loss
|0751] Albumin fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention, could be used to increase the adherence of skin grafts to a wound bed and to stimulate re-tpithelialization from the wound bed. The following arc types of grafts that fusion proteins of the invention and/or polynudeotides encoding albumin fusion proteins of the invention, could be used to increase adherence to a wound bed: autografts, artificial skin, allografts, autodermic graft, autocpdcrmic grafts, avacular grafts, Blair-Brown grafts, bone graft, brcphoplastic
grafts, cutis graft, delayed gralt, dermic graft, epidermic graft, fascia graft, full thickness graft, hcterologous graft, xenogratt, homologous graft, hyperplastic graft, lamellar graft, mesh graft, mucosal graft, Ollier-Thiersch graft, omenpal graft, patch graft, pedicle graft, penetrating graft, split |07S21 It is believed that fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention, will also produce changes in hepatocyte proliferation, and epithelial cell proliferation in the lung, breast, pancreas, stomach, small intestine, and large intestine. Albumin fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention, could promote proliferation of epithelial cells such as sebocytes, hair follicles, hepatocytes, type 11 pneumocytes, mucin-producing goblet cells, and other epithelial cells and their progenitors contained within the skin, lung, liver, and gastrointestinal tract. Albumin fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention, may promote proliferation of endothelial cells, keratinocytes, and basal keratinocytes.
|0753] Albumin fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention, could also be used to reduce the side effects of gut toxicity that result from radiation, chemotherapy treatments or viral infections. Albumin fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention, may have a cytoprotective effect on the small intestine mucosa. Albumin fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention, may also stimulate healing of mucositis (mouth ulcers) that result from chemotherapy and viral infections.
|0754| Albumin fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention, could further be used in full regeneration of skin in full and partial thickness skin defects, including burns, (i.e., repopulation of hair follicles, sweat glands, and sebaceous glands), treatment of other skin defects such as psoriasis. Albumin fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention, could be used to treat epidermolysis bullosa, a defect in adherence of the epidermis to the underlying dermis which results in frequent, open and painful blisters by accelerating reepithelialization of these lesions. Albumin fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention, could also be used to treat gastnc and doudenal ulcers and help heal by scar formation of the mucosal lining and regeneration of glandular mucosa and duodenal mucosal lining more rapidly. Inflammatory bowel diseases, such as Crohn's disease and ulcerative colitis, are diseases which result in destruction of the mucosal surface of the small or large intestine, respectively. Thus, fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention, could be used to promote the resurfacing of the mucosal surface to aid more rapid healing and to prevent progression of inflammatory bowel disease. Treatment with fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention, is expected to have a significant effect on the production of mucus throughout the gastrointestinal tract and could be used to protect the intestinal mucosa from injurious substances that are ingested or following surgery. Albumin fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention, could be used to treat diseases associate with the under expression.
|0755] Moreover, fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention, could be used to prevent and heal damage to the lungs due to various pathological states. Albumin fusion proteins of the invention and/or polynuclcotides encoding albumin fusion proteins of the invention, which could stimulate proliferation and differentiation and promote the repair of alveoli and brochiolar epithelium lo prevent or treat acute or chronic lung damage. For example, emphysema, which results in the progressive loss of aveoli, and inhalation injuries, i.e., resulting from smoke inhalation and bums, that cause necrosis of the bronchiolar epithelium and alveoli could be effectively treated using polynucleotides or polypcptides, agonists or antagonists of the present invention. Also fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention, could be used to stimulate the proliferation of and differentiation of type II pneumocytes, which may help treat or prevent disease such as hyaline membrane diseases, such as infant respiratory distress syndrome and bronchopulmonary displasia, in premature infants.
|0756| Albumin fusion proteins of the invention anoVor polynucleotides encoding albumin fusion proteins of the invention, could stimulate the proliferation and differentiation of hepatocytes and, thus, could be used to alleviate or treat liver diseases and pathologies such as fulminant liver failure caused by cirrhosis, liver damage caused by viral hepatitis and toxic substances (i.e., acetaminophen, carbon tctraholoride and other hepatoloxins known in the art).
[0757| In addition, fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention, could be used treat or prevent the onset of diabetes mcllitus. In patients with newly diagnosed Types I and II diabetes, where some islet cell function remains, fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention, could be used to maintain the islet function so as to alleviate, delay or prevent permanent manifestation of the disease. Also, fusion proteins of the invention and/or polynuclcotides encoding albumin fusion proteins of the invention, could be used as an auxiliary in islet cell transplantation lo improve or promote islet cell function.
Neural Activity and Neurological Diseases
|07S8| The albumin fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention may be used for (he diagnosis and/or treatment of diseases, disorders, damage or injury of the brain and/or nervous system. Nervous system disorders that can be
treated with the compositions ot the invention (e.g., fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins ot the invention), include, but are not limited to, nervous system injuries, and diseases or disorders which result in either a disconnection of axons, a diminution or degeneration of neurons, or demyi:lin;ition. Nervous system lesions which may be treated in a patient (including human and non-human mammalian patients) according to the methods of the invention, include but are not limited to, the following lesions of either the central (including spinal cord, brain) or peripheral nervous systems: (1) ischemic lesions, in which a lack of oxygen in a portion of the nervous system results in neuronal injury or death, including cerebral infarction or ischemia, or spinal cord infarction or ischemia; (2) traumatic lesions, including lesions caused by physical injury or associated with surgery, for example, lesions which sever a portion of the nervous system, or compression injuries; (3) malignant lesions, in which a portion of the nervous system is destroyed or injured by malignant tissue which is either a nervous system associated malignancy or a malignancy derived from non-nervous system tissue; (4) infectious lesions, in which a portion of the nervous system is destroyed or injured as a result of infection, for example, by an abscess or associated with infection by human immunodeficiency virus, herpes zoster, or herpes simplex virus or with Lyme disease, tuberculosis, or syphilis; (5) degenerative lesions, in which a portion of the nervous system is destroyed or injured as a result of a degenerative process including but not limited to, degeneration associated with Parkinson's disease, Alzheimer's disease, Huntington's chorea, or amyotrophic lateral sclerosis (ALS); (6) lesions associated with nutritional diseases or disorders, in which a portion of the nervous system is destroyed or injured by a nutritional disorder or disorder of metabolism including, but not limited to, vitamin B12 deficiency, folic acid deficiency, Wemicke disease, tobacco-alcohol amblyopia, Marchiafava-Bignami disease (primary degeneration of the corpus callosum), and alcoholic cerebellar degeneration; (7) neurological lesions associated with systemic diseases including, but not limited to, diabetes (diabetic neuropathy, Bell's palsy), systemic lupus erythematosus, carcinoma, or sarcoidosis; (8) lesions caused by toxic substances including alcohol, lead, or particular neurotoxins; and (9) demyelinated lesions in which a portion of the nervous system is destroyed or injured by a demyelinating disease including, but not limited to, multiple sclerosis, human immunodeficiency virus-associated myelopathy, transverse myelopathy or various etiologies, progressive multifocal leukoencephalopathy, and central pontine myelinolysis.
[0759] In one embodiment, the albumin fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention are used to protect neural cells from the damaging effects of hypoxia. In a further preferred embodiment, the albumin fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention are used to protect neural cells from the damaging effects of cerebral hypoxia. According to this embodiment, the compositions of the invention are used to treat or prevent neural cell injury associated with cerebral hypoxia. In one non-exclusive aspect of this embodiment, the albumin fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention, are used to treat or prevent neural cell injury associated with cerebral ischemia. In another non-exclusive aspect of this embodiment, the albumin fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention are used to treat or prevent neural cell injury associated with cerebral infarction.
|0760] In another preferred embodiment, albumin fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention are used to treat or prevent neural cell injury associated with a stroke. In a specific embodiment, albumin fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention are used to treat or prevent cerebral neural cell injury associated with a stroke.
|0761| In another preferred embodiment, albumin fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention are used to treat or prevent neural cell injury associated with a heart attack. In a specific embodiment, albumin fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention are used to treat or prevent cerebral neural cell injury associated with a heart attack.
|0762| The compositions ol the invention which are useful for treating or preventing a nervous system disorder may be selected by testing for biological activity in promoting the survival or differentiation of neurons. For example, and not by way of limitation, compositions of the invention which elicit any of the following effects may be useful according to the invention: (I) increased survival time of neurons in culture either in the presence or absence of hypoxia or hypoxic conditions; (2) increased sprouting of neurons in culture or in vivo; (3) increased production of a neuron-associated molecule in culture or in vivo, e.g., choline acetyltransferase or acelylcholincsterase with respect to motor neurons; or (4) decreased symptoms of neuron dysfunction in vivo. Such effects may be measured by any method known in the art. In preferred, non-limiting embodiments, increased survival of neurons may routinely be measured using a method set forth herein or otherwise known in the art, such as, for example, in Zhang et ill., Proc Nail Acad Sci USA 97:3637-42 (2000) or in Arakawael «/.. J. Neurosci., /0:3507-15 (1990); increased sprouting of neurons may be detected by methods known in the art, such as, for example, the methods set forth in Pestronk et al., Exp. Neural., 70:65-82 (1980), or Brown el ai. Ann. Rev. Ncurosci., 4:\1-A,1 (1981); increased production of neuron-associated molecules may be measured by bioassay, enzymatic assay, antibody binding, Northern blot assay, etc., using techniques blown in the art and depending on the molecule to be measured; and motor neuron dysfunction may be measured by assessing the physical manifestation of motor neuron disorder, e.g., weakness, motor neuron conduction velocity, or functional disability.
|076J] in specific embodiments, motor neuron disorders that may be treated according to the invention include, but are not limited to, disorders such as infarction, infection, exposure to toxin, trauma, surgical damage, degenerative disease or malignancy that may affect motor neurons as well
as other components ot the nervous system, as well as disorders that selectively affect neurons such as amyotrophic lateral sclerosis, and including, hut not limited to, progressive spinal muscular atrophy, progressive bulbar palsy, primary lateral sclerosis, infantile and juvenile muscular atrophy, progressive bulbar paralysis of childhood (Fazio-Londe syndrome), poliomyelitis and the post polio syndrome, and Hereditary Motorsensory Neuropathy (Charcot-Marie-Tooth Disease).
(0764) Further, fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention may play a role in neuronal survival; synapse formation; conductance, neural differentiation, etc. Thus, compositions of the invention (including fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention) may be used to diagnose and/or treat or prevent diseases or disorders associated with these roles, including, but not limited to, learning and/or cognition disorders. The compositions of the invention may also be useful in the treatment or prevention of neurodegenerative disease states and/or behavioural disorders. Such neurodegenerative disease states and/or behavioral disorders include, but are not limited to, Alzheimer's Disease, Parkinson's Disease, Huntington's Disease, Tourette Syndrome, schizophrenia, mama, dementia, paranoia, obsessive compulsive disorder, panic disorder, learning disabilities, ALS, psychoses, autism, and altered behaviors, including disorders in feeding, sleep patterns, balance, and perception. In addition, compositions of the invention may also play a role in the treatment, prevention and/or detection of developmental disorders associated with the developing embryo, or sexually-linked disorders. |0765| Additionally, fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention, may be useful in protecting neural cells from diseases, damage, disorders, or injury, associated with cerebrovascular disorders including, but not limited to, carotid artery diseases (e.g., carotid artery thrombosis, carotid stenosis, or Moyamoya Disease), cerebral amyloid angiopathy, cerebral aneurysm, cerebral anoxia, cerebral arteriosclerosis, cerebral arteriovenous malformations, cerebral artery diseases, cerebral embolism and thrombosis (e.g., carotid artery thrombosis, sinus thrombosis, or Wallenberg's Syndrome), cerebral hemorrhage (e.g., epidura! or subdural hematoma, or subarachnoid hemorrhage), cerebral infarction, cerebral ischemia (e.g., transient cerebral ischemia, Subclavian Steal Syndrome, or vertebrobasilar insufficiency), vascular dementia (e.g., multi-infarct), leukomalacia, periventricular, and vascular headache (e.g., cluster headache or migraines). |0766] In accordance with yet a further aspect of the present invention, there is provided a process for utilizing fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention, for therapeutic purposes, for example, to stimulate neurological cell proliferation and/or differentiation. Therefore, fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention may be used to treat and/or detect neurologic diseases. 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 nervous system disease or disorder. [0767] Examples of neurologic diseases which can be treated or detected with fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention include, brain diseases, such as metabolic brain diseases which includes phcnylketonuria such as maternal phenylketonuria, pyruvate carboxylase deficiency, pyruvate dehydrogenase complex deficiency, Wemickc's Encephalopathy, brain edema, brain neoplasms such as ccrcbcllar neoplasms which include infratentorial neoplasms, cerebral ventricle neoplasms such as choroid plexus neoplasms, hypothiilamic neoplasms, supratcntorial neoplasms, canavan disease, ccrcbellar diseases such as cerebellar ataxia which include spinoccrcbellar degeneration such as ataxia telangiectasia, cerebellar dyssynergia, Friederich's Ataxia, Machado-Joseph Disease, olivopontocerebellar atrophy, cerebellar neoplasms such as infratentorial neoplasms, diffuse cerebral sclerosis such as encephalitis periaxialis, globoid cell leukodystrophy, metachromatic leukodystrophy and subacute sclerosing panencephalitis.
|0768| Additional neurologic diseases which can be treated or detected with fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention include cerebrovascular disorders (such as carotid artery diseases which include carotid artery thrombosis, carotid stenosis and Moyamoya Disease), cerebral amyloid angiopathy, cerebral aneurysm, cerebral anoxia, cerebral arteriosclerosis, cerebral arteriovenous malformations, cerebral artery diseases, cerebral embolism and thrombosis such as carotid artery thrombosis, sinus thrombosis and Wallenberg's Syndrome, cerebral hemorrhage such as epidural hematoma, subdural hematoma and subarachnoid hemorrhage, cerebral infarction, cerebral ischemia such as transient cerebral ischemia, Subclavian Steal Syndrome and vertebrobasilar insufficiency, vascular dementia such as mulli-infarct dementia, periventricular leukomalacia, vascular headache such as cluster headache and migraine.
|0769| Additional neurologic diseases which can be treated or detected with fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention include dementia such as AIDS Dementia Complex, presenile dementia such as Alzheimer's Disease and Creutzfeldt-Jakob Syndrome, senile dementia such as Alzheimer's Disease and progressive supranuclear palsy, vascular dementia such as multi-infarct dementia, encephalitis which include encephalitis periaxialis, viral encephalitis such as epidemic encephalitis, Japanese Encephalitis, St. i.ouis Encephalitis, tick-borne encephalitis and West Nile Fever, acute disseminated encephalomyelitis, meningoencephalitis such as uveomeningoencephalitic syndrome, Postencephalitic Parkinson Disease and subacute sclerosing panencephalitis, encephalomalacia such as periventricular leukomalacia, epilepsy such as generalized epilepsy which includes infantile spasms, absence epilepsy, myoclonic epilepsy which includes MERRF Syndrome, tonic-clonic epilepsy, partial epilepsy such as complex partial epilepsy, frontal lobe epilepsy and temporal lobe epilepsy, post-traumatic epilepsy, status epilepticus such as Epilcpsia Partialis Continue, and Hallervorden-Spatz Syndrome.
J0770] Additional neurologic diseases which can be treated or detected with fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention include hydroccphalus such as Dandy-Walker Syndrome and normal pressure hydrocephalus, hypothalamic
diseases such as hypothalamic neoplasms, cerebral malaria, narcolepsy which includes cataplexy, bulbar poliomyelitis, cerebn pseudotumor, Kelt Syndrome, Reye's Syndrome, thalamic diseases, cerebral toxoplasmosis, intracranial tuberculoma and Zellweger Syndrome, central nervous system infections such as AIDS Dementia Complex, Drain Abscess, subdural empyema, encephalomyclitis such as Equine Encephalomyelitis. Venezuelan Equine Encephalomyelitis, Necrotizing Hemorrhagic Encephalomyelitis, Visna, and cerebral malaria.
[07711 Additional neurologic diseases which can be treated or detected with fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention include meningitis such as arachnoiditis, aseptic mcningtitis such as viral meningtitis which includes lymphocytic choriomeningitis, Bacterial meningtitis which includes Haemophilus Mcningtitis, Ltsteria Meningtitis, Meningococcal Menmgtilis such as Waterhouse-Friderichsen Syndrome, Pneumococcal Meningtitis and meningeal tuberculosis, fungal meningitis such as Cryptococcal Meningtilis, subdural effusion, meningoencephalitis such as uvemeningoencephalitic syndrome, myelitis such as transverse myelitis, neurosyphilis such as tabes dorsalis, poliomyelitis which includes bulbar poliomyelitis and postpoliomyelitis syndrome, prion diseases (such as Creutzfeldt-Jakob Syndrome, Bovine Spongiform Encephalopathy, Gerstmann-Straussler Syndrome, Kuril, Scrapie), and cerebral toxoplasmosis.
|0772| Additional neurologic diseases which can be treated or detected with fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention include central nervous system neoplasms such as brain neoplasms that include cerebellar neoplasms such as infratentorial neoplasms, cerebral ventricle neoplasms such as choroid plexus neoplasms, hypothalamic neoplasms and supratentorial neoplasms, meningeal neoplasms, spinal cord neoplasms which include epidural neoplasms, demyelinating diseases such as Canavan Diseases, diffuse cerebral sceloris which includes adrenoleukodystrophy, encephalitis periaxialis, globoid cell leukodystrophy, diffuse cerebral sclerosis such as mctachromatic leukodystrophy, allergic encephalomyelitis, necrotizing hemorrhagic cncephalomyelitis, progressive multifocal leukoencephalopathy, multiple sclerosis, central pontine myelinolysis, transverse myelitis, neuromyclitis optica, Scrapie, Swayback, Chronic Fatigue Syndrome, Visna, High Pressure Nervous Syndrome, Meningism, spinal cord diseases such as amyotonia congenita, amyotrophic lateral sclerosis, spinal muscular atrophy such as Werdnig-Hoffmann Disease, spinal cord compression, spinal cord neoplasms such as epidural neoplasms, syringomyelia, Tabes Dorsalis, Stiff-Man Syndrome, mental retardation such as Angelman Syndrome, Cri-du-Chat Syndrome, De Lange's Syndrome, Down Syndrome, Gangliosidoses such as gangliosidoses G(M1), Sandhoff Disease, Tay-Sachs Disease, Hartnup Disease, homocystinuria, Laurence-Moon- Biedl Syndrome, Lesch-Nyhan Syndrome, Maple Syrup Urine Disease, mucolipidosis such as fucosidosis, neuronal ceroid-lipofuscinosis, ocuiocerebrorenal syndrome, phenylketonuria such as maternal phenylketonuria, Prader-Willi Syndrome, Reft Syndrome, Rubinstein-Taybi Syndrome, Tuberous Sclerosis, WAGR Syndrome, nervous system abnormalities such as holoprosencephaly, neural tube defects such as anencephaly which includes hydrangencephaly, Arnold-Chairi Deformity, cncephalocele, meningocele, meningomyelocele, spinal dysraphism such as spina bifida cystica and spina bifida occulta.
|0773| Additional neurologic diseases which can be treated or detected with fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention include hereditary motor and sensory neuropathies which include Charcot-Marie Disease, Hereditary optic atrophy, Refsum's Disease, hereditary spastic paraplegia, Werdnig-Hoffmann Disease, Hereditary Sensory and Autonomic Neuropathies such as Congenital Analgesia and Familial Dysautonomia, Neurologic manifestations (such as agnosia that include Gerstmann's Syndrome, Amnesia such as retrograde amnesia, apraxia, neurogenic bladder, cataplexy, communicative disorders such as hearing disorders that includes deafness, partial hearing loss, loudness recruitment and tinnitus, language disorders such as aphasia which include agraphia, anomia, broca aphasia, and Wemicke Aphasia, Dyslexia such as Acquired Dyslexia, language development disorders, speech disorders such as aphasia which includes anomia, broca aphasia and Wernicke Aphasia, articulation disorders, communicative disorders such as speech disorders which include dysarthria, echolalia, mutism and stuttering, voice disorders such as aphonia and hoarseness, decerebrate state, delirium, fasciculation, hallucinations, meningism, movement disorders such as angelman syndrome, ataxia, athetosis, chorea, dystonia, hypokinesia, muscle hypotonia, myoclonus, tic, torticollis and tremor, muscle hypertonia such as muscle rigidity such as stiff-man syndrome, muscle spasticity, paralysis such as facial paralysis which includes Herpes Zoster Oticus, Gastroparcsis, Hcmiplegia, ophthalmoplegia such as diplopia, Duane's Syndrome, Homer's Syndrome, Chronic progressive external ophthalmoplegia such as Keams Syndrome, Bulbar Paralysis, Tropical Spastic Paraparesis, Paraplegia such as Brown-Sequard Syndrome, quadriplegia, respiratory paralysis and vocal cord paralysis, paresis, phantom limb, taste disorders such as ageusia and dysgeusia, vision disorders such as amblyopia, blindness, color vision defects, diplopia, hemianopsia, scotoma and subnormal vision, sleep disorders such as hypersomnia which includes KJeine-Levin Syndrome, insomnia, and somnambulism, spasm such as trismus, unconsciousness such as coma, persistent vegetative state and syncope and vertigo, neuromuscular diseases such as amyotonia congenita, amyotrophic lateral sclerosis, Lambert-Eaton Myasthenic Syndrome, motor neuron disease, muscular atrophy such as spinal muscular atrophy, Charcot-Maric Disease and Werdnig-Hoffmann Disease, Postpoliomyelitis Syndrome, Muscular Dystrophy, Myasthenia Gravis, Myotonia Atrophica, Myotonia Confenita, Nemaline Myopathy, Familial Periodic Paralysis, Multiplex Pararnyloclonus, Tropical Spastic Paraparesis and Stiff-Man Syndrome, peripheral nervous system diseases such as ncrodynia, amyloid neuropathies, autonomic nervous system diseases such as Adie's Syndrome, Barre-Lieou Syndrome, Familial Dysautonomia, Homer's Syndrome, Reflex Sympathetic Dystrophy and Shy-Drager Syndrome, Cranial Nerve Diseases such as Acoustic Nerve Diseases such as Acoustic Neuroma which includes Ncurofibromatosis 2, Facial Nerve Diseases such as Facial Neuralgia,Melkersson-Roscnthal Syndrome, ocular motility disorders which includes amblyopia, nystagmus, oculomotor nerve paralysis, ophthalmoplegia such as Duane's Syndrome, Horncr's
Syndrome, Chronic Progressive External Ophthalmoplegia which includes Keams Syndrome, Strabismus such as Esotropia and Exolropia, Oculomotor Nerve Paralysis, Optic Nerve Diseases such as Optic Atrophy which includes Hereditary Optic Atrophy, Optic Disk Drusen, Optic Neuritis such as Neuromyelitis Optica, Papilledcma, Trigeminal Neuralgia, Vocal Cord Paralysis, Demyelinating Diseases such as Neuromyelitis Optica and Swayback, and Diabetic neuropathies such as diabetic foot.
|0774] Additional neurologic diseases which can be treated or detected with fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention include nerve compression syndromes such as carpal tunnel syndrome, tarsal tunnel syndrome, thoracic outlet syndrome such as cervical rib syndrome, ulnar nerve compression syndrome, neuralgia such as causalgia, cervico-brachial neuralgia, facial neuralgia and trigeminal neuralgia, neuritis such as experimental allergic neuritis, optic neuritis, polyneuritis, polyradiculoneuritis and radiculities such as polyradiculitis, hereditary motor and sensory neuropathies such as Charcot-Marie Disease, Hereditary Optic Atrophy, Refsum's Disease, Hereditary Spastic Paraplegia and Werdnig-Hoffmann Disease, Hereditary Sensory and Autonomic Neuropathies which include Congenital Analgesia and Familial Dysautonomia, POEMS Syndrome, Sciatica, Gustatory Sweating and Tetany).
Endocrine Disorders
|0775) Albumin fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention, may be used to treat, prevent, diagnose, and/or prognosc disorders and/or diseases related to hormone imbalance, and/or disorders or diseases of the endocrine system. (0776J Hormones secreted by the glands of the endocrine system control physical growth, sexual function, metabolism, and other functions. Disorders may be classified in two ways: disturbances in the production of hormones, and the inability of tissues to respond to hormones. The etiology of these hormone imbalance or endocrine system diseases, disorders or conditions may be genetic, somatic, such as cancer and some autoimmune diseases, acquired (e.g., by chemotherapy, injury 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 disease or disorder related to the endocrine system and/or hormone imbalance.
|0777| Endocrine system and/or hormone imbalance and/or diseases encompass disorders of uterine motility including, but not limited to: complications with pregnancy and labor (e.g., pre-term labor, post-term pregnancy, spontaneous abortion, and slow or stopped labor); and disorders and/or diseases of the menstrual cycle (e.g., dysrnenorrhea and cndometriosis).
|0778| Endocrine system and/or hormone imbalance disorders and/or diseases include disorders and/or diseases of the pancreas, such as, for example, diabetes mellitus, diabetes insipidus, congenital pancreatic agenesis, pheochromocytoma-islet cell tumor syndrome; disorders and/or diseases of the adrenal glands such as, for example, Addison's Disease, corticosteroid deficiency, virilizing disease, hirsiitism, Cushing's Syndrome, hyperaldosteronism, pheochromocytoma; disorders and/or diseases of the pituitary gland, such as, for example, hvperpituilarism, hypopituitari;,m, pituitary dwarfism, pituitary adenoma, panhypopituitarism, acromegaly, gigantism; disorders and/or diseases of the thyroid, including but not limited to, hyperthyroidism, hypothyroidism, Plummer's disease, Graves' disease (toxic diffuse goiter), toxic nodular goiter, thyroiditis (Hashimoto's thyroiditis, subacute granulomatous thyroiditis, and silent lymphocytic thyroiditis), Pendred's syndrome, myxedema, cretinism, thyrotoxicosis, thyroid hormone coupling defect, thymic aplasia, Hurthle cell tumours of the thyroid, thyroid cancer, thyroid carcinoma, Medullary thyroid carcinoma, disorders and/or diseases of the parathyroid, such as, for example, hyperparathyroidism, hvpoparathyroidism; disorders and/or diseases of the hypothatamus.
[0779] In addition, endocrine system and/or hormone imbalance disorders and/or diseases may also include disorders and/or diseases of the lesles or ovaries, including cancer. Other disorders and/or diseases of the testes or ovaries further include, for example, ovarian cancer, polycystic ovary syndrome, Klinefelter's syndrome, vanishing testes syndrome (bilateral anorchia), congenital absence of Leydig's cells, cryplorchidism, Noonan's syndrome, myotonic dystrophy, capillary haernangioma of the testis (benign), neoplasias of the testis and nco-testis. |0780| Moreover, endocrine system and/or hormone imbalance disorders and/or diseases may also include disorders and'or diseases such as, for example, polyglandular deficiency syndromes, pheochromocytoma, neuroblastoma, multiple Endocrine neoplasia, and disorders and/or cancers of endocrine tissues.
|078I| In another 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 endocrine diseases and/or disorders associated with the tissue(s) in which the Therapeutic protein corresponding to the Therapeutic protein portion of the albumin protein of the invention is expressed,
Reproductive System Disorders
|0782| The albumin fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention may be used for the diagnosis, treatment, or prevention of diseases and/or disorders of the reproductive system. Reproductive system disorders that can be treated by the compositions of the invention, include, but are not limited to, reproductive system injuries, infections, neoplastic disorders, congenital defects, and diseases or disorders which result in infertility, complications with pregnancy, labor, or parturition, and postpartum difficulties. |0783| Reproductive system disorders and/or diseases include diseases and/or disorders of the testes, including testicular atrophy, testicular fcminiyation, cryptorchisin (unilateral and bilateral), anorchia, ectopic testis, epididymitis and orchitis (typically resulting from infections such as, for example, gonorrhea, mumps, tuberculosis, and syphilis), testicular torsion, vasitis nodosa, germ cell tumors (e.g., scminomas, embryonal cell
carcinomas, teratocarcinomas, chonocarcmomas, yolk sac tumors, and teratomas), stromal tumors (e.g., Leydig cell tumors), hydrocelc, hcm.itocele, varicocele, spermatocele, inguinal hcmia, and disorders of sperm production (e.g., immotile cilia syndrome, aspcrmia, asthenozoospermia, azoospcrmia, oligo^nf-nnia, and teratozoospermiaY
[0784] Reproductive system disorders also include disorders of the prostate gland, such as acute non-bacterial prostatitis, chronic non-hacterial prostatitis, acute bacterial prostatitis, chronic bacteria] prostatitis, prostatodystonia, prostatosis, granulomatous prostatitis, malacoplakia, benign prostatic hypertrophy or hyperplasia, and prostate neoplastic disorders, including adenocarcinomas, transitional cell carcinomas, ductal carcinomas, and squamous cell carcinomas.
(0785) Additionally, the compositions of the invention may be useful in the diagnosis, treatment, and/or prevention of disorders or diseases of the penis and urethra, including inflammatory disorders, such as balanoposlhitis, balanitis xerotica obliterans, phimosis, paraphimosis, syphilis, herpes simplex virus, gonorrhea, non-gonococcal urcthritis, chlamydia, mycoplasma, trichomonas, HIV, AIDS, Reiter's syndrome, condyloma acuminatum, condyloma latum, and pearly penile papules; urethral abnormalities, such as hypospadias, epicardia’s, and phimosis; premalignant lesions, including Erythroplasia of Queyrat, Bowen's disease, Bowenoid paplosis, giant condyloma of Buscke-Lowenstein, and varrucous carcinoma; penile cancers, including squamous cell carcinomas, carcinoma in situ, verrucous carcinoma, and disseminated penile carcinoma; urethral neoplastic disorders, including penile urethral carcinoma, bulb membranous urethral carcinoma, and prostatic urethral carcinoma; and erectile disorders, such as priapism, Peyronie's disease, erectile dysfunction, and impotence.
[0786] Moreover, diseases and/or disorders of the vas deferens include masculinities and CBAVD (congenital bilateral absence of the vas deferens); additionally, the albumin fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention may be used in the diagnosis, treatment, and/or prevention of diseases and/or disorders of the seminal vesicles, including hydatid disease, congenital chloride diarrhea, and polycystic kidney disease.
(0787] Other disorders and/or diseases of the male reproductive system include, for example, Klinefelter's syndrome, Young's syndrome, premature ejaculation, diabetes rnellitus, cystic fibrosis, Kartagcner's syndrome, high fever, multiple sclerosis, and gynecomastia. [0788] Further, the polynucleotides, fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention may be used in the diagnosis, treatment, and/or prevention of diseases and/or disorders of the vagina and vulva, including bacterial vaginosis, Candida vaginitis, herpes simplex virus, chancroid, granuloma inguinale, lymphogranuloma vcnercum, scabies, human papillomavims, vaginal trauma, vulvar trauma, adenosis, chlamydia vaginitis, gonorrhea, trichomonas vaginitis, condyloma acuminatum, syphilis, molluscum contagiosum, airophic vaginitis, Paget's disease, lichen sclerosus, lichen planus, vulvodynia, toxic shock syndrome, vaginismus, vulvovaginitis, vulvar vcstibulitis, and neoplastic disorders, such as squamous cell hyperplasia, clear cell carcinoma, basal cell carcinoma, melanomas, cancer of Bartholin's gland, and vulvar intraepithelial neoplasia.
[0789] Disorders and/or diseases of the uterus include dysmcnonhca, retroverted uterus, cndometriosis, fibroids, adenomyosis, anovulatory bleeding, amenorrhea, Cushing's syndrome, hydatidiform moles, Asherman's syndrome, premature menopause, precocious puberty, uterine polyps, dysfunctional uterine bleeding (e.g., due to aberrant hormonal signals), and neoplastic disorders, such as adenocarcinomas, keiomyosarcornas, and sarcomas. Additionally, the albumin fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention may be useful as a marker or detector of, as well as in the diagnosis, treatment, and/or prevention of congenital uterine abnormalities, such as bicornuate uterus, septate uterus, simple unicomuatc uterus, unicornuate uterus with a noncavitary rudimentary hom, unicornuate uterus with a non-communicating cavitary rudimentary horn, unicornuate uterus with a communicating cavitary hom, arcuate uterus, uterine didelfus, and T-shaped uterus.
[0790] Ovarian diseases and/or disorders include anovulation, polycystic ovary syndrome (Stein-Leventhal syndrome), ovarian cysts, ovarian hypofunction, ovarian insensitivity to gonadotropins, ovarian overproduction of androgens, right ovarian vein syndrome, amenorrhea, hirutism, and ovarian cancer (including, but not limited to, primary and secondary cancerous growth, Sertoli-Lcydig tumors, endometriod carcinoma of the ovary, ovarian papillary serous adenocarcinoma, ovarian rnucinous adenocarcinoma, and Ovarian Krukenberg tumors).
[0791] Cervical diseases and/or disorders include ccrvicitis, chronic cervicitis, mucopurulent cervicitis, cervical dysplasia, cervical polyps, Nabothian cysts, cervical erosion, cervical incompetence, and cervical neoplasms (including, for example, cervical carcinoma, squamous metaplasia, squamous cell carcinoma, adenosquamous cell neoplasia, and columnar cell neoplasia).
|0792| Additionally, diseases and/or disorders of the reproductive system include disorders and/or diseases of pregnancy, including miscarriage and stillbirth, such as early abortion, late abortion, spontaneous abortion, induced abortion, therapeutic abortion, threatened abortion, missed abortion, incomplete abortion, complete abortion, habitual abortion, missed abortion, and septic abortion; ectopic pregnancy, anemia, Rh incompatibility, vaginal bleeding during pregnancy, gestational diabetes, intrauterine growth retardation, polyhydramnios, HELLP syndrome, abruptio placentae, placenta previa, hypcremesis, preeclampsia, eclampsia, herpes gestationis, and urticaria of pregnancy. Additionally, the albumin fusion prolcins of the invention and/or polynucleoiides encoding albumin fusion proteins of the invention may be used in the diagnosis, treatment, and/or prevention of diseases that can complicate pregnancy, including heart disease, heart failure, rheumatic heart disease, congenital heart disease, mitral valve prolapse, high blood pressure, anemia, kidney disease, infectious disease (e.g., rubella, cytomegalovirus, toxoplasmosis, infectious
hepatitis, cmamyclia, HIV, ailo, ana genital herpes), diabetes mellitus, Graves' disease, thyroiditis, hypothyroidism, Hashimoto's thyroiditis,
chronic active hepatitis, cirrhosis of the liver, primary biliary cirrhosis, aslhma, systemic lupus eryematosis, rheumatoid arthritis, myasthenia gravis,
idiopathic thrombocytopcnic purpura, appendicitis, ovarian cysts, gallbladder disorders.and obstruction of the intestine.
|0793] Complications associated with labor and parturition include premature rupture of the membranes, pre-term labor, post-term pregnancy,
postmaturity, labor that progresses too slowly, fetal distress (e.g., abnormal heart rate (fetal or maternal), breathing problems, and abnormal fetal
position), shoulder dystocia, prolapsed umbilical cord, amniotic fluid embolism, and aberrant uterine bleeding.
10794) Further, diseases and/or disorders of the postdelivery period, including endometritis, myometritis, parametritis, peritonitis, pelvic
thrombophlebitis, pulmonary embolism, endotoxemia, pyelonephritis, saphenous thrombophlebitis, mastitis, cystitis, postpartum hemorrhage, and
inverted uterus.
(0795| Other disorders and/or diseases of the female reproductive system that may be diagnosed, treated, and/or prevented by the albumin
fusion proteins of the invention and/or polynucleotidcs encoding albumin fusion proteins of the invention include, for example, Turner's syndrome,
pseudohermaphroditism, premenstrual syndrome, pelvic inflammatory disease, pelvic congestion (vascular engorgement), frigidity, anorgasmia,
dyspareunia, ruptured fallopian tube, and Mittelschmerz.
Infectious Disease
(0796] Albumin fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention can be used to treat or detect infectious agents. For example, by increasing the immune response, particularly increasing the proliferation and differentiation of B and/or T cells, infectious diseases may be treated. 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, without necessarily eliciting an immune response.
[0797] Viruses arc one example of an infectious agent that can cause disease or symptoms that can be treated or detected by albumin fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention. Examples of viruses, include, but are not limited to Examples of viruses, include, but are not limited to the following DNA and RNA viruses and viral families: Arbovirus, Adenoviridae, Arenaviridae, Arterivirus, Bimaviridae, Bunyavirkiac, Caliciviridae, Circoviridae, Coronaviridae, Dengue, EBV, HIV, Flaviviridae, Hepadnaviridae (Hepatitis), I Icrpcsviridae (such as, Cytomegalovirus, Herpes Simplex, Herpes Zoster), Mononegavirus (e.g., Paramyxoviridae, Morbillivirus, Rhabdoviridae), Orthomyxoviridae (e.g., Influenza A, Influenza B, and parainfluenza), Papiloma virus, Papovaviridac, Parvoviridae, Picornaviridae, Foxviridae (such as Smallpox or Vaccinia), Reoviridae (e.g., Rotavirus), Retroviridae (HTLV-I, HTLV-II, Lentivirus), and Togaviridae (e.g., Rubivirus). Viruses falling within these families can cause a variety of diseases or symptoms, including, but not limited to: arthritis, bronchiollitis, respiratory syncytial virus, encephalitis, eye infections (e.g., conjunctivitis, keratitis), chronic fatigue syndrome, hepatitis (A, B, C, E, Chronic Active, Delta), Japanese B encephalitis, junin, Chikungunya, Rift Valley fever, yellow fever, meningitis, opportunistic infections (e.g., AIDS), pneumonia, Burkitt's Lymphoma, chickenpox, hemorrhagic fever, Measles, Mumps, Parainfluenza, Rabies, the common cold, Polio, leukemia, Rubella, sexually transmitted diseases, skin diseases (e.g., Kaposi's, warts), and viremia. Albumin fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention, can be used to treat or detect any of these symptoms or diseases. In specific embodiments, fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention are used to treat: meningitis, Dengue, EBV, and/or hepatitis (e.g., hepatitis B). In an additional specific embodiment fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention are used to treat patients nonresponsive to one or more other commercially available hepatitis vaccines. In a further specific embodiment fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention are used Id treat AIDS.
[0798| Similarly, bacterial and fungal agents that can cause disease or symptoms and that can be treated or detected by albumin fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention include, but not limited to, the following Gram-Negative and Gram-positive bacteria, bacterial families, and fungi: Actinomyces (e.g., Norcardia), Acinetobacter, Cryplococcus neoformans, Aspergillus, Bacillaceae (e.g., Bacillus aalhrasis), Bacteroides (e.g., Oacteroides fragilis), Blastomycosis, Bordetella, Borrelia (e.g., Borrelia burgilorferi), Urucella, Candidia, Campylobacter, Chlamydia, Clostridium (e.g., Closlridium boliilinum, Closlridium dijici/e, Closlridium perfriiigens, Clostriiiium Iclani), Coccidioides, Corynebacterium (e.g., Corynebaclcrium diplheriae), Cryptococcus, Dermatocycoses, E. coli (e.g, Enterotoxigenic E, coli and Enterohemorrhagic E. coli), Enterobacter (e.g. Enterobticter aerogenes), Enterobacteriaceae (Klebsiella, Salmonella (eg., Salmonella typhi, Salmonella entcritidis, Salmonella typhi), Serratia, Yersinia, Shigella), Erysipelothrix, Haemophilus (e.g., Haemophilus influcnia type B), Helicobactcr, Legionella (e.g., Legionel/a pneumophila), Leptospira, Listcria (e.g., Listeria monocylogenes), Mycoplasma, Mycobacterium (e.g , Mycobacierium leprae and Mycobncterium tuberculosis). Vibrio (e.g., Vibrio cholerae), Neisseriaceac (e.g., Neisseria gonorrhea, Neisseria meningiiidis), Pasteurellacea, Proteus, Pseudomoras (e.g., Pseudomonns aeruginosa), Rickettsiaceae, Spirochetes (e.g., Trcponcma spp., Leptospira spp., Borrelia spp.), Shigella spp., Staphylococcus (e.g., Slaphylococcus aureus), Meningiococcus, Pncumococcus and Streptococcus (e.g., Streptococcus pneumonias and Groups A, B, and C Streptococci), and Ureaplasmas. These bacterial, parasitic, and fungal families can cause diseases or symptoms, including, but not limited to: antibiotic-resistant infections, bacteremia, endocarditis, septicemia, eye
infections (e.g., conjunctivitis), uveitis, tuberculosis, gingivitis, bacterial diarrhea, opportunistic infections (e.g., AIDS related infections), paronychia, prosthesis-related infections, dental caries, Reiter's Disease, respiratory tract infections, such as Whooping Cough or Empyerna, sepsis, Lyme Disease, Cat-Scratch Disease, dysentery, paratyphoid fever, food poisoning, Legionella disease, chronic and acute inflammation, erythema, yeast infections, typhoid, pneumonia, gonorrhea, meningitis (e.g., mengitis types A and B), chlamydia, syphillis, diphtheria, leprosy, brucellosis, peptic ulcers, anthrax, spontaneous abortions, birth defects, pneumonia, lung infections, ear infections, deafness, blindness, lethargy, malaise, vomiting, chronic diarrhea, Crohn's disease, colitis, vaginosis, sterility, pelvic inflammatory diseases, candidiasis, paratuberculosis, tuberculosis, lupus, botulism, gangrene, tetanus, impetigo, Rheumatic Fever, Scarlet Fever, sexually transmitted diseases, skin diseases (e.g., cellulitis, dcrmatocycoses), toxemia, urinary tract infections, wound infections, noscomial infections. Albumin fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention, can be used to treat or detect any of these symptoms or diseases. In specific embodiments, fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention are used to treat: tetanus, diptheria, botulism, and/or meningitis type B.
[0799] Moreover, parasitic agents causing disease or symptoms that can be treated, prevented, and/or diagnosed by fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention include, but not limited to, the following families or class: Amebiasis, Babesiosis, Coccidiosis, Cryptosporidiosis, Dientamoebiasis, Dourine, Ectoparasitic, Giardias, Helminthiasis, Leishmaniasis, Schistisoma, Theilcriasis, Toxoplasmosis, Trypanosomiasis, and Trichomonas and Sporozoans (e.g., Plasmodium virax, Plasmodiumfalciparium, I'lasmodium malarias and Plasmodium ovule). These parasites can cause a variety of diseases or symptoms, including, but not limited to: Scabies, Trombiculiasis, eye infections, intestinal disease (e.g., dysentery, giardiasis), liver disease, lung disease, opportunistic infections (e.g., AIDS related), malaria, pregnancy complications, and toxoplasmosis. Albumin fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention, can be used to treat, prevent, and/or diagnose any of these symptoms or diseases. In specific embodiments, fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention are used to treat, prevent, and/or diagnose malaria.
[0800] Albumin fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention could either be by administering an effective amount of an albumin fusion protein of the invention to the patient, or by removing cells from the patient, supplying the cells with a polynucleotide of the present invention, and returning the engineered cells to the patient (ex vivo therapy). Moreover, the albumin fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention can be used as an antigen in a vaccine to raise an immune response against infectious disease.
Regeneration
[0801] Albumin fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention can be used lo differentiate, proliferate, and attract cells, leading lo the regeneration of tissues. (See, Science 276:59-87 (1997)). The regeneration of tissues could be used to repair, replace, or protect tissue damaged by congenital defects, trauma (wounds, bums, incisions, or ulcers), age, disease (e.g. osteoporosis, osteocarthritis, periodontal disease, liver failure), surgery, including cosmetic plastic surgery, tibrosis, rcperfusion injury, or systemic cytokine damage.
[0802| Tissues that could be regenerated using the present invention include organs (e.g., pancreas, liver, intestine, kidney, skin, endothelium), muscle (smooth, skeletal or cardiac), vasculature (including vascular and lymphatics), nervous, hematopoietic, and skeletal (bone, cartilage, tendon, and ligament) tissue. Preferably, regeneration occurs without or decreased scarring. Regeneration also may include angiogenesis. [0803J Moreover, fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention, may increase regeneration of tissues difficult to heal. For example, increased tendon/ligament regeneration would quicken recovery time after damage. Albumin fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention could also be used prophylactically in an efforl to avoid damage. Specific diseases that could be treated include of tendinitis, carpal tunnel syndrome, and other tendon or ligament defects. A further example of tissue regeneration of non-healing wounds includes pressure ulcers, ulcers associated with vascular insufficiency, surgical, and traumatic wounds.
[0804] Similarly, nerve and brain tissue could also be regenerated by using fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention, to proliferate and differentiate nerve cells. Diseases that could be treated using this method include central and peripheral nervous system diseases, neuropathies, or mechanical and traumatic disorders (e.g., spinal cord disorders, head trauma, cerebrovascular disease, and stoke). Specifically, diseases associated with peripheral nerve injuries, peripheral neuropathy (e.g., resulting from chemotherapy or other medical therapies), localized neuropathies, and central nervous system diseases (e.g., Alzheimer's disease, Parkinson's disease, Huntington's disease, amyotrophic lateral sclerosis, and Shy-Drager syndrome), could all be treated using the albumin fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention.
Gastrointestinal Disorders
|080S| Albumin 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 gastrointestinal disorders, including inflammatory diseases and/or conditions, infections, cancers (e.g., intestinal
neoplasms (carcinoid tumor ot the small intestine, non-Hodgkin's lymphoma of the small intestine, small bow] lymphoma)), and ulcers, such as peptic ulcers.
10806) Gastrointestinal disorders include dysphagia, odynophagia, inflammation of the esophagus, peptic esophagitis. gastric reflux, submucosal fibrosis and stricturing, Mallory-Weiss lesions, leiomyomas, lipomas, epidermal cancers, adeoncarcinomas, gastric retention disorders, gastroenteritis, gastric atrophy, gastric/stomach cancers, polyps of the stomach, autoimmune disorders such as pernicious anemia, pyloric stenosis, gastritis (bacterial, viral, eosinophilic, stress-induced, chronic erosive, atrophic, plasma cell, and Menetrier's), and peritoneal diseases (e.g., chyloperioncum, hemoperitoncum, mesenteric cyst, mesenteric lymphadenitis, mescnteric vascular occlusion, panniculitis, neoplasms, peritonitis, pneumopcritoneum, bubphrenic abscess,).
|0807] Gastrointestinal disorders also include disorders associated with the small intestine, such as malabsorption syndromes, distension, irritable bowel syndrome, sugar intolerance, celiac disease, duodenal ulcers, duodenitis, tropical sprue, Whipple's disease, intestinal lymphangiectasia, Crohn's disease, appendicitis, obstructions of the ileum, Mcckel's diverticulum, multiple diverticula, failure of complete rotation of the small and large intestine, lymphoma, and bacterial and parasitic diseases (such as Traveler's diarrhea, typhoid and paratyphoid, cholera, infection by Roundworms (Ascariasis lumbricoides), Hookworms (Ancylosloma duodenale). Threadworms (Enterobius vermicularis), Tapeworms (Taenia saginata, Echinococcus granulosus, Diphyllobothrium spp., and '/'. solium).
[0808| Liver diseases and/or disorders include intrahepatic cholestasis (alagille syndrome, biliary liver cirrhosis), fatty liver (alcoholic fatty liver, reye syndrome), hepatic vein thrombosis, hepatolentricular degeneration, hepatomegaly, hepatopulmoriary syndrome, hepatorenal syndrome, portal hypertension (esophagcal and gastric varices), liver abscess (amebic liver abscess), liver cirrhosis (alcoholic, biliary and experimental), alcoholic liver diseases (fatty liver, hepatitis, cirrhosis), parasitic (hepatic cchinococcosis, fascioliasis, amebic liver abscess), jaundice (hcmolytic, hepatocellular, and choleslatic), cholestasis, portal hypertension, liver enlargement, ascites, hepatitis (alcoholic hepatitis, animal hepatitis, chronic hepatitis (autoimmune, hepatitis B, hepatitis C, hepatitis D, drug induced), toxic hepatitis, viral human hepatitis (hepatitis A, hepatitis B, hepatitis C, hepatitis D, hepatitis E), Wilson's disease, granulomatous hepatitis, secondary biliary cirrhosis, hepatic encephalopathy, portal hypertension, varices, hepatic encephalopathy, primary biliary cirrhosis, primary sclerosing cholangitis, hepatocellular adenoma, hemangiomas, bile stones, liver failure (hepatic encephalopathy, acute liver failure), and liver neoplasms (angiomyolipoma, calcified liver metastases, cystic liver metastases, epithelial tumors, fibrolamellar hepatocarcinoma, focal nodular hyperplasia, hepatic adenoma, hepatobiliary cystadenoma, hepatoblastoma, hepatocellular carcinoma, hepatoma, liver cancer, liver hemangioendothclioma, mesenchymal hamartoma, mesencriymal tumors of liver, nodular regenerative hyperplasia, benign liver tumors (Hepatic cysts [Simple cysts, Polycystic liver disease, Hepatobiliary cystadenoma, Choledochal cyst], Mesenchymal tumors [Mesenchymal hamartoma, Infantile hemangioendothelioma, Hemangioma, Peliosis hepatis, Lipomas, Inflammatory pseudotumor, Miscellaneous], Epithelial tumors [Bile duct epithelium (Bile duct hamartoma, Bile duct adenoma), Hepatocyte (Adenoma, Focal nodular hyperplasia, Nodular regenerative hyperplasia)], malignant liver tumors [hepatocellular, hepatoblastoma, hepatocellular carcinoma, cholangioccllular, cholangiocarcinoma, cystadenocarcinoma, tumors of blood vessels, angiosarcoma, Karposi's sarcoma, hemangioendothelioma, other tumors, embryonal sarcoma, fibrosarcoma, leiomyosarcoma, rhabdomyosarcoma, carcinosarcorna, teratoma, carcinoid, squamous carcinoma, primary lymphoma]), peliosis hepatis, erythrohepatic porphyria, hepatic porphyria (acute intermittent porphyria, porphyria cutanea larda), Zellweger syndrome).
|0809| Pancreatic diseases and/or disorders include acute pancreatitis, chronic pancreatitis (acute necrotizing pancreatitis, alcoholic pancreatitis), neoplasms (adenocarcinoma of the pancreas, cystadenocarcinoma, insulinoma, gastrinoma, and glucagonoma, cystic neoplasms, islet-cell tumors, pancreoblastoma), and other pancreatic diseases (e.g., cystic fibrosis, cyst (pancreatic pseudocyst, pancreatic fistula, insufficiency)). |0810] Gallbladder diseases include gallstones (cholelithiasis and choledocholithiasis), postcholecystectomy syndrome, diverticulosis of the gallbladder, acute cholecystitis, chronic cholecystitis, bile duct tumors, and mucoccle.
|0811] Diseases and/or disorders of the large intestine include antibiotic-associated colitis, divcrticulitis. ulcerative colitis, acquired mcgacolon, abscesses, fungal and bacterial infections, anorectal disorders (e.g., fissures, hemorrhoids), colonic diseases (colitis, colonic neoplasms [colon cancer, adcnomatous colon polyps (e.g., villous adenoma), colon carcinoma, colorectal cancer], colonic diverticulitis, colonic diverticulosis, megacolon [Mirschsprung disease, toxic megacolon]; sigrnoid diseases [proctocolitis, sigmoin neoplasms]), constipation, Crohn's disease, diarrhea (infantile diarrhea, dysentery), duodenal diseases (duodenal neoplasms, duodenal obstruction, duodenal ulcer, duodenitis), enteritis (entcrocolitis), HIV cntcropathy, ilcal diseases (ileal neoplasms, ileitis), immunoproliferative small intestinal disease, inflammatory bowel disease (ulcerative colitis, Crohn's disease), intestinal atresia, parasitic diseases (anisakiasis, balantidiasis, blastocystis infections, cryptosporidiosis, dientamoebiasis, amebic dysentery, giardiasis), intestinal fistula (rectal fistula), intestinal neoplasms (cccal neoplasms, colonic neoplasms, duodenal neoplasms, ileal neoplasms, intestinal polyps, jejuna! neoplasms, rectal neoplasms), intestinal obstruction (afferent loop syndrome, duodenal obstruction, impacted fcccs, intestinal pseudo-obstruction [cecal volvulus], intussusception), intestinal perforation, intestinal polyps (colonic polyps, gardner syndrome, peutz-jeghers syndrome), jejuna] diseases (jejunal neoplasms), malabsorption syndromes (blind loop syndrome, celiac disease, lactose intolerance, short bowl syndrome, tropical sprue, whipple's disease), mesenteric vascular occlusion, pneumatosis cystoides intestinalis, protein-losing enteropathies (intestinal lymphagiectasis), rectal diseases (anus diseases, fecal incontinence, hemorrhoids, proctitis, recta) fistula, rectal prolapse,
rectocele), peptic ulcer (duodenal ulcer, peptic esophagitis, hemorrhage, perforation, stomach ulcer, Zollinger-Ellison syndrome), postgastrectomy syndromes (dumping syndrome), stomach diseases (e.g., achlorhydria, duodenogastric reflux (bile reflux), gastric antral vascular ectasia, gastric fistula, gastric outlet obstruction, gastritis (atrophic or hypertrophic), gastroparesis, stomach dilatation, stomach divcrticulum, stomach neoplasms (gastric cancer, gastric polyps, gastric adenocarcinama, hyperplastic gastric polyp), stomach rupture, stomach ulcer, stomach volvulus), tuberculosis, visceroptosis, vomiting (e.g., hematemesis, hyperernesis gravidarum, postoperative nausea and vomiting) and hemorrhagic colitis. |0812] Further diseases and/or disorders of the gastrointestinal system include biliary tract diseases, such as, gastroschisis, fistula (e.g., biliary fistula, esophageal fistula, gastric fistula, intestinal fistula, pancreatic fistula), neoplasms (e.g., biliary tract neoplasms, esophagcal neoplasms, such as adenocarcinoma of the esophagus, esophageal squamous cell carcinoma, gastrointestinal neoplasms, pancreatic neoplasms, such as adenocarcinoma of the pancreas, mucinous cystic neoplasm of the pancreas, pancreatic cystic neoplasms, pancreatoblastoma, and peritoneal neoplasms), esophageal disease (e.g., bullous diseases, candidiasis, glycogenic acanthosis, ulceration, barren esophagus varices, atresia, cyst, divcrticulum (e.g., Zenker's diverticulum), fistula (e.g., tracheoesophageal fistula), motility disorders (e.g., CREST syndrome, deglutition disorders, achalasia, spasm, gastroesophagcal reflux), neoplasms, perforation (e.g., Boerhaave syndrome, Mallory-Weiss syndrome), stenosis, esophagitis, diaphragmatic hernia (e.g., hiatal hernia); gastrointestinal diseases, such as, gastroenteritis (e.g., cholera morbus, norwalk virus infection), hemorrhage (e.g., hematemesis, mclena, peptic ulcer hemorrhage), stomach neoplasms (gastric cancer, gastric polyps, gastric adenocarcinoma, stomach cancer)), hernia (e.g., congenital diaphragmatic hernia, femoral hernia, inguinal hernia, obturator hernia, umbilical hernia, ventral hernia), and intestinal diseases (e.g., cecal diseases (appendicitis, cecal neoplasms)).
Chemotaxis
|0813] Albumin fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention may have chemotaxis activity. A chemotaxic molecule attracts or mobilizes cells (e.g., monocytes, fibroblasts, neutrophils, T-cells, mast cells, eosinophils, epithelial and/or endothelial cells) to a particular site in the body, such as inflammation, infection, or site of hyperproliferation. The mobilized cells can then tight off and/or heal the particular trauma or abnormality.
|0814| Albumin fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention may increase chemotaxic activity of particular cells. These chernotaclic molecules can then be used to treat inflammation, infection, hyperproliferative disorders, or any immune system disorder by increasing the number of cells targeted to a particular location in the body. For example, chemotaxic molecules can be used to treat wounds and other trauma to tissues by attracting immune cells to the injured location. Chcmotactic molecules of the present invention can also attract fibroblasts, which can be used to treat wounds.
|08I5] It is also contemplated that fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention may inhibit chcmotactic activity. These molecules could also be used to treat disorders. Thus, fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention could be used as an inhibitor of chemotaxis.
Binding Activity
|0816) Albumin fusion proteins of the invention may be used to screen for molecules that bind to the Therapeutic protein portion of the fusion protein or for molecules to which the Therapeutic protein portion of the fusion protein binds. The binding of the fusion protein and the molecule may activate (agonist), increase, inhibit (antagonist), or decrease activity of the fusion protein or the molecule bound. Examples of such molecules include antibodies, oligonucleolides, proteins (e.g., leceptors), or small molecules.
|()817| Preferably, the molecule is closely related to the natural ligand of the Therapeutic protein portion of the fusion protein of the invention, e.g., a fragment of the ligand, or a natural substrate, a ligand, a structural or functional mimetic. (See, Coligan et al., Current Protocols in Immunology l(2):Chapter 5 (1991)). Similarly, the molecule can be closely related to the natural receptor to which the Therapeutic protein portion of an albumin fusion protein of the invention binds, or at least, a fragment of the receptor capable of being bound by the Therapeutic protein portion of an albumin fusion protein of the invention (e.g., active site). In either case, the molecule can be rationally designed using known techniques. |0818| Preferably, the screening for these molecules involves producing appropriate cells which express the albumin fusion proteins of the invention. Preferred cells include cells from mammals, yeast, Drosophila, or E. coli.
|0819] The assay may simply test binding of a candidate compound to an albumin fusion protein of the invention, wherein binding is detected by a label, or in an assay involving competition with a labeled competitor. Further, the assay may test whether the candidate compound results in a signal generated by binding to the fusion protein.
|0820] Alternatively, the assay can be carried out using cell-free preparations, fusion protein/molecule affixed to a solid support, chemical libraries, or natural product mixtures. The assay may also simply comprise the steps of mixing a candidate compound with a solution containing an albumin fusion protein, measuring fusion proteinAnolecuIe activity or binding, and comparing the fusion protein/molecule activity or binding to a standard.
|0821| Preferably, an EL1SA assay can measure fusion protein level or activity in a sample (e.g., biological sample) using a monoclonal or polyclonal antibody. The antibody can measure fusion protein level or activity by either binding, directly or indirectly, to the albumin fusion protein or by competing with the albumin fusion protein for a substrate.
Additionally, tne rccquur iu which a Therapeutic protein portion of an albumin fusion protein of the invention binds can be identified by numerous methods known to those of skill in the art, for example, ligand panning and FACS sorting (Coligan, et al.F Current Protocols in Immun., \(7) Chapter 5, (19911). For example, in cases wherein the Therapeutic protein portion of the fusion protein corresponds to FGF, expression cloning may be employed wherein polyadenylated RNA is prepared from a cell responsive to the albumin fusion protein, for example, NIH3T3 cells which are known to contain multiple receptors for the FGF family proteins, and SC-3 cells, and a cDNA library created from this RNA is divided into pools and used to transfcct COS cells or other cells that arc not responsive to the albumin fusion protein. Transfected cells which are grown on glass slides are exposed to the albumin fusion protein of the present invention, after they have been labeled. The albumin fusion proteins can be labeled by a variety of means including iodination or inclusion of a recognition site for a site-specific protein kinase.
[0823] Following fixation and incubation, the slides are subjected to auto-radiographic analysis. Positive pools are identified and sub-pools are prepared and rc-transfectcd using an iterative sub-pooling and re-scrccning process, eventually yielding a single clones that encodes the putative receptor.
|0824| As an alternative approach for receptor identification, a labeled albumin fusion protein can be photoaffinity linked with cell membrane or extract preparations that express the receptor molecule for the Therapeutoc protein component of an albumin fusion protein of the invention, the linked material may be resolved by PAGE analysis and exposed to X-ray film. The labeled complex containing the receptors of the fusion protein can be excised, resolved into peptide fragments, and subjected to protein microsequencing. The amino acid sequence obtained from microsequencing would be used to design a set of degenerate oligonucleotide probes to screen a cDNA library to identify the genes encoding the putative receptors.
|U825| Moreover, the techniques of gene-shuffling, motif-shuffling, exon-shuffling, and/or codon-shuffling (collectively referred to as "DNA shuffling") may be employed to modulate the activities of the fusion protein, and/or Therapeutic protein portion or albumin component of an albumin fusion protein of the present invention, thereby effectively generating agonists and antagonists of an albumin fusion protein of the present invention. See generally, U.S. Patent Nos. 5,605,793, 5,811,238, 5,830,721, 5,834,252, and 5,837,458, and Patten, P. A., et ai. Curr. Opinion Biotcchnol. 8:724-33 (1997); Harayama, S. Trends Biolechnol. 16(2):76-82 (1998); Hansson, L. 0., et ai, J. Mol. Biol. 287:265-76 (1999); and Lorenzo, M. M. and Blasco, R. Biotechniques 24(2):308-13 (1998); each of these patents and publications are hereby incorporated by reference). In one embodiment, alteration of polynucleotides encoding albumin fusion proteins of the invention and thus, the albumin fusion proteins encoded thereby, may be achieved by DNA shuffling. DNA shuffling involves the assembly of two or more DNA segments into a desired molecule by homologous, or site-specific, recombination. In another embodiment, polynucleotides encoding albumin fusion proteins of the invention and thus, the albumin fusion proteins encoded thereby, may be altered by being subjected to random mutagenesis by error-prone PCR, random nucleotide insertion or other methods prior to recombination. In another embodiment, one or more components, motifs, sections, parts, domains, fragments, etc., of an albumin fusion protein of the present invention may be recombined with one or more components, motifs, sections, parts, domains, fragments, etc. of one or more hcterologous molecules. In preferred embodiments, the heterologous molecules are family members. In further preferred embodiments, the heterologous molecule is a growth factor such as, for example, platelet-derived growth factor (PDGF), insulin-like growth factor (1GF-I), transforming growth factor (TGF)-alpha, epidermal growth factor (EOF), fibroblast growth factor (FGF), TGF-beta, bone morphogenetic protein (BMP)-2, BMP-4, BMP-5, BMP-6, BMP-7, activins A and B, decapentaplegic(dpp), 60A, OP-2, dorsalin, growth differentiation factors (GDFs), nodal, MIS, inhibin-alpha, TGF-betal, TGF-beta2, TGF-beta3, TGF-beta5, and glial-derived neurotrophic factor (GDNF).
|0826| Other preferred fragments are biologically active fragments of the Therapeutic protein portion and/or albumin component of the albumin fusion proteins of the present invention. Biologically active fragments are those exhibiting activity similar, but not necessarily identical, to an activity of a Therapeutic protein portion and/or albumin component of the albumin fusion proteins of the present invention. The biological activity of the fragments may include an improved desired activity, or a decreased undesirable activity.
|0827] Additionally, this invention provides a method of screening compounds to identify those which modulate the action of an albumin fusion protein of the present invention. An example of such an assay comprises combining a mammalian fibroblast cell, an albumin fusion protein of the
present invention, and the compound to be screened and [HJ thymidine under cell culture conditions where the fibroblast cell would normally proliferate. A control assay may be performed in the absence of the compound to be screened and compared to the amount of fibroblast
proliferation in the presence of the compound to determine if the compound stimulates proliferation by determining the uptake of 3[H] Ihymidine in
each case. The amount of fibroblast cell proliferation is measured by liquid scintillation chromatography which measures the incorporation of [H] thymidine. Both agonist and antagonist compounds may be identified by this procedure.
[0828| In another method, a mammalian cell or membrane preparation expressing a receptor for the Therapeutic protien component of a fusion piotine of the invention is incubated with a labeled fusion protein of the present invention in the presence of the compound. The ability of the compound to enhance or block this interaction could then be measured. Alternatively, the response of a known second messenger system following interaction of a compound to be screened and the receptor is measured and the ability of the compound to bind to the receptor and elicit a second
messenger response is measured to determine if the compound is a potential fusion protein. Such second messenger systems include but are not
limited to, cAMP guanylate cyclase, ion channels or phosphoinositide hydrolysis.
[08291 All of these above assays can be used as diagnostic or prognostic markers. The molecules discovered using these assays can be used to
treat disease or to bring about a particular result in a patient (e.g., blood vessel growth) by activating or inhibiting the fusion protein/molecule.
Moreover, the assays can discover agents which may inhibit or enhance the production of the albumin fusion proteins of the invention from suitably
manipulated cells or tissues.
|0830| Therefore, the invention includes a method of identifying compounds which bind to an albumin fusion protein of the invention
comprising the steps of: (a) incubating a candidate binding compound with an albumin fusion protein of the present invention; and (b) determining
if binding has occurred. Moreover, the invention includes a method of identifying agonists/antagonists comprising the steps of: (a) incubating a
candidate compound with an albumin fusion protein of the present invention, (b) assaying a biological activity, and (b) determining if a biological
activity of the fusion protein has been altered. Targeted Delivery
[0831] In another embodiment, the invention provides a method of delivering compositions to targeted cells expressing a receptor for a component of an albumin fusion protein of the invention.
[0832| As discussed herein, fusion proteins of the invention may be associated with heterologous polypeptides, heterologous nucleic acids, toxins, or prodrugs via hydrophobic, hydrophilic, ionic and/or covalenl interactions. In one embodiment, the invention provides a method for the specific delivery of compositions of the invention to cells by administering fusion proteins of the invention (including 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., antiscnse or ribozymcs) 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.
[0833] In another embodiment, the invention provides a method for the specific destruction of cells (e.g., the destruction of tumor cells) by administering an albumin fusion protein of the invention (e.g., polypeptides of the invention or antibodies of the invention) in association with toxins or cytotoxic prodrugs.
[0834] By "toxin" is meant 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, thymidme kinase, cndonuclease, RNAse, alpha toxin, ricin, abrin, Pseudomonas exotoxin A, diphtheria toxin, saporin, momordin, gelonin, pokeweed antiviral protein, alpha-sarcin and cholera toxin. By "cytotoxic prodrug" is meant a non-toxic compound that is convened by an enzyme, normally present in the cell, into a cytotoxic compound. Cytotoxic prodrugs that may be used according to the methods of the invention include, but are not limited to, glutamyl derivatives of bcnzoic acid mustard alkylating agent, phosphate derivatives of etoposide or mitomycin C, cytosinc arabinoside, daunorubisin, and phenoxyacetamide derivatives of doxorubicin.
Drug Screening
|0835| Further contemplated is the use of the albumin fusion proteins of the present invention, or the polynucleotides encoding these fusion proteins, to screen for molecules which modify the activities of the albumin fusion protein of the present invention or proteins corresponding to the Therapeutic protein portion of the albumin fusion protein. Such a method would include contacting the fusion protein with a selected compound(s) suspected of having antagonist or agonist activity, and assaying the activity of the fusion protein following binding.
|()836[ This invention is particularly useful for screening therapeutic compounds by using the albumin fusion proteins of the present invention, or binding fragments thereof, in any of a variety of drug screening techniques. The albumin fusion protein employed in such a test may be affixed to a solid support, expressed on a cell surface, free in solution, or located intracellularly. One method of drug screening utilizes eukaryotic or prokaryotic host cells which are stably transformed with recombinant nucleic acids expressing the albumin fusion protein. Drugs are screened against such transformed cells or supernatants obtained from culturing such cells, in competitive binding assays. One may measure, for example, the formulation of complexes between the agent being tested and an albumin fusion protein of the present invention.
(0837] Thus, the present invention provides methods of screening for drugs or any other agents which affect activities mediated by the albumin fusion proteins of the present invention. These methods comprise contacting such an agent with an albumin fusion protein of the present invention or a fragment thereof and assaying for the presence of a complex between the agent and the albumin fusion protein or a fragment thereof, by methods well known in the art. In such a competitive binding assay, the agents to screen are typically labeled. Following incubation, free agent is separated from that present in bound form, and the amount of free or uncomplexed label is a measure of the ability of a particular agent to bind to ihe albumin fusion protein of the present invention.
|OK38| Another technique for drug screening provides high throughput screening for compounds having suitable binding affinity to an albumin
fusion protein of the present invention, and is described in great detail in European Patent Application 84/03564, published on September 13, 1984, which is incorporated herein by reference herein. Briefly stated, large numbers of different small pcptide test compounds are synthesized on a solid substrate, such as plastic pins or some other surface. The pcptide test compounds are reacted with an albumin fusion protein of the present invention and washed. Bound peptides are then detected by methods well known in the art. Purified albumin fusion protein may be coated directly onto plates for use in the aforementioned drug screening techniques, in addition, non-neutralizing antibodies may be used to capture the peptide and immobilize it on the solid support.
(0839) This invention also contemplates the use of competitive drug screening assays in which neutralizing antibodies capable of binding an albumin fusion protein of the present invention specifically compete with a test compound for binding to the albumin fusion protein or fragments thereof. In this manner, the antibodies are used to detect the presence of any peptide which shares one or more antigenic epitopes with an albumin fusion protein of the invention.
Binding Peptides and Other Molecules
|0840] The invention also encompasses screening methods for identifying polypeptides and nonpolypeptides that bind albumin fusion proteins of the invention, and the binding molecules identified thereby. These binding molecules are useful, for example, as agonists and antagonists of the albumin fusion proteins of the invention. Such agonists and antagonists can be used, in accordance with the invention, in the therapeutic embodiments described in detail, below.
|0841] This method comprises the steps of: contacting an albumin fusion protein of the invention with a plurality of molecules; and identifying a molecule that binds the albumin fusion protein.
|0842) The step of contacting the albumin fusion protein of the invention with the plurality of molecules may be effected in a number of ways. For example, one may contemplate immobilizing the albumin fusion protein on a solid support and bringing a solution of the plurality of molecules in contact with the immobilized polypeptides. Such a procedure would be akin to an affinity chromatographic process, with the affinity matrix being comprised of the immobilized albumin fusion protein of the invention. The molecules having a selective affinity for the albumin fusion protein can then be purified by affinity selection. The nature of the solid support, process for attachment of the albumin fusion protein to the solid support, solvent, and conditions of the affinity isolation or selection are largely conventional and well known to those of ordinary skill in the art. |0843] Alternatively, one may also separate a plurality of polypeptides into substantially separate fractions comprising a subset of or individual polypeptides. For instance, one can separate the plurality of polypeptides by gel electrophoresis, column chromatography, or like method known to those of ordinary skill for the separation of polypeptides. The individual polypeptides can also be produced by a transformed host cell in such a way as to be expressed on or about its outer surface (e.g., a rccombinant phage). Individual isolates can then be "probed" by an albumin fusion protein of the invention, optionally in the presence of an inducer should one be required for expression, to determine if any selective affinity interaction takes place between the albumin fusion protein and the individual clone. Prior to contacting the albumin fusion protein with each fraction comprising individual polypeptides, the polypeptides could first be transferred to a solid support for additional convenience. Such a solid support may simply be a piece of filter membrane, such as one made of nitrocellulose or nylon. In this manner, positive clones could be identified from a collection of transformed host cells of an expression library, which harbor a DNA construct encoding a polypeptide having a selective affinity for an albumin fusion protein of the invention. Furthermore, the amino acid sequence of the polypeptide having a selective affinity for an albumin fusion protein of the invention can be determined directly by conventional means or the coding sequence of the DNA encoding the polypeptide can frequently be determined more conveniently. The primary sequence can then be deduced from the corresponding DNA sequence. If the amino acid sequence is to be determined from the polypeptide itself, one may use microsequcncing techniques. The sequencing technique may include mass spectroscopy. |0844| In certain situations, it may be desirable to wash away any unbound polypeptides from a mixture of an albumin fusion protein of the invention and the plurality of polypeptides prior to attempting to determine or to detect the presence of a selective affinity interaction. Such a wash step may be particularly desirable when the albumin fusion protein of the invention or the plurality of polypeptides are bound to a solid support. (08451 The plurality of molecules provided according to this method may be provided by way of diversity libraries, such as random or combinatorial peptide or nonpeptide libraries which can be screened for molecules that specifically bind an albumin fusion protein of the invention. Many libraries are known in the art that can be used, e.g., chemically synthesized libraries, rccombinant (e.g., phage display libraries), and in vitro translation-based libraries. Examples of chemically synthesized libraries are described in Fodor etal., Science 251:767-773 (1991); Houghten etal.. Nature 354:84-86 (1991); Lam ct al.. Nature 354:82-84 (1991); Medynski, Bio/Technology 12:709-710 (1994); Gallop et al., J. Medicinal Chemistry 37(9):1233-1251 (1994); Ohlmeyer et al., Proc. Natl. Acad. Sci. USA 90:10922-10926 (1993); Erb et al., Proc. Natl. Acad. Sci. USA 91:11422-11426 (1994); Houghten et al., Biotechniques 13:412 (1992); Jayawickremc et al., Proc. Natl. Acad. Sci. USA 91:1614-1618 (1994); Salmon et al., Proc. Natl. Acad. Sci. USA 90:11708-11712 (1993); PCT Publication No. WO 93/20242; and Brenner and Lerner, Proc. Natl. Acad. Sci. USA 89:5381-5383(1992).
10846] Examples of phage display libraries are described in Scott et al., Science 249:386-390 (1990); Devlin et al., Science, 249:404^106 (1990); Christian et al., 1992, J. Mol. Biol. 227:711-718 1992); Lenstra, J. Immune!. Meth. 152:149-157 (1992); Kay et al., Gene 128:59-65 (1993): and PCT Publication No. WO 94/18318 dated Aug. 18, 1994.
|U847| In vitro translation-oaseu iimaries include but are not limited to those described in PCT Publication No. WO 91/05058 dated Apr. 18,
1991; and Mattheakis et al., Proc. Natl. Acad. Sci. USA 91:9022-9026 (1994).
|0848| By way of examples of nonpeptide libraries, a benzodiazepine library (see e.g., Bunin et al., Proc. Nail. Acad. Sci. USA 91:4708-4712
(1994)) can be adapted for use. Peptoid libraries (Simon ct al., Proc. Natl. Acad. Sci. USA 89:9367-9371 (1992)) can also be used. Another
example of a library that can be used, in which the amide functionalities in peptides have been permethylated to generate a chemically transformed
combinatorial library, is described by Ostresh et al. (Proc. Natl. Acad. Sci. USA 91:11138-11142 (1994)).
(0849) The variety of non-peptide libraries that are useful in the present invention is great. For example, Ecker and Crooke (Bio/Technology
13:351-360 (1995) list bcnzodiazepines, hydantoins, piperazinediones, biphenyls, sugar analogs, beta-mercaptoketones, arylacetic acids,
acylpiperidines, benzopyrans, cubanes, xanthines, aminimides, and oxazolones as among the chemical species that form the basis of various
libraries.
[0850| Non-peptide libraries can be classified broadly into two types: decorated monomers and oligomers. Decorated monomer libraries employ
a relatively simple scaffold structure upon which a variety functional groups is added. Often the scaffold will be a molecule with a known useful
pharmacological activity. For example, the scaffold might be the benzodiazepine structure.
|0851| Non-peptide oligomer libraries utilize a large number of monomers that are assembled together in ways that create new shapes that
depend on the order of the monomers. Among the monomer units that have been used are carbamates, pyrrolinoncs, and morpholinos. Peptoids,
peptide-likc oligomers in which the side chain is attached to the alpha amino group rather than the alpha carbon, form the basis of another version of
non-peptide oligomer libraries. The first non-peptide oligomer libraries utilized a single type of monomer and thus contained a repeating backbone.
Recent libraries have utilized more than one monomer, giving the libraries added flexibility.
|0852] Screening the libraries can be accomplished by any of a variety of commonly known methods. See, e.g., the following references, which
disclose screening of peptide libraries: Parmley et al., Adv. Exp. Med. Biol. 251:215-218 (1989); Scott et al,. Science 249:386-390 (1990); Howlkes
ct al., BioTechniques 13:422^127 (1992); Oldenburg et al., Proc. Natl. Acad. Sci. USA 89:5393-5397 (1992); Yu et al., Cell 76:933-945 (1994);
Staudt et al., Science 241:577-580 (1988); Bock el al., Nature 355:564-566 (1992); Tuerk et al., Proc. Natl. Acad. Sci. USA 89:6988-6992 (1992);
Ellington et al., Nature 355:850-852 (1992); U.S. Pat. No. 5,096,815, U.S. Pat. No. 5,223,409, and U.S. Pat. No. 5,198,346, all to Ladner et al.;
Rebar et al., Science 263:671-673 (1993); and PCT Publication No. WO 94/18318.
|0853| In a specific embodiment, screening to identify a molecule that binds an albumin fusion protein of the invention can be carried out by
contacting the library members with an albumin fusion protein of the invention immobilized on a solid phase and harvesting those library members
that bind to the albumin fusion protein. Examples of such screening methods, termed "panning" techniques are described by way of example in
Parmley et al., Gene 73:305-318 (1988); Fowlkcs et al., BioTechniques 13:422-427 (1992); PCT Publication No. WO 94/18318; and in references
cited herein.
[OH54J In another embodiment, the two-hybrid system for selecting interacting proteins in yeast (Fields et al., Nature 340:245-246 (1989);
Chien et al., Proc. Natl. Acad. Sci. USA 88:9578-9582 (1991) can be used to identify molecules that specifically bind to polypeptides of the
invention.
|08SS| Where the binding molecule is a polypcptide, the polypeptide can be conveniently selected from any peptide library, including random
pcptidc libraries, combinatorial peptide libraries, or biased peptide libraries. The term "biased" is used herein to mean that the method of generating
the library is manipulated so as to restrict one or more parameters that govern the diversity of the resulting collection of molecules, in this case
peptides.
(0856] Thus, a truly random peptide library would generate a collection of peptides in which the probability of finding a particular amino acid at
a given position of the peptide is the same for all 20 amino acids. A bias can be introduced into the library, however, by specifying, for example, that
a lysine occur every fifth amino acid or that positions 4, 8, and 9 of a decapcptidc library be fixed to include only arginine. Clearly, many types of
biases can be contemplated, and the present invention is not restricted to any particular bias. Furthermore, the present invention contemplates
specific types of peptide libraries, such as phage displayed peplide libraries and those that utilize a DNA construct comprising a lambda phage
vector with a DNA insert.
|0857| As mentioned above, in the case of a binding molecule that is a polypeptide, the polypeptide may have about 6 to less than about 60
jmino acid residues, preferably about 6 to about 10 amino acid residues, and most preferably, about 6 to about 22 amino acids. In another
embodiment, a binding polypeptide has in the range of 15-100 amino acids, or 20-50 amino acids.
|0858| The selected binding polypeptide can be obtained by chemical synthesis or recombinant expression.
Other Activities
|0859| An albumin fusion protein of the invention and/or polynucleotide encoding an albumin fusion protein of the invention, may be employed in treatment for stimulating re-vascularization of ischemic tissues due to various disease conditions such as thrombosis, arteriosclerosis, and other cardiovascular conditions. The albumin fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention may also be employed to stimulate angiogenesis and limb regeneration, as discussed above.
|UX6l)| An albumin fusion protein 01 the invention and/or polynucleotide encoding an albumin fusion protein of the invention may also be
employed for treating wounds due to injuries, burns, post-operative tissue repair, and ulcers since they are mitogenic to various cells of different
"rig'iis, such as fihrohlast cells and skeletal musr.lc cells, and therefore, facilitate the repair or replacement of damaged or diseased tissue.
|0861| An albumin fusion protein of the invention andVor polynucleotide encoding an albumin fusion protein of the invention may also be
employed stimulate neuronal growth and to treat and prevent neuronal damage which occurs in certain neuronal disorders or neuro-degenerative
conditions such as Alzheimer's disease, Parkinson's disease, and AIDS-rclated complex. An albumin fusion protein of the invention and/or
polynucleotide encoding an albumin fusion protein of the invention may have the ability to stimulate chondrocyte growth, therefore, they may be
employed to enhance bone and periodontal regeneration and aid in tissue transplants or bone grafts.
|0862] An albumin fusion protein of the invention and/or polynucleotide encoding an albumin fusion protein of the invention may be also be
employed to prevent skin aging due to sunburn by stimulating kcratinocyte growth.
|0863] An albumin fusion protein of the invention and/or polynucleotide encoding an albumin fusion protein of the invention may also be
employed for preventing hair loss. Along the same lines, an albumin fusion protein of the invention and/or polynucleotide encoding an albumin
fusion protein of the invention may be employed to stimulate growth and differentiation of hematopoietic cells and bone marrow cells when used in
combination with other cytokines.
|0864) An albumin fusion protein of the invention and/or polynucleotide encoding an albumin fusion protein of the invention may also be
employed to maintain organs before transplantation or for supporting cell culture of primary tissues. An albumin fusion protein of the invention
and/or polynucleotide encoding an albumin fusion protein of the invention may also be employed for inducing tissue of mesodermal origin to
differentiate in early embryos.
[086S] An albumin fusion protein of the invention and/or polynucleotide encoding an albumin fusion protein of the invention may also increase
or decrease the differentiation or proliferation of embryonic stern cells, besides, as discussed above, hematopoietic lineage.
|0866| An albumin fusion protein of the invention and/or polynucleotide encoding an albumin fusion protein of the invention may also be used
to modulate mammalian characteristics, such as body height, weight, hair color, eye color, skin, percentage of adipose tissue, pigmentation, size, and
shape (e.g., cosmetic surgery). Similarly, an albumin fusion protein of the invention and/or polynucleotide encoding an albumin fusion protein of
the invention may be used to modulate mammalian metabolism affecting catabolism, anabolism, processing, utilization, and storage of energy.
[0867] An albumin fusion protein of the invention and/or polynucleotide, encoding an albumin fusion protein of the invention may be used to
change a mammal's mental state or physical state by influencing biorhythms, caricadic rhythms, depression (including depressive disorders),
tendency for violence, tolerance for pain, reproductive capabilities (preferably by Activin or mhibin-like activity), hormonal or endocrine levels,
appetite, libido, memory, stress, or other cognitive qualities.
|0868) An albumin fusion protein of the invention and/or polynucleotide encoding an albumin fusion protein of the invention may also be used
as a food additive or preservative, such as to increase or decrease storage capabilities, fat content, lipid, protein, carbohydrate, vitamins, minerals,
cofactors or other nutritional components.
|0869| The above-recited applications have uses in a wide variety of hosts. Such hosts include, but are not limited to, human, murine, rabbit,
goat, guinea pig, camel, horse, mouse, rat, hamster, pig, micro-pig, chicken, goat, cow, sheep, dog, cat, non-human primate, and human. In specific
embodiments, the host is a mouse, rabbit, goat, guinea pig, chicken, rat, hamster, pig, sheep, dog or cat. In preferred embodiments, the host is a
mammal. In most preferred embodiments, the host is a human.
|0870] Having generally described the invention, the same will be more readily understood by reference to the following examples, which are
provided byway of illustration and are not intended as limiting.
|087I| Without further description, it is believed that one of ordinary skill in the art can, using the preceding description and the following
illustrative examples, make and utilize the alterations detected in the present invention and practice the claimed methods. The following working
examples therefore, specifically point out preferred embodiments of the present invention, and are not to be construed as limiting in any way the
remainder of the disclosure.
EXAMPLES.
EXAMPLE]: Generation of pScNHSA and pScCHSA.
[0872] The vectors pScNHSA (ATCC Deposit No. PTA-3279) and pScCHSA (ATCC Deposit No. PTA-3276) arc derivatives of pPPCOOOS (ATCC Deposit No. PTA-3278) and are used as cloning vectors into which polynuclcotidcs encoding a therapeutic protein or fragment or variant thereof is inserted adjacent to and in translation frame with polynucleotides encoding human serum albumin "HSA". pScCHSA may be used for generating Therapeutic protein-IISA fusions, while pScNHSA may be used to generate HSA-Thcrapcutic protein fusions.
Generation ofpScCHSA: albumin fusion with the albumin moiety C-lerminal to the therapeutic portion.
|0873| A vector to facilitate cloning DNA encoding a Therapeutic protein N-terminal to DNA encoding the mature albumin protein was made by altering the nucleic acid sequence that encodes the chimeric HSA signal peptide in pPPCOOOS to include the Xho [ and Clti 1 restriction sites
|08;4|' " TTrst, 'me Mo \ and Cla I sites inherent to pPPC0005 (located 3' of the ADH1 terminator sequence) were eliminated by digesting pPPCOOOS with Xho I and Cla I, filling in the sticky ends with T4 DNA polymerase, and religating the blunt ends to create pPPC0006. |087S| Second, the Xho I and Cla I restriction sites were engineered into the nucleic acid sequence that encodes the signal peptide of HSA (a chimera of the HSA leader and a kex2 site from mating factor alpha, "MAF") in pPPC0006 using two rounds of PCR. In the first round of PCR, amplification with primers shown as SEQ ID NO:36 and SEQ ID NO:37 was performed. The primer whose sequence is shown as SEQ ID N0:36 comprises a nucleic acid sequence that encodes part of the signal peptide sequence of HSA, a kex2 site from the mating factor alpha leader sequence, and part of the amino-terminus of the mature form of HSA. Four point mutations were introduced in the sequence, creating the Xho I and Cla I sites found at the junction of the chimeric signal peptide and the mature form of HSA. These four mutations are underlined in the sequence shown below. In pPPCOOOS the nucleotides at these four positions from 5' to 3' are T, G, T, and 0.
i'-UCCrcGAGAAAAGAGATGCACACAAGA(}fGAGG'ITGCTCATCGATn'AAAGA'nTGGG-3' (SEQ ID NO:36) and 5^AATCGATGAGCAACCTCACTCrrGTGTGCATCTCTTrTCTCGAGGCTCCTGGAATAAGC-3' (SEQ ID NO:37). A second round of PCR was then performed with an upstream flanking primer, S'-TACAAACTTAAGAGTCCAA'ITAGC-S1 (SEQ ID NO:38) and a downstream flanking primer 5'-CACTTGTCTAGAGTGGTTTCATATGTCTT-3' (SEQ ID NO:39). The resulting PCR product was then purified and digested with Aft II and Xba \ and ligated into the same sites in pPPC0006 creating pScCHSA. The resulting plasmid has Xho I and Cla I sites engineered into the signal sequence. The presence of the Xho I site creates a single amino acid change in the end of the signal sequence from LDKR to LEKR. The D to E change will not be present in the final albumin fusion protein expression plasmid when a nucleic acid sequence comprising a polynuclcotide encoding the Therapeutic portion of the albumin fusion protein with a 5' Sal 1 site (which is compatible with the Xho I site) and a 3' Cla I site is ligated into the Xho I and Cla I sites of pScCHSA. Ligation of Sal I to Xho I restores the original amino acid sequence of the signal peptide sequence. DNA encoding the Therapeutic portion of the albumin fusion protein may be inserted after the Kex2 site (K.ex2 cleaves after the dibasic amino acid sequence KR at the end of the signal peptide) and prior to the Cla I site.
Generation oCpScNH.SA: albumin fusion with the albumin moiety N-terminal to the therapeutic portion.
|0876] A vector to facilitate cloning DNA encoding a Therapeutic protein portion C-terminal to DNA encoding the mature albumin protein, was made by adding three, eight-base-pair restriction sites to pScCHSA. The Asc I, Fsc I, and Pme I restriction sites were added in between the Bsu)6 I and Hind III sites at the end of the nucleic acid sequence encoding the mature HSA protein. This was accomplished through the use of two complementary synthetic primers containing the Asc I, Fse I, and Pine I restriction sites underlined (SEQ ID NO:40 and SEQ ID N0:41). S'-AAGCTGCC1TACGCrrATAATAAGGCGCGCCCGCCGGCCGTn-AAACTAAGCTTAATTCT-3' (SEQ ID N0:40) and 5-AnAAri'AAGCTTAGrrTAAACGGCCGGCCGGCGCGCCTTATTATAAGCCTAAGGCAGCTT-3' (SEQ ID NO:41). These primers were annealed and digested with Bsu36 I and Hind III and ligated into the same sites in pScCHSA creating pScNHSA.
EXAMPLE 2: General Construct Generation for Yeast Transformation.
|0877| The vectors pScNHSA and pScCHSA may be used as cloning vectors into which polynuclcotides encoding a therapeutic protein or fragment or variant thereof is inserted adjacent to polynucleotides encoding mature human serum albumin "HSA". pScCHSA is used for generating Therapeutic protein-HSA fusions, while pScNHSA may be used to generate HSA-Therapeutic protein fusions.
Generation uf albumin fusion constructs comprising HSA-Therapeiitic prolein fusion products.
(0878) DNA encoding a Therapeutic protein (e.g., sequences shown in SEQ ID NO:X or known in the art) may be PCR amplified using the primers which facilitate the generation of a fusion construct (e.g., by adding restriction sites, encoding seamless fusions, encoding linker sequences, etc.) For example, one skilled in the art could design a 5' primer that adds polynucleotides encoding the last four amino acids of the mature form of USA (and containing Ihe Biu36I site) onto the 5' end of DNA encoding a Therapeutic protein; and a 3' primer that adds a STOP codon and appropriate cloning sites onto the 3' end of the Therapeutic protein coding sequence. For instance, the forward primer used to amplify DNA encoding a Therapeutic protein might have the sequence, 5'-aagctGCCTTAGGCTTA(N),s-3' (SEQ ID N0:42) where the underlined sequence is a Bsu36\ site, the upper case nucleotides encode the last four amino acids of the mature HSA protein (ALGL), and (N),s is identical to the first 15 nucleotides encoding the Therapetic protein of interest. Similarly, the reverse primer used to amplify DNA encoding a Therapeutic protein might have the sequence, S'-GCGCGCCr/T/l/ Generation of albumin fusion constructs comprising ^ru'-rlSA fusion products.
|0880| Similar to the method described above, DNA encoding 3 Therapeutic protein may be PCR amplified using the following primers: A 5' primer that adds polynucleotides containing a Satl site and encoding the last three amino acids of the HSA leader sequence, DK.R, onto the 5' end of DNA encoding a Therapeutic protein; and a 3' primer that adds polynucleotides encoding the first few amino acids of the mature HSA containing a
Clti I site onto the 3' end of UNA encoding a Therapeutic protein. For instance, the forward primer used to amplify the DNA encoding a Therapeutic protein might have the sequence, 5'-aggagcgtcGACAAAAGA(N)ls-3' (SEQ ID NO:46) where the underlined sequence is a Sat I site, the upper case nucleotides encode the last three amino acids of the USA leader sequence (DKR), and (N)L, is identical to the first 15 nucleotides encoding the Thcrapetic protein of interest. Similarly, the reverse primer used to amplify the DNA encoding a Therapeutic protein might have the sequence, 5'-CTTTAA^ TCGATOAGCAACCTCACTCTTGTGTGCATCfN), Generation of albumin fusion construct compatible for expression in venal S. cerevisiae.
|0881| The Not I fragment containing the DNA encoding either an N-terminal or C-terminal albumin fusion protein generated from pScNHSA or pScCHSA may then be cloned into the Not I site of pSAC35 which has a LEU2 selectable marker. The resulting vector is then used in transformation of a yeast S. cerevisiae expression system.
EXAMPLE 3: General Expression in Yeast S. cerevisiae,
|0882| An expression vector compatible with yeast expression can be transformed into yeast S. cerevisiae by lithium acetate transformation, electroporation, or other methods known in the art and or as described in part in Sambrook, Fritsch, and Maniatis. 1989. "Molecular Cloning: A Laboratory Manual, 2nd edition", volumes 1-3, and in Ausubel et al. 2000. Massachusetts General Hospital and Harvard Medical School "Current Protocols in Molecular Biology", volumes 1-4. The expression vectors are introduced into S. cerevisiae strains DXY1, D88, or BXP10 by transformation, individual transformants can be grown, for example, for 3 days at 30°C in 10 mL YEPD (1% w/v yeast extract, 2 % w/v, peptone, 2 % w/v, dextrose), and cells can be collected at stationary phase after 60 hours of growth. Supernatants are collected by clarifying cells at 3000g for 10 minutes.
|0883| pSAC35 (Sleep et al., 1990, Biotechnology 8:42 and see Figure 3) comprises, in addition to the LEU2 selectable marker, the entire yeast 2 urn plasmid to provide replication functions, the PRBI promoter, and the ADH1 termination signal.
EXAMPLE 4: General Purification of an Albumin Fusion Protein Expressed from an Albumin Fusion in Yeast S. cerevisiae. |0884| In preferred embodiments, albumin fusion proteins of the invention comprise the mature form of HSA fused to either the N- or C-terminus of the mature form of a therapeutic protein or portions thereof (e.g., the mature form of a therapeutic protein listed in Table 1, or the mature form of a therapeutic protein shown in Table 2 as SEQ ID NO:Z). In one embodiment of the invention, albumin fusion proteins of the invention further comprise a signal sequence which directs the nascent fusion polypeptide in the secretory pathways of the host used for expression. In a preferred embodiment, the signal pcptide encoded by the signal sequence is removed, and the mature albumin fusion protein is secreted directly into the culture medium. Albumin fusion proteins of the invention preferably comprise heterologous signal sequences (e.g., the non-native signal sequence of a particular therapeutic protein) including, but not limited to, MAP, 1NV, Ig, Fibulin B, Clusterin, Insulin-Like Growth Factor Binding Protein 4, variant HSA leader sequences including, but not limited to, a chimeric HSA/MAF leader sequence, or other heterologous signal sequences known in the art. Especially preferred as those signal sequence listed in Table 2 and/or the signal sequence listed in the "Expression of Fusion Proteins" and/or "Additional Methods of Rccombinant and Synthetic Production of Albumin Fusion Proteins" section of the specification, above. In preferred embodiments, the fusion proteins of the invention further comprise an N-terminal methionine residue. Polynucleotides encoding these polypeptidcs, including fragments and/or variants, are also encompassed by the invention.
[0885] Albumin fusion proteins expressed in yeast as described above can be purified on a small-scale over a Dyax peptide affinity column as follows. Supernatants from yeast expressing an albumin fusion protein is diafiltrated against 3 mM phosphate buffer pH 6.2, 20 mM NaCI and 0.01% Tween 20 to reduce the volume and to remove the pigments. The solution is then filtered through a 0.22 urn device. The filtrate is loaded onto a Dyax peptide affinity column. The column is eluted with 100 mM Tris/HCI, pH 8.2 buffer. The peak fractions containing protein are collected and analyzed on SDS-PAGE after concentrating 5-fold.
|0886| For large scale purification, the following method can be utilized. The supernatant in excess of 2 L is diafiltered and concentrated to 500 rnL in 20 mM Tris/HCI pH 8.0. The concentrated protein solution is loaded onto a pre-equilibrated 50 mL DEAE-Sepharose Fast Flow column, the column is washed, and the protein is eluted with a linear gradient of NaCI from 0 to 0.4 M NaCI in 20 mM Tris/HCI, pH 8.0. Those fractions containing the protein are pooled, adjusted to pH 6.8 with 0.5 M sodium phosphate (NaHjPO,). A final concentration of 0.9 M (NH linear gradient from 0 to 0.5 M. The tractions containing the protein of interest are combined, the buffer is changed to 10 mM Na2HPO,/citric acid pH 6.25 with an Amicon concentrator, the conductivity is EXAMPLE 5: General Construct Generation for Mammalian Cell Transfection. Generation of albumin fusion construct compatible for expression in mammalian cell-lines.
[0887] Albumin fusion constructs can be generated in expression vectors for use in mammalian cell culture systems. DNA encoding a therapeutic protein can be cloned N-terminus or C-terminus to USA in a mammalian expression vector by standard methods known in the art (e.g., PCR amplification, restriction digestion, and ligation). Once the expression vector has been constructed, transfection into a mammalian expression system can proceed. Suitable vectors are known in the art including, but not limited to, for example, the pC4 vector, and/or vectors available from I-onza Biologies, Inc. (Portsmouth, NH).
(0888| The DNA encoding human serum albumin has been cloned into the pC4 vector which is suitable for mammalian culture systems, creating plasmid pC4:HSA (ATCC Deposit # PTA-3277). This vector has a DiHydroFolatc Rcductasc, "DHFR", gene that will allow for selection in the presence of methotrexate.
|0889] The pC4:HSA vector is suitable for expression of albumin fusion proteins in CHO cells. For expression, in other mammalian cell culture systems, it may be desirable to subclone a fragment comprising, or alternatively consisting of, DNA which encodes for an albumin fusion protein into an alternative expression vector. For example, a fragment comprising, or alternatively consisting, of DNA which encodes for a mature albumin fusion protein may be subcloned into another expression vector including, but not limited to, any of the mammalian expression vectors described herein.
[0890] In a preferred embodiment, DNA encoding an albumin fusion construct is subcloned into vectors provided by Lonza Biologies, Inc. (Portsmouth, NH) by procedures known in the art for expression in NSO cells.
Generation of albumin fusion constructs comprising USA-Therapeutic Protein fusion products.
[0891 ] Using pC4:HSA (ATCC Deposit # PTA-3277), albumin fusion constructs can be generated in which the Therapeutic protein portion is C terminal to the mature albumin sequence. For example, one can clone DNA encoding a Therapeutic protein of fragment or variant thereof between the Bsu 361 and Asc I restriction sites of the vector. When cloning into the Bsu 361 and Asc I, the same primer design used to clone into the yeast vector system (SEQ ID NO:42 and 43) may be employed (see Example 2).
Generation of albumin fusion constructs comprising gene-USA fusion products. (
|()892| Using pC4:HSA (ATCC Deposit # PTA-3277), albumin fusion constructs can be generated in which a Therapeutic protein portion is cloned N terminal to the mature albumin sequence. For example, one can clone DNA encoding a Therapeutic protein that has its own signal sequence between the Bam HI (or Hind HI) and Cla I sites of pC4:HSA. When cloning into either the Bam HI or Hind III site, it is prefcrrablc to include a Kozak sequence (CCGCCACCATG, SEQ ID NO:49) prior to the translational start codon of the DNA encoding the Therapeutic protein. If a Therapeutic protein does not have a signal sequence, DNA encoding that Therapeutic protein may be cloned in between the Xlio I and Cla I sites ofpC4.HSA. When using the X)w I site, the following 5' (SEQ ID NO:50) and 3' (SEQ ID NO:51) exemplary PCR primers may be used: S'-CCGCCGCTCGAGGGGTGTGTTTCCTCGA(N)i.-3' (SEQ ID NO: 50) 5l-AGTCCCATCGATGAGCAACCTCACTCrrGTG'raCATC(N)i.-3' (SEQ ID NO:51)
[0893] In the 5' primer (SEQ ID N0:50), the underlined sequence is a Xho I site; and the Xlio I site and the DNA following the Xho I site code for the last seven amino acids of the leader sequence of natural human serum albumin. In SEQ ID NO:50, "(N),B" is DNA identical to the first 18 nuclcotides encoding the Therapeutic protein of interest. In the 3' primer (SEQ ID N0:51), the underlined sequence is a Cla I site; and the Cla I site and the DNA following it are the reverse complement of the DNA encoding the first 10 amino acids of the mature USA protein (SEQ ID N0:l). In SEQ ID NO:5I "(N)i«" is the reverse complement of DNA encoding the last 18 nucleotides encoding the Therapeutic protein of interest. Using these two primers, one may PCR amplify the Therapeutic protein of interest, purify the PCR product, digest it with Xho I and Cla I restriction enzymes and clone it into the Xho I and Cla 1 sites in the pC4:USA vector.
|0894| If an alternative leader sequence is desired, the native albumin leader sequence can be replaced with the chimeric albumin leader, i.e., the HSA-kex2 signal pcptide, or an alternative leader by standard methods known in the art. (For example, one skilled in the art could routinely PCR amplify an alternate leader and sub clone the PCR product into an albumin fusion construct in place of the albumin leader while maintaining the reading frame).
EXAMPLE 6: General Expression in Mammalian Cell-Lines.
[0895] An albumin fusion construct generated in an expression vector compatible with expression in mammalian cell-lines can be transacted into appropriate cell-lines by calcium phosphate precipitation, lipofectamine, clcctroporation, or other transfect ion methods known in the art and/or as described in Sambrook, Fritsch, and Maniatis. 1989. "Molecular Cloning: A Laboratory Manual, 21"1 edition" and in Ausubel et al. 2000. Massachusetts General Hospital and Harvard Medical School "Current Protocols in Molecular Biology", volumes 1-4. "Hie transfected cells are then
selected lor Dy tne presence 01 a seiixung agent determined by the selectable marker in the expression vector.
(0896| The pC4 expression vector (ATCC Accession No. 209646) is a derivative of the plasmid pSV2-DHFR (ATCC Accession No. 37146).
pC4 contains the strong promoter Ixmg Terminal Repeats "LTR" of the Rous Sarcoma Virus (Cullen et al., March 1985, Molecular and Cellular
Biology, 438-447) and a fragment of the CytoMegaloVirus "CMV'-enhancer (Boshart et al., 1985, Cell 41: 521-530). The vector also contains the
3' intron, (lie polyadenylation and termination signal of the rat preproinsulin gene, and the mouse DHFR gene under control of the SV40 early
promoler. Chinese hamster ovary "CHO" cells or other cell-lines lacking an active DHFR gene are used for transfcction. Transfection of an
albumin fusion construct in pC4 into CHO cells by methods known in the art will allow for the expression of the albumin fusion protein in CHO
cells, followed by leader sequence cleavage, and secretion into the supernatant. The albumin fusion protein is then further purified from the
supernatant.
[0897] The pEEI2.1 expression vector is provided by Lonza Biologies, Inc. (Portsmouth, NH) and is a derivative of pEE6 (Stephens and
Cockctt, 1989, Nucl. Acids Res. 17: 7110). This vector Comprises a promoter, enhancer and complete 5'-untranslated region of the Major
Immediate Early gene of the human CytoMegaloVirus, "hCMV-MIE" (International Publication # WO89/01036), upstream of a sequence of
interest, and a Glutamine Synthetase gene (Murphy et al., 1991, Uiochem J. 227: 277-279; Bebbington et al., 1992, Bio/Technology 10:169-175;
US patent US 5,122,464) for purposes of selection of transfected cells in selective methionine sulphoximine containing medium. Transfection of
albumin fusion constructs made in pEE12.1 into NSO cells (International Publication # WO86/05807) by methods known in the art will allow for the
expression of the albumin fusion protein in NSO cells, followed by leader sequence cleavage, and secretion into the supernatant. The albumin fusion
protein is then further purified from the supernatant using techniques described herein or otherwise known in the art.
[0898] Expression uf an albumin fusion protein may be analyzed, for example, by SDS-PAGE and Western blot, reversed phase HPLC analysis,
or other methods known in the art.
|0899| Stable CHO and NSO cell-lines transfected with albumin fusion constructs are generated by methods known in the art (e.g.,
lipofectamine transfection) and selected, for example, with 100 nM methotrexate for vectors having the DiHydroFolate Reductase 'DHFR' gene as a
selectable marker or through growth in the absence of glutamine. Expression levels can be examined for example, by immunoblotting, primarily,
with an anti-HSA senirn as the primary antibody, or, secondarily, with serum containing antibodies directed to the Therapeutic protein portion of a
given albumin fusion protein as the primary antibody.
[0900| Expression levels are examined by immunoblot detection with anti-HSA serum as the primary antibody. The specific productivity rales
are determined via ELISA in which the capture antibody can be a monoclonal antibody towards the therapeutic protein portion of the albumin fusion
and the detecting antibody can be the monoclonal anti-HSA-biotinylated antibody (or vice versa), followed by horseradish peroxidase/streptavidin
binding and analysis according to the manufacturer's protocol.
EXAMPLE 7: Kxpression of an Albumin fusion Protein In Mammalian Cells.
|0901| The albumin fusion proteins of the present invention can be expressed in a mammalian cell. A typical mammalian expression vector contains a promoter element, which mediates the initiation of transcription of mRNA, a protein coding sequence, and signals required for the termination of transcription and polyadenylation of the transcript Additional elements include enhancers, Kozak sequences and intervening sequences Hanked by donor and acceptor sites for KNA splicing. Highly efficient transcription is achieved with the early and late promoters from SV40, the long terminal repeals (LTRs) from Rctroviruses, e.g., RSV, HTLVI, II1VI and the early promoter of the cytomcgalovirus (CMV). However, cellular elements can also be used (e.g., the human actin promoter).
|0902| Suitable expression vectors for use in practicing the present invention include, for example, vectors such as, pSVL and pMSG (Pharmacia, Uppsala, Sweden), pRSVcat (ATCC 37152), pSV2dhfr (ATCC 37146), pBCI2MI (ATCC 67109), pCMVSport 2.0, and pCMVSport 3.0. Mammalian host cells that could be used include, but are not limited to, human Hela, 293, H9 and Jurkat cells, mouse NIH3T3 and C127 cells, Cos 1, Cos 7 and CV1, quail QC1-3 cells, mouse L cells and Chinese hamster ovary (CHO) cells.
|0903| Alternatively, the albumin fusion protein can be expressed in slable cell lines containing the polynucleotide encoding the albumin fusion protein integrated into a chromosome. The co-transfection with a selectable marker such as IDHFR, gpt, neomycin, or hygromycin allows the identification and isolation of the transfected cells.
|0904] The transfected polynucleotide encoding the fusion protein can also be amplified to express large amounts of the encoded fusion protein. The DHFR (dihydrofolate reduclase) marker is useful in developing cell lines that carry several hundred or even several thousand copies of the gene of interest. (See, e.g., Alt et al., J. Biol. Chem. 253:1357-1370 (1978), Hamlinetal., Diochem. et Biophys. Acta, 1097:107-143 (1990); Page et al., Biotechnology 9:64-68 (1991)). Another useful selection marker is the enzyme glutamine synthase (GS) (Murphy et al., Biochem J. 227:277-279 (1991); Bebbington el al., Bio/Technology 10:169-175 (1992). Using these markers, the mammalian cells are grown in selective medium and the cells with the highest resistance are selected. These cell lines contain the amplified gene(s) integrated into a chromosome. Chinese hamster ovary (CHO) and NSO cells are often used for the production of proteins.
|0905| Derivatives of the plasmid pSV2-dhfr (ATCC Accession No. 37146), the expression vectors pC4 (ATCC Accession No. 209646) and pC6 (ATCC Accession No 209647) contain the strong promoter (LTR) of the Rous Sarcoma Virus (Cullen et al., Molecular and Cellular Biology,
438-447 (March, 1985)) plus a tragment of the CMV-enhanccr (Boshart et al., Cell 41:521-530 (1985)). Multiple cloning sites, e.g., with the
restriction enzyme cleavage sites BarnHI, Xbal and Asp718, facilitate the cloning of the gene of interest. The vectors also contain the 3' intron, the
polyadenylatioii and termination signal of the rat preproinsulin gene, and the mouse DHFR gene under control of the SV40 early promoter.
[0906] Specifically, the plasmid pC6, for example, is digested with appropriate restriction enzymes and then dephosphorylated using calf
intestinal phosphates by procedures known in the art. The vector is then isolated from a 1% agarose gel.
|0907] A polynucleotide encoding an albumin fusion protein of the present invention is generated using techniques known in the art and this
polyrmcleotide is amplified using PCR technology known in the art. If a naturally occurring signal sequence is used to produce the fusion protein of
the present invention, the vector does not need a second signal peptide. Alternatively, if a naturally occurring signal sequence is not used, the vector
can be modified to include a heterologous signal sequence. (See, e.g., International Publication No. WO 96/34891.)
|0908| The amplified fragment encoding the fusion protein of the invention is isolated from a 1% agarose gel using a commercially available kit
("Geneclean," BIO 101 Inc., La Jolla, Ca.). The fragment then is digested with appropriate restriction enzymes and again purified on a 1% agarose
gel-
|0909| The amplified fragment encoding the albumin fusion protein of the invention is then digested with the same restriction enzyme and
purified on a 1% agarose gel. The isolated fragment and the dephosphorylated vector are then ligated with T4 DNA ligase. E. coli HB101 or XL-I
Blue cells are then transformed and bacteria are identified that contain the fragment inserted into plasmid pC6 using, for instance, restriction enzyme
analysis.
J0910] Chinese hamster ovary cells lacking an active DHFR gene is used for transfection. Five /jg of the expression plasmid pC6 or pC4 is
cotransfected with 0.5 jig of the plasmid pSVnco using lipofcctin (Feigner et al., supra). The plasmid pSV2-nco contains a dominant selectable
marker, the nco gene from Tn5 encoding an enzyme that confers resistance to a group of antibiotics including G4I8. The cells are seeded in alpha
minus MEM supplemented with 1 mg/ml G418. After 2 days, the cells are trypsinized and seeded in hybridoma cloning plates (Greiner, Germany)
in alpha minus MEM supplemented with 10, 25, or 50 ng/ml of methotrexate plus 1 mg/ml G418. After about 10-14 days single clones are
trypsinized and then seeded in 6-well pctri dishes or 10 ml flasks using different concentrations of methotrexate (50 nM, 100 nM, 200 nM, 400 nM,
800 nM). Clones growing at the highest concentrations of methotrexate are then transferred to new 6-well plates containing even higher
concentrations of methotrexate (1 /iM, 2 /xM, 5 jiM, 10 mM, 20 mM). The same procedure is repeated until clones are obtained which grow at a
concentration of 100 - 200 nM. Expression of the desired fusion protein is analyzed, for instance, by SDS-PAGE and Western blot or by reversed
phase HPLC analysis.
EXAM PI K X: General Purification of an Albumin Fusion Protein Expressed from an Albumin Fusion Construct in Mammalian
Cell-lines.
|09111 In preferred embodiments, albumin fusion proteins of the invention comprise the mature form of HSA fused to either the N- or C-terminus of the mature form of a therapeutic protein or portions thereof (e.g., the mature form of a therapeutic protein listed in Table 1, or the mature form of a therapeutic protein shown in Table 2 as SKQ ID NO:Z). In one embodiment of the invention, albumin fusion proteins of the invention further comprise a signal sequence which directs the nascent fusion polypeptide in the secretory pathways of the host used for expression. In a preferred embodiment, the signal peptide encoded by the signal sequence is removed, and the mature albumin fusion protein is secreted directly into the culture medium Albumin fusion proteins of the invention preferably comprise heterologous signal sequences (e.g., the non-native signal sequence of a particular therapeutic protein) including, but not limited to, MAP, INV, Ig, Fibulin B, Clusterin, Insulin-Like Growth Factor Binding Protein 4, variant HSA leader sequences including, but not limited to, a chimeric HSA/MAF leader sequence, or other heterologous signal sequences known in the art. Especially preferred as those signal sequence listed in Table 2 and/or the signal sequence listed in the "Expression of Fusion Proteins" and/or "Additional Methods of Recombinant and Synthetic Production of Albumin Fusion Proteins" section of the specification, above. In preferred embodiments, the fusion proteins of the invention further comprise an N-terminal methionine residue. Polynuclcotides encoding these polypeptides, including fragments and/or variants, are also encompassed by the invention.
[0912| Albumin fusion proteins from mammalian cell-line supematants are purified according to different protocols depending on the expression system used.
Purification from CHO and 293T cell-lines.
|0913| Purification of an albumin fusion protein from CHO cell supernatant or from transiently transfectcd 293T cell supernatant may involve initial capture with an anionic HQ resin using a sodium phosphate buffer and a phosphate gradient elution, followed by affinity chromatography on a Blue Sepharose FF column using a salt gradient elution. Blue Sepharose FF removes the main BSA/fetuin contaminants. Further purification over the Poros PI 50 resin with a phosphate gradient may remove and lower endotoxin contamination as well as concentrate the albumin fusion protein.
Purification from NSQ cell-line.
(0914\ Purification of an albumin-fusion protein from NSO cell supernatant may involve Q-Sepharose anion exchange chromatography, followed by SP-sepharose purification with a step elution, followed by Phenyl-650M purification with a step elution, and, ultimately, diafiltration. |09I 51 The purified protein may then be formulated by buffer exchange.
EXAMPLE 9: Bacterial Expression of an Albumin Fusion Protein.
|0916| A polynucleotide encoding an albumin fusion protein of the present invention comprising a bacterial signal sequence is amplified using PCR oligonucleotide primers corresponding to the 5' and 3' ends of the DNA sequence, to synthesize insertion fragments. The primers used to amplify the polynucleotide encoding insert should preferably contain restriction sites, such as BamHI and Xbal, at the 5' end of the primers in order to clone the amplified product into the expression vector. For example, BamHI and Xbal correspond to the restriction enzyme sites on the bacterial expression vector pQE-9. (Qiagen, Inc., Chatsworth, CA). This plasmid vector encodes antibiotic resistance (Ampr), a bacterial origin of replication (ori), an IPTG-regulatable promoter/operator (P/O). a ribosome binding site (RBS), a 6-histidine tag (6-His), and restriction enzyme cloning sites. (0917] The pQE-9 vector is digested with BamHI and Xbal and the amplified fragment is ligated into the pQE-9 vector maintaining the reading frame initiated at the bacterial RDS. The ligation mixture is then used to transform the E. coli strain MI5/rep4 (Qiagen, Inc.) which contains
multiple copies of the plasmid pREP4, which expresses the lad represser and also confers kanamycin resistance (Kanr). Transforrnants are
identified by their ability to grow on LB plates and ampicillin/kanamycin resistant colonies are selected. Plasmid DNA is isolated and confirmed by
restriction analysis.
[0918] Clones containing the desired constructs are grown overnight (O/N) in liquid culture in LB media supplemented with both Amp (100
ug/ml) and Kan (25 ug/ml). The O/N culture is used to inoculate a large culture at a ratio of 1:100 to 1:250. The cells are grown to an optical
density 600 (O.D.*00) of between 0.4 and 0.6. IPTG (Isopropyl-B-D-thiogalacto pyrarioside) is then added to a final concentration of 1 mM. 1PTG
induces by inactivating the lad repressor, clearing the P/O leading to increased gene expression.
[0919] Cells are grown for an extra 3 to 4 hours. Cells are then harvested by centrifugation (20 mins at 6000Xg). The cell pellet is solubilized
in the chaotropic agent 6 Molar Guanidine HC1 or preferably in 8 M urea and concentrations greater than 0.14 M 2-mercaptoethanol by stirring for
3-4 hours at 4°C (see, e.g., Burton ct al., Eur. J. Biochem. /7P/379-387 (1989)). The cell debris is removed by centrifugation, and the supernatant
containing the polypcptide is loaded onto a m'ckel-nitrilo-tri-acetic acid ("Ni-NTA") affinity resin column (available from QIAGEN, Inc., supra).
Proteins with a 6 x His tag bind to the Ni-NTA resin with high affinity and can be purified in a simple one-step procedure (for details see: The
QIAexpressionist (1995) QIAGEN, Inc., supra).
[0920] Briefly, the supernatant is loaded onto the column in 6 M guanidine-HCI, pH 8. The column is first washed with 10 volumes of 6 M
guanidinc-HCI, pH 8, then washed with 10 volumes of 6 M guanidine-HCI pH 6, and finally the polypcptide is eluted with 6 M guanidine-HCI, pH
5.
|09211 The purified protein is then renaturcd by dialyzing it against phosphate-buffered saline (PBS) or 50 mM Na-acelale, pH 6 buffer plus
200 mM NaCI. Alternatively, the protein can be successfully refolded while immobilized on the Ni-NTA column Exemplary conditions are as
follows: renature using a linear 6M-1M urea gradient in 500 mM NaCI, 20% glycerol, 20 mM Tris/HCI pH 7.4, containing protease inhibitors. The
rcnaturation should he performed over a period of 1.5 hours or more. After rcnaturation the proteins are eluted by the addition of 250 mM
immidazole. Immidazolc is removed by a final dialyzing step against PBS or 50 mM sodium acetate pH 6 buffer plus 200 mM NaCI. The purified
protein is stored at 4°C or frozen at -80° C.
|0922| In addition to the above expression vector, the present invention further includes an expression vector, called pHE4a (ATCC Accession
Number 209645, deposited on February 25, 1998) which contains phage operator and promoter elements operatively linked to a polynucleotide
encoding an albumin fusion protein of the present invention, called pHE4a. (ATCC Accession Number 209645, deposited on February 25, 1998.)
This vector contains: 1) a neomycmphosphotransferase gene as a selection marker, 2) an E. coli origin of replication, 3) a T5 phage promoter
sequence, 4) two lac operator sequences, 5) a Shine-Delgamo sequence, and 6) the lactose operon represser gene (laclq). The origin of replication
(oriC) is derived from pUC19 (LTI, Gaithersburg, MD). The promoter and operator sequences are made synthetically.
[0923] DNA can be inserted into the pHE4a by restricting the vector with Ndcl and Xbal, BamHI, Xhol, or Asp718, running the restricted
product on a gel, and isolating the larger fragment (the stuffer fragment should be about 310 base pairs). The DNA insert is generated according to
PCR protocols described herein or otherwise known in the art, using PCR primers having restriction sites for Ndcl (5' primer) and Xbal, BamHI,
Xhol, or Asp718 (3' primer). The PCR insert is gel purified and restricted with compatible enzymes. The insert and vector are ligated according to
standard protocols.
10924] The engineered vector may be substituted in the above protocol to express protein in a bacterial system.
EXAMPLE 10: Isolation of a Selected cDNA Clone From the Deposited Sample.
[092S| Many of the albumin fusion constructs of the invention have been deposited with the ATCC as shown in Table 3. The albumin fusion constructs may comprise any one of the following expression vectors: the yeast S. cerevisiae expression vector pSAC35, the mammalian expression vector pC4, or the mammalian expression vector pEE12.1.
|0926] pSAC35 (Sleep ei al., 1990, Biotechnology 8:42), pC4 (ATCC Accession No. 209646, Cullen ct a!.. Molecular and Cellular Biology, 438-447 (1985); Boshart el al. Cell 41: 521-530 (1985)), and pEE12.1 (I-onza Biologies, Inc.; Stephens and Cocketl, Nucl. Acids Res. 17: 7110 (1989); International Publication SW089/01036; Murphy cl al.. Biochem J. 227: 277-279 (1991); Behbington el al.. Biotechnology 10:169-175
(1992); US patent US 5,122,464; International Publication #WO86/05807) vectors comprise an ampicillin resistance gene for growth in bacterial
cells. These vectors and/or an albumin fusion construct comprising them can be transformed into an E. coli strain such as Stratagene XL-1 Blue
(Stratagene Cloning Systems, Inc , 11011 N. Torrey Pines Road, I.a Jolla, CA, 92037) using techniques described in the art such as Hanahan, spread
onto Luna-Broth agar plates containing 100 ug/rnL ampicillin, and grown overnight at 37 °C.
[0927] The deposited material in the sample assigned the ATCC Deposit Number cited in Table 3 for any given albumin fusion construct also
may contain one or more additional albumin fusion constructs, each encoding different albumin fusion proteins. Thus, deposits sharing the same
ATCC Deposit Number contain at least an albumin fusion construct identified in the corresponding row of Table 3.
[0928] Two approaches can be used to isolate a particular albumin fusion construct from the deposited sample of plasmid DNAs cited for that
albumin fusion construct in Table 3.
Mrthad I: Screening
|0929] First, an albumin fusion construct may be directly isolated by screening the sample of deposited plasmid DNAs using a polynucleotide probe corresponding to SEQ ID NO:X for an individual construct ID number in Table 1, using methods known in the art. For example, a specific polynucleotide with 30-40 nucleotides may be synthesized using an Applied Biosystems DNA synthesizer according to the sequence reported. The oligonucleotide can be labeled, for instance, with J2P-y-ATP using T4 polynucleotide kinase and purified according to routine methods. (E.g., Maniatis et al., Molecular Cloning: A Laboratory Manual, Cold Spring Harbor Press, Cold Spring, NY (1982)). The albumin fusion construct from a given ATCC deposit is transformed into a suitable host, as indicated above (such as XL-1 Blue (Stratagene)) using techniques known to those of skill in the art, such as those provided by the vector supplier or in related publications or patents cited above. The transformants are plated on 1.5% agar plates (containing the appropriate selection agent, e.g., ampicillin) to a density of about 150 transformants (colonies) per plate. These plates are screened using Nylon membranes according to routine methods for bacterial colony screening (e.g., Sambrook et al., Molecular Cloning: A Laboratory Manual, 2nd Edit., (1989), Cold Spring Harbor Laboratory Press, pages 1.93 to 1.104), or other techniques known to those of skill in the art.
Method 2: PCR
[0930] Alternatively, DNA encoding a given albumin fusion protein may be amplified from a sample of a deposited albumin fusion construct with SEQ ID NO:X, for example, by using two primers of 17-20 nucleotides that hybridize to the deposited albumin fusion construct 5' and 3' to the DNA encoding a given albumin fusion protein. The polymerase chain reaction is carried out under routine conditions, for instance, in 25 /il of reaction mixture with 0.5 ug of the above cDNA template. A convenient reaction mixture is 1.5-5 mM MgCI2, 0.01% (w/v) gelatin, 20 ^M each of dATP, dCTP, dGTP, dTTP, 25 pmol of each primer and 0.25 Unit of Taq polymerase. Thirty five cycles of PCR (denaturation at 94°C for I min; annealing at 55°C for 1 min; elongation at 72°C for 1 min) are performed with a Perkin-Elmcr Cetus automated thermal cycler. The amplified product is analyzed by agarose gel clectrophoresis and the DNA band with expected molecular weight is excised and purified. The PCR product is verified to be the selected sequence by subcloning and sequencing the DNA product.
[0931| Several methods are available for the identification of the 5' or 3' non-coding portions of a gene which may not be present in the deposited clone. These methods include but arc not limited to, filter probing, clone enrichment using specific probes, and protocols similar or identical to 5' and 3' "RACE" protocols which are known in the art. For instance, a method similar to 5' RACE is available for generating the missing 5'end of a desired full-length transcript. (Fremont-Racine et al., Nucleic Acids Res., 21(7): 1683-1684 (1993)).
|0932| Briefly, a specific RNA oligonucleotide is ligated to the 5' ends of a population of RNA presumably containing full-length gene RNA transcripts. A primer set containing a primer specific to the ligated RNA oligonucleotide and a primer specific to a known sequence of the gene of interest is used to PCR amplify the 5' portion of the desired full-length gene. This amplified product may then be sequcnced and used to generate the full length gene.
|0933| This above method starts with total RNA isolated from the desired source, although poly-A+ RNA can be used. The RNA preparation can then be treated with phosphatasc if necessary to eliminate 5' phosphate groups on degraded or damaged RNA which may interfere with the later RNA ligase step. The phosphatase should then be inactivated and the RNA treated with tobacco acid pyrophosphatase in order to remove the cap structure present at the 5' ends of messenger RNAs. This reaction leaves a 5' phosphate group at the 5' end of the cap cleaved RNA which can then he ligated to an RNA oligonucleotide using T4 RNA ligase.
[0934| This modified RNA preparation is used as a template for first strand cDNA synthesis using a gene specific oligonucleotide. The first strand synthesis reaction is used as a template for PCR amplification of the desired 5' end using a primer specific to the ligated RNA oligonucleolide and a primer specific to the known sequence ofthc gene of interest. The resultant product is then sequenccd and analyzed to confirm that the 5' end sequence belongs to the desired gene.
EXAMPLE 11: Multifusion Fusions.
[0935] The albumin fusion proteins (e.g.. containing a Therapeutic protein (or fragment or variant thereof) fused to albumin (or a fragment or variant thereof)) may additionally be fused to other proteins to generate "multifusion proteins". These multifusion proteins can be used for a variety
of applications For example, fusion of the albumin fusion proteins of the invention to His-tag, HA-tag, protein A, IgG domains, and maltose
binding protein facilitates puntication. (see e.g.. EP A 394,827; Traunecker et al., Nature 331:84-86 (1988)). Nuclear localization signals fused to
the polypeptides of the present invention can target the protein to a specific subcellular localization, while covalent heterodimer or homodimers can
increase or decrease the nrtivity of an albumin fusion protein. Furthermore, the fusion of additional protein sequences to the albumin fusion
proteins of the invention may further increase the solubility and/or stability of the fusion protein. The fusion proteins described above can he made
using or routinely modifting techniques known in the art and/or by modifying the following protocol, which outlines the fusion of a polypeptide to an
IgG molecule.
(0936] Briefly, the human Fc portion of the IgG molecule can be PCR amplified, using primers that span the 5' and 3' ends of the sequence
described below. These primers also should have convenient restriction enzyme sites that will facilitate cloning into an expression vector, preferably
a mammalian or yeast expression vector.
|0937] For example, if pC4 (ATCC Accession No. 209646) is used, the human Fc portion can be ligated into the BamHI cloning site. Note that
the 3' BamHI site should be destroyed. Next, the vector containing the human Fc portion is re-restricted with BamHI, linearizing the vector, and a
polynucleotide encoding an albumin fusion protein of the present invention (generateed and isolated using techniques known in the art), is ligated
into this BamMl site. Note that the polynucleotide encoding the fusion protein of the invention is cloned without a stop codon, otherwise a Fc
containing fusion protein will not be produced.
[0938] If the naturally occurring signal sequence is used to produce the albumin fusion protein of the present invention, pC4 does not need a
second signal peptide. Alternatively, if the naturally occurring signal sequence is not used, the vector can be modified to include a heterologuus
signal sequence. (See, e.g., International Publication No. WO 96/34891.)
|0939] Human IgG Fc region:
GGGATCCGGAGCCCAAATC'ITCTGACAAAACTCACACATGCCCACCGTGCCCAGCACCTGAATTCGAGGGTGCACCGTCAGTCTI'CC
CTGAGGTCAAGTTCAACTGGTACGTGGACGGCGTGGAGGTGCATAATGCCAAGACAAAGCCGCGGGAGGAGCAGTACAACAGCACG CCAACCCCCATCGAGAAAACCATCTCCAAAGCCAAAGGGCAGCCCCGAGAACCACAGGTGTACACCCTGCCCCCATCCCGGGATGA AGCCGGAGAACAACTACAAGACCACGCCTCCCGTGCTGGACTCCGACGGCTCCITCTTCCTCTACAGCAAGCTCACCGTGGACAAGA
CTCCGGGTAAATGAGTGCGACGGCCGCGACTCTAGAGGAT (SEQ ID NO:52)
EXAMPLE 12: Production of an Antibody frvm an Albumin Fusion Protein.
Hvbridoma Technology
(0940] Antibodies that bind the albumin fusion proteins of the present invention and portions of the albumin fusion proteins of the present invention (e.g., the Therapeutic protein portion or albumin portion of the fusion protein) can be prepared by a variety of methods. (See, Current Protocols, Chapter 2.) As one example of such methods, a preparation of an albumin fusion protein of the invention or a portion of an albumin fusion protein of the invention is prepared and purified to render it substantially free of natural contaminants. Such a preparation is then introduced into an animal in order to produce polyclonal antisera of greater specific activity.
|0941| Monoclonal antibodies specific for an albumin fusion protein of the invention, or a portion of an albumin fusion protein of the invention, are prepared using hybridoma technology (Kohler et al.. Nature 256:495 (1975); Kohler ct al., Eur. J. Immunol. 6:511 (1976); Kohleret al., Hur. J. Immunol. 6:292 (1976); Hammerling et al., in: Monoclonal Antibodies and T-Cell Hybridomas, Elsevier, N.Y., pp. 563-681 (1981)). In general, an animal (preferably a mouse) is immunized with an albumin fusion protein of the invention, or a portion of an albumin fusion protein of the invention. The splcnocytes of such mice arc extracted and fused with a suitable myeloma cell line. Any suitable myeloma cell line may be employed in accordance with the present invention; however, it is preferable to employ the parent myeloma cell line (SP2O), available from the ATCC. After fusion, the resulting hybridoma cells arc selectively maintained in HAT medium, and then cloned by limiting dilution as described by Wands ct al. (Gastroenterology 80:225-232 (1981)). The hybridoma cells obtained through such a selection are then assayed to identify clones which secrete antibodies capable of binding an albumin fusion protein of the invention, or a portion of an albumin fusion protein of the invention. |0942| Alternatively, additional antibodies capable of binding to an albumin fusion protein of the invention, or a portion of an albumin fusion protein of the invention can be produced in a two-step procedure using anti-idiotypic antibodies. Such a method makes use of the fact that antibodies are themselves antigens, and therefore, it is possible to obtain an antibody which binds to a second antibody. In accordance with this meihod, protein specific antibodies are used to immunize an animal, preferably a mouse. The splenocytes of such an animal are then used to produce hybridoma cells, and the hybridoma cells are screened to identify clones which produce an antibody whose ability to bind to the an albumin fusion protein of the invention (or portion of an albumin fusion protein of the invention) -specific antibody can be blocked by the fusion protein of ihe invention, or a portion of an albumin fusion protein of the invention. Such antibodies comprise anti-idiotypic antibodies to the fusion protein of the invention (or portion of an albumin fusion protein of the invention) -specific antibody and are used to immunize an animal to induce formation
ol Hirther fusion protein of the invention (or portion of an albumin fusion protein of (he invention) -specific antibodies.
|0943| For in vivo use of antibodies in humans, an antibody is "humanized". Such antibodies can be produced using genetic constructs derived from hybridoma cells producing the monoclonal antibodies described above. Methods for producing chimeric and humanized antibodies are known in the art and are discussed herein. (Sec, for review, Morrison, Science 229:1202 (1985);Oi et al., BioTechniques 4:214(1986); Cabilly et al., U.S. Patent No. 4,816,567; Taniguchi et al, EP 1714%; Morrison et al.. EP 173494; Neuberger et al., WO 8601533; Robinson ct al., International Publication No. WO 8702671; Boulianne ct al., Nature 312:643 (1984); Neuberger et al,, Nature 314:268 (1985)).
[0944] Isolation Of Antibody Fragments Directed Against an albumin fusion protein of the invention, or a portion of an albumin fusion protein of the invention From A Library Of scFvs. Naturally occurring V-genes isolated from human P3Ls are constructed into a library of antibody fragments which contain reactivities against an albumin fusion protein of the invention, or a portion of an albumin fusion protein of the invention, to which the donor mayor may not have been exposed (see e.g., U.S. Patent 5,885,793 incorporated herein by reference in its entirety). [0945) Rescue of the Library. A library of scFvs is constructed from the RNA of human PBLs as described in International Publication No. WO 92/01047. To rescue phage displaying antibody fragments, approximately 10' E. coli harboring the phagemid are used to inoculate 50 ml of 2xTY containing 1% glucose and 100 /Jg/ml of ampicillin (2xTY-AMP-GLU) and grown to an O.D. of 0.8 with shaking. Five ml of this culture is i used to inoculate 50 ml of 2xTY-AMI'-GLU, 2 x 108 TU of delta gene 3 helper (M13 delta gene III, see International Publication No. WO 92/01047) are added and the culture incubated at 37°C for 45 minutes without shaking and then at 37°C for 45 minutes with shaking. The culture is ccnlrifnged at 4000 r.p.m. for 10 min. and the pellet resuspcnded in 2 liters of 2xTY containing 100 ^g/mi ampicillin and 50 ug/ml kanamycin and grown overnight. I'hage are prepared as described in International Publication No. WO 92/01047.
|0946] M13 delta gene 111 is prepared as follows: M13 delta gene III helper phage docs not encode gene 111 protein, hence the phage(mtd) displaying antibody fragments have a greater avidity of binding to antigen. Infectious M13 delta gene [[I particles are made by growing the helper phage in cells harboring a pUCl9 derivative supplying the wild type gene III protein during phage morphogenesis. The culture is incubated for I hour at 37° C without shaking and then for a further hour at 37°C with shaking. Cells are spun down (lEC-Centra 8,400 r.p.m. for 10 min), resuspcnded in 300 ml 2xTY broth containing 100 ;jg ampicillin/ml and 25 /ig kanamycin/ml (2xTY-AMP-KAN) and grown overnight, shaking at 37DC. I'hage particles are purified and concentrated from the culture medium by two PEG-precipilations (Sambrook et al., 1990), resuspended in 2 ml PBS and passed through a 0.45 fim filter (Minisart NML; Sartorius) to give a final concentration of approximately 10" transducing units/ml (ampicillin-resistant clones).
|0947| Panning of the Library. Immunotubes (Nunc) are coated overnight in PBS with 4 ml of either 100 /ig/ml or 10 /ig/ml of an albumin fusion protein of the invention, or a portion of an albumin fusion protein of the invention. Tubes arc blocked with 2% Marvel-PBS for 2 hours at 37°C and then washed 3 times in PBS. Approximately I01J TU of phage is applied to the tube and incubated for 30 minutes at room temperature Himbling on an over and under turntable and then left to stand for another 1.5 hours. Tubes arc washed [Q times with PBS 0.1% Tween-20 and 10 limes with PBS. Phage are eluted by adding I ml of 100 mM triethylamine and rotating 15 minutes on an under and over turntable after which the solution is immediately neutralized with 0.5 nil of l.OM Tris-HCl, pH 7.4. Phage are then used to infect !0 ml of mid-log E. coli TGI by incubating eluted phage with bacteria for 30 minutes at 37°C. The E. coli are then plated on TYE plates containing 1% glucose and 100 /Jg/ml ampicillin. The resulting bacterial library is then rescued with delta gene 3 helper phage as described above to prepare phage for a subsequent round of selection. This process is then repeated for a tola! of 4 rounds of affinity purification with tube-washing increased to 20 times with PBS, 0.1 % Tween-20 and 20 times with PBS for rounds 3 and 4.
|(1948| Characterization of Binders, Eluled phage from the 3rd and 4th rounds of selection are used to infect E. coli HB 2151 and soluble scFv is pioduced (Marks, ct al., 199!) from single colonies for assay. ELISAs are performed with microtitre plates coated with cither 10 pg/ml of an albumin fusion prolein of the invention, or a portion of an albumin fusion protein of the invention, in 50 mM bicarbonate pH 9.6. Clones positive in EIJSA are further characterized by PCR fingerprinting (see, e.g.. International Publication No. WO 92/01047) and then by sequencing. These El.ISA positive clones may also be further characterized by techniques known in the art. such as, for example, epitope mapping, binding affinity, receptor signal transduction, ability to block or competitively inhibit antibody/antigen binding, and competitive agonistic or antagonistic activity.
EXAMPLE 11: l'HI-2-Deoxvulncose Uptake Assay.
|0'M9) Adipose, skeletal muscle, and liver are insulin-sensitive tissues. Insulin can stimulate glucose uptake/transport into these tissues, (n the ca.se of adipose and skeletal muscle, insulin initiates the signal transduction that eventually leads to the translocalion of the glucose transporter 4 molecule, GLUT4, from a specialized intracellular compartment to the cell surface. Once on the celi surface, GLUT4 allows for glucose uptake/lransport.
lJHI-2-Deaxyglucose Uptnkc
|OVSO] A number of adipose and muscle related cell-lines can be used to test for glucose uptake/transport activity in the absence or presence of a combination of any one or more of the therapeutic drugs listed for the treatment of diabetes mellitus. In particular, the 3T3-LI muriiie fibroblast cells and the L6 murine skeletal muscle cells can be differentiated inlo 3T3-L1 adipocyles and into myotuhes, respectively, to serve as appropriate in w/re models fcr the (3H]-2-deoxyglucose uptake assay (Urso et al., J Biol Chem, 274(43): 30864-73 (1999); Wang et al., J Mol Endocrinol, 19(3).
24i;» (iyy//;~Haspel"et"al., J Menibr Biol, 169 (1J: 45-53 (!999); Tsakiridis et al., Endocrinology, 136(10): 4315-22 (1995)). Briefly, 2 x 10s cells/100 uL of adipocytes or differentiated L6 cells are transferred ;a 96-well Tissue-Culture, '"I'C", treated, i.e., coated with 50 jjg/ml. of poly-L-lysine, plates in post-differentiation medium and arc incubated overnight at 37 °C in 5% CO2. The cells are first washed once with serum free low glucose DMEM medium and are then starved with 100 u,L/well of the same medium and with 100 uL/well of either buffer or of a combination of any one or more of the therapeutic drugs listed for the treatment of diabetes mellitus, for example, increasing concentrations of I nM, 10 n.M, and 100 nM of the therapeutics of the subject invention (e.g., specific fusions disclosed as SEQ ID NO:Y and fragments and variants thereof) for 16 hours at 37 CC in the absence or presence of 1 nM insulin. The plates are washed three times with 100 ^L/well of HEPES buffered saline. Insulin is added at 1 nM in HEPES buffered saline for 30 min at 37 °C in the presence of 10 uM labeled [!H]-2-deoxyglucose (Amersham, 0TRK672) and 10 fiM unlabeled 2-deoxyglucose (SIGMA, D-3179). As control, the same conditions are carried out except in the absence of insulin. A final concentration of 10 uM cytochalasin B (SIGMA, C6762) is added at 100 |jIAvell in a separate well to measure the non-specific uptake. The cells arc washed three limes with HEI'ES buffered saline. Labeled, i.e., 10 uM of [3H]-2-deoxyglucosc, and unlabeled, i.e., 10 uM of 2-deoxyglucose, are added for 10 minutes at room temperature. The cells are washed three times with cold Phosphate Buffered &/ine, "PBS". The cells arc lysettupon the addition of 150 u,IVwell of 0.2 N NaOII and subsequent incubation with shaking for 20 minutes at room temperature. Samples aie then transferred to a scintillation vial to which is added 5 mL of scintillation fluid. The vials are counted in a Beta-Scintillation counter. Uptake in duplicate conditions, the difference being the absence or presence of insulin, is determined with the following equation: [(Insulin counts per minute "cpm" - Non-Specific cpm)/(No Insulin cpm - Non-Specific cprn)]. Average responses fall within the limits of about 5-fold and 3-fold that of controls for adipocytes and myotubes, respectively.
DiSTernntialion of Cells
[0951] The cells are allowed to become fully confluent in a T-75 cm2 flask. The medium is removed and replaced with 25 mL of pre-differentiation medium for 48 hours. The cells are incubated at 37 °C, in 5% C02, 85% humidity. After 48 hours, the prc-differentiation medium is removed and replaced with 25 mL differentiation medium for 48 hours. The cells are again incubated at 37 "C, in 5% COi, 85% humidity. After 48 hours, the medium is removed and replaced with 30 mL post-differentiation medium. Post-differentiation medium is maintained for 14-20 days or until complete differentiation is achieved. The medium is changed every 2-3 days. Human adipocytes can be purchased from Zen-Bio, INC (# SA-1096).
EXAMPLE 14: In vitro Assay of fH}-Th\midinc Incorporation into Pancreatic Cell-lines.
t
[0952] It has recently been shown that GLP-1 induces differentiation of the rat pancreatic ductal epithelial cell-line ARIP in a time- and dose-dependent manner which is associated with an increase in Islet Duodenal Homeobox-1 (1DX-1) and insulin mRNA levels (Hui et al., 2001, Diabetes, 50(4): 785-96). The IDX-I in turn increases mRNA levels of the GLP-1 receptor.
Cells Types Tested
|0953J RIN-M cells: These cells arc available from the American Type Tissue Culture Collection (ATCC Cell Line Number CRL-2057 ). The RIN-M cell line was derived from a radiation induced transplanlahle rat islet cell tumor. The line was established from a nude mouse xenografl of Ihe lumor. The cells produce and secrete islet polypeptide hormones, and produce L-dopa decarboxylase (a marker for cells having amine precursor uptake and decarboxylation, or APUD, activity).
[0954| ARIP cells: These are pancreatic exocrme cells of epithelial morphology available from the American Type Tissue Culture Collection (ATCC Cell Line Number CRL-1674). See also, references: Jessop, N.W. and Hay, R.I, "Characteristics of Iwo rat pancreatic cxocrine cell lines derived from transplantabte tumors," In Vitro 16: 212, (1980); Cockell, M. et al., "Identification of a cell-specific DNA-binding activity that interacts with a trarscriptional activator uf genes expressed in the acinar pancreas," Mol. Cell. Biol. 9: 2464-2476, (19X9); Roux, E., et al. "The cell-specific transcription factor P'l'FI contains two different suhunits thai interact with the DNA" Genes Dev. 3: 1613-1624, (1989); and, Hui, II , et al., "Glucagon-like peptide I induces differentiation of islet duodenal homcobox-t-positive pancreatic ductal cells into insulin-secreting cells," Diabetes 50: 785-796(2001).
Preparation nf Cells
[0955] The RFN-M cell-line is grown in RPMI 1640 medium (Hyclone, #SH300027.0I) with 10% fetal bovine serum (HyCIone, #SH30088.03) and is subcuitured every 6 to 8 days at a ralio of 1:3 to 1:6. The medium is changed every 3 to 4 days.
(0956) The ARIP (ATCC ftCRL-1674) cell-line is grown in Hani's F12K medium (ATCC, #30-2004) with 2 mM L-glutamine adjusted to contain 1 5 g/'L sodium bicarbonate and 10% fetal bovine serum. The ARIP cell-line is subcultured at a ratio of 1:3 to 1:6 twice per week. The medium is changed every 3 to 4 days.
.4-v.yny Proiocol
|0957| The cells are seeded at 4000 cells/well in 96-wel! plates and cultured for 48 to 72 hours to 50% confluence. The cells are switched to scrum-free media at 100 uL/well. After incubation lor 48-72 hours, serum and/or the therapeutics of the subject invention (e.g., albumin fusion proteins of the invention and fragments and variants thereof) arc added to the weil Incubation persists for an additional 36 hours. []l!]-Thyrmdine
(5-20 Ci/mmol) (Amersham Pharmacia, #TRK.I20) is diluted to 1 microCuries/5 tnicrolitcrs. After the 3.6 hour incubation, 5 microliters is added per well for a further 24 hours. The reaction is terminated by washing the cells gently with cold Phosphate-Buffered Sal ine, "PBS", once. The cells are men fixed with 100 miciolittn of 10% ice cold TCA for 15 mm at 4 °C. The PBS is removed and 200 microliters of 0.7 N NaOH is added. The plates are incubated for I hour at room temperature with shaking. The solution is transferred to a scintillation vial and 5 mL of scintillation fluid compatible with aqueous solutions i.s added and mixed vigorously. The vials are counted in a beta scintillation counter. As negative control, only buffer is used. As a positive control fetal calf serum is used.
EXAMPLE 15: Assaying for Glycasuria.
|0958J Glycosuria (i.e., excess sugar in the urine), can be readily assayed to provide an index of the disease state of diabetes mellitus. Excess urine in a patient sample as compared with a normal patient sample is symptomatic of IDDM and NIDDM. Efficacy of treatment of such a patient having IDDM and NIDDM is indicated by a resulting decrease in the amount of excess glucose in the urine. In a preferred embodiment for IDDM and NIDDM monitoring, urine samples from patients are assayed for the presence of glucose using techniques known in the art. Glycosuria in humans is defined by a urinary glucose concentration exceeding 100 mg per 100 ml. Excess sugar levels in those patients exhibiting glycosuria can be measured even more precisely by obtaining blood samples and assaying serum glucose.
EXAMPLE 16: Assays Delecting Stimulation or Inhibition of B cell Proliferation and Differentiation.
(09591 Generation of functional humoral immune responses requires both soluble and cognate signaling between B-lineage cells and their microcnvironment. Signals may impart a positive stimulus that allows a B-lineage cell to continue its programmed development, or a negative stimulus that instructs the cell to arrest its current developmental pathway. To date, numerous stimulatory and inhibitory signals have been found to influence B cell responsiveness including IL-2, ILA, IL-5, IL-6, IL-7, IL10, IL-13, 1L-14 and IL-15. Interestingly, these signals are by themselves weak effectors but can, in combination with various co-stimulatory proteins, induce activation, proliferation, differentiation, homing, tolerance and death among B cell populations.
|0960| One of the best studied classes of B-cell co-stimulatory proteins is the TNF-supcrfamily. Within this family CD40, CD27, and CD30 along with their respective ligands CD154, CD70, and CD153 have been found to regulate a variety of immune responses. Assays which allow for the detection and/or observation of the proliferation and differentiation of these B-cell populations and their precursors are valuable tools in determining the effects various proteins may have on these B-cell populations in terms of proliferation and differentiation. Listed below are two assays designed to allow for the detection of the differentiation, proliferation, or inhibition of B-cell populations and their precursors. [0961| In Vitro Assay- Albumin fusion proteins of the invention (including fusion proteins containing fragments or variants of Therapeutic proteins and/or albumin or fragments or variants of albumin) can be assessed for its ability to induce activation, proliferation, differentiation or inhibition and/or death in B-cell populations and their precursors. The activity of an albumin fusion protein of the invention on purified human tonsillar B cells, measured qualitatively over the dose range from 0.1 to 10,000 ng/mL, is assessed in a standard B-lymphocyte co-stimulation assay in which purified tonsillar B cells are cultured in the presence of either formalin-fixed Staphylococcus aureus Cowan I (SAC) or immobilized anti-human IgM antibody as the priming agent. Second signals such as IL-2 and IL-15 synergize with SAC and IgM crosslinking to elicit B cell proliferation as measured by tritiated-thymidine incorporation. Novel synergizing agents can be readily identified using this assay. The assay involves isolating tmrnan tonsillar B cells by magnetic bead (MACS) depiction of CD3-positive cells. The resulting cell population is greater than 95% B cells as assessed by expression of CD45R(B220).
|0962| Various dilutions of each sample are placed into individual wells of a 96-well plate to which are added 10s B-cells suspended in culture medium (RPMI 1640 containing 10% FBS, 5 X 10'!M 2ME, lOOU/ml penicillin, lOug/ml streptomycin, and 10"5 dilution of SAC) in a total volume of 150ul. Proliferation or inhibition is quantitated by a 20h pulse (luCi/well) with 3H-thymidine (6.7 Ci/mM) beginning 72h post factor addition. The positive and negative controls are IL2 and medium respectively.
|0963| In vivo Assay- BALB/c mice are injected (i.p.) twice per day with buffer only, or 2 nig/Kg of an albumin fusion protein of the invention (including fusion proteins containing fragments or variants of Therapeutic proteins and/or albumin or fragments or variants of albumin). Mice receive this treatment for 4 consecutive days, at which time they are sacrificed and various tissues and serum collected for analyses. Comparison of ll&li sections from normal spleens and spleens treated with the albumin fusion protein of the invention identify the results of the activity of the fusion protein on spleen cells, such as the diffusion of peri-arterial lymphatic sheaths, and/or significant increases in the nucleated cellularity of the red pulp regions, which may indicate the activation of the differentiation and proliferation of B-cell populations. Immunohislochemical studies using a D cell marker, anti-CD45R(B220), are used to determine whether any physiological changes to splenic cells, such as splenic disorganization, are due lo increased B-cell representation within loosely defined B-cell zones that infiltrate established T-cell regions.
|0964| Flow cytometric analyses of the spleens from mice treated with the albumin fusion protein is used to indicate whether the albumin fusion protein specifically increases the proportion of ThB+, CD45R(B220)dull B cells over that which is observed in control mice.
|0965| Likewise, a predicted consequence of increased mature B-cell representation in vivo is a relative increase in scrum Ig tilers. Accordingly, scrum IgM and IgA levels are compared between buffer and fusion protein treated mice. K\'AMPLE 17: T Celt Proliferation Assay.
|0966| A CD3-induced proliferation assay is performed on PBMCs and is measured by the uptake of 3H-thymidine. The assay is performed as follows. Ninety-six well plates are coated with 100 ul/well of mAb to CD3 (HIT3a, Pharmingen) or isotype-matched control mAb (B33.1) overnight at 4 degrees C (1 ug/ml in .05M bicarbonate buffer, pll 9.5), then washed three times with PBS. PBMC are isolated by F/H gradient centrifugation from human peripheral blood and added to quadruplicate wells (5 x lO'/well) of mAb coated plates in RPMI containing 10% PCS and P/S in the presence of varying concentrations of an albumin fusion protein of the invention (including fusion proteins containing fragments or variants of Therapeutic proteins and/or albumin or fragments or variants of albumin) (total volume 200 ul). Relevant protein buffer and medium alone are controls. After 48 hr. culture at 37 degrees C, plates arc spun for 2 min. at 1000 rpm and 100 ul of supernatant is removed and stored -20 degrees C for measurement of IL-2 (or other cytokines) if effect on proliferation is observed. Wells are supplemented with 100 ul of medium containing 0.5 uCi of 3H-thymidine and cultured at 37 degrees C for 18-24 hr. Wells are harvested and incorporation of JH-thymidine used as a measure of proliferation. Anti-CD3 alone is the positive control for proliferation. IL-2 (100 U/ml) is also used as a control which enhances proliferation. Control antibody which docs not induce proliferation off cells is used as the negative control for the effects of fusion proteins of the invention.
EXAMPLE 18: Effect of Fusion Proteins of the Invention on the Expression of MHC Class II, Costimulatorv and Adhesion Molecules and Celt Differentiation of Monocytes and Monocyte-Derived Human Dendritic Cells.
|0967] Dendritic cells are generated by the expansion of proliferating precursors found in the peripheral blood: adherent PBMC or elutriated monocytic fractions are cultured for 7-10 days with GM-CSF (50 ng/ml) and IL-4 (20 ng/ml). These dendritic cells have the characteristic phenotype of immature cells (expression of GDI, CD80, CD86, CD40 and MHC class II antigens). Treatment with activating factors, such as TNF-a, causes a rapid change in surface phcnotypc (increased expression of MHC class I and II, costimulatory and adhesion molecules, downregulation of FCyRH, upregulation of CD83). These changes correlate with increased antigen-presenting capacity and with functional maturation of the dendritic cells.
[0968] FACS analysis of surface antigens is performed as follows. Cells are treated 1-3 days with increasing concentrations of an albumin fusion protein of the invention or LPS (positive control), washed with I'BS containing 1% BSA and 0.02 mM sodium azide, and then incubated with 1:20 dilution of appropriate FITC- or PE-labeled monoclonal antibodies for 30 minutes at 4 degrees C. After an additional wash, the labeled cells are analyzed by flow cytometry on a FACScan (Becton Dickinson).
[0969] F.ffecl on the production of cvtokines. Cytokines generated by dendritic cells, in particular IL-12, are important in the initiation of T-cell dependent immune responses. IL-12 strongly influences the development of Thl helper T-cell immune response, and induces cytotoxic T and NK cell function. An El.ISA is used to measure the IL-12 release as follows. Dendritic cells (lO'/ml) arc treated with increasing concentrations of an albumin fusion protein of the invention for 24 hours. LPS (100 ng/ml) is added to the cell culture as positive control. Supernatants from the cell cultures are then collected and analyzed for IL-12 content using commercial ELISA kit (e.g., R & D Systems (Minneapolis, MN)). The standard protocols provided with the kits are used.
[0970] Effect on the expression of MHC Class II, costimulalory and adhesion molecules. Three major families of cell surface antigens can be identified on monocytcs: adhesion molecules, molecules involved in antigen presentation, and Fc receptor. Modulation of the expression of MHC class II antigens and other costimulatory molecules, such as B7 and ICAM-I, may result in changes in the antigen presenting capacity of monocytes and ability to induce T cell activation. Increased expression of Fc receptors may correlate with improved monocyte cytotoxic activity, cytokme release and phagocytosis.
|097I| FACS analysis is used to examine the surface antigens as follows. Monocytes are treated 1-5 days with increasing concentrations of an albumin fusion protein of the invention or LPS (positive control), washed with PBS containing 1% BSA and 0.02 mM sodium azidc, and then incubated with 1:20 dilution of appropriate FITC- or PE-labeled monoclonal antibodies for 30 minutes at 4 degrees C. After an additional wash, the labeled cells are analyzed by flow cytornetry on a FACScan (Becton Dickinson).
|0972] Monocvte activation and/or increased survival. Assays for molecules that activate (or alternatively, inactivate) monocytcs and/or increase monocyte survival (or alternatively, decrease monocyte survival) are known in the art and may routinely be applied to determine whether a molecule of the invention functions as an inhibitor or activator of monocytcs. Albumin fusion proteins of the invention can be screened using the three assays described below. For each of these assays. Peripheral blood mononuclcar cells (PBMC) are purified from single donor leukopacks (American Red Cross, Baltimore, MD) by centrifugation through a I listopaque gradient (Sigma). Monocytes are isolated from PBMC by counterflow centrifugal elutnation.
[0973| Monocyte Survival Assay. Human peripheral blood monocytes progressively lose viability when cultured in absence of serum or other stimuli. Their death results from internally regulated processes (apoplosis). Addition to the culture of activating factors, such as TNF-alpha dramatically improves cell survival and prevents DNA fragmentation. Propidium iodide (PI) staining is used to measure apoptosis as follows. Monocytes are cultured for 48 hours in polypropylene tubes in serum-free medium (positive control), in the presence of 100 ng/ml TNF-alpha (negative control), and in the presence of varying concentrations of the fusion protein to be tested. Cells are suspended at a concentration of 2 x
l()'/ml m CBS containing PI at a final concentration of 5 ng/ml, and then incubated at room temperature for 5 minutes before FACScan analysis. PI
uptake nas oeen aemonsiraieu tu uuncnue with DNA fragmentation in this experimental paradigm.
|0974] Effect an cytokinejelease. An important function of monocytes/macrophages is their regulatory activity on other cellular populations of the immune system through the release of cytokines after stimulation. An ELISA to measure cytokine release is performed as follows. Human monocytes are incubated at a density of 5x10s cells/ml with increasing concentrations of an albumin fusion protein of the invention and under the same conditions, but in the absence of the fusion protein. For 1L-12 production, the cells are primed overnight with IFN (100 U/ml) in the presence of the fusion protein. LI'S (10 ng/ml) is then added. Conditioned media are collected after 24h and kept frozen until use. Measurement of TNF-alpha, 1L-10, MCP-1 and IL-8 is then performed using a commercially available ELISA kit (e.g., R & D Systems (Minneapolis, MN)) and applying the standard protocols provided with the kit.
[0975| Oxidativc burst. Purified monocytes are plated in 96-w plate at 2-lxlO5 cell/well. Increasing concentrations of an albumin fusion protein of the invention are added to the wells in a total volume of 0.2 ml culture medium (RPMI 1640 -t- 10% PCS, glutamine and antibiotics). After 3 days incubation, the plates are centrifuged and the medium is removed from the wells. To the macrophage monolayers, 0.2 ml per well of phenol red solution (140 mM NaCI, 10 mM potassium phosphate buffer pH 7.0, 5.5 mM dextrose, 0.56 mM phenol red and 19 U/ml of HRPO) is added, together with the stimulant (200 nM PMA). The plates are incubated at 37°C for 2 hours and the reaction is stopped by adding 20 ji\ IN NaOH per well. The absorbance is read at 610 nm. To calculate the amount of HjO2 produced by the macrophages, a standard curve of a H2O] solution of known molarity is performed for each experiment.
EXAMPLE 19: The Effect of Albumin Fusion Proteins of the Invention on the Growth of Vascular Endothelial Cells.
[0976J On day 1, human umbilical vein cndothelial cells (HUVL;C) are seeded at 2-5x10* cells/35 mm dish density in M199 medium containing 4% fetal bovine scrum (FBS), 16 units/ml heparin, and 50 units/ml endothelial cell growth supplements (ECGS, Biotechnique, Inc.). On day 2, the medium is replaced with Ml 99 containing 10% FBS, 8 units/ml heparin. An albumin fusion protein of the invention, and positive controls, such as VEGF and basic FGF (bFGF) are added, at varying concentrations. On days 4 and 6, the medium is replaced. On day 8, cell number is determined with a Coulter Counter.
|0977| An increase in the number of HUVEC cells indicates that the fusion protein may proliferate vascular endothelial cells, while a decrease in the number of HUVF.C cells indicates that the fusion protein inhibits vascular endothelial cells.
EXAMPLE 20: Rat Cornea! Wound Heating Model
|0978| This animal model shows the effect of an albumin fusion protein of the invention on neovascularization. The experimental protocol includes:
Making a 1-1.5 mm long incision from the center of cornea into the stromal layer. Inserting a spatula below the lip of the incision facing the outer comer of the eye. Making a pocket (its base is 1-1.5 mm form the edge of the eye).
Positioning a pellet, containing 50ng- 5ug of an albumin fusion protein of the invention, within the pocket.
Treatment with an an albumin fusion protein of the invention can also be applied topically to the comeal wounds in a dosage range of 20mg - 500mg (daily treatment for five days).
EXAMPLE 2t: Diabetic Mouse and Glucocorticoid-lmpaired Wound Healins Models. Diabetic db+/db+ Mouse Model.
|0979| To demonstrate that an albumin fusion protein of the invention accelerates the healing process, the genetically diabetic mouse model of wound healing is used. The full thickness wound healing model in the db+/db+ mouse is a well characterized, clinically relevant and reproducible model uf impaired wound healing. Healing of the diabetic wound is dependent on formation of granulation tissue and rc-epithelialization rather than contraction (Gartner, M.H. etai.J. Surg. Res. .52:389 (1992); Grcenhalgh, D.G. et al. Am. J. Pathol. A?(5:I235 (1990)).
J09XOJ Hie diabetic animals have many of the characteristic features observed in Type II diabetes mellitus. Homozygous (db+/db+) mice are obese in comparison to their normal heterozygous (db+/+m) littermates. Mutant diabetic (db+/dbl-) mice have a single autosomal recessive mutation on chromosome 4 (db+) (Colcman et al. I'roc. Nail. Acad. Sci. USA 77:283-293 (1982)). Animals show polyphagia, polydipsia and polyuria. Mutant diabetic mice (db+/db+) have elevated blood glucose, increased or normal insulin levels, and suppressed cell-mediated immunity (Mandel eial.,J. Immunol. 120:1115 (1978); Debray-Sachs, M. el al.. Clin. Exp. Immunol. 51(!):\-1 (1983); Leiter etal.,Am. J. ofPnlhol. 114:46-55 (1985)). Peripheral neuropathy, myocardial complications, and microvascular lesions, basement membrane thickening and glomerular filtration abnormalities have been described in these animals (Norido, F. et al., Exp. Neural. 83(2):22\-232 (1984); Robertson el a/., Diabetes 29(l):60-67 (1980); Giacomelli et tit.. Lab Invest. 40(^:460-413 (1979); Coleman, D.L., Diabetes 31 (Suppl):\-6 (1982)). These homozygous diabetic mice develop hyperglycernia that is resislant lo insulin analogous to human type II diabetes (Mandel et al.,J. Immunol. 120:1375-1377 (1978)). |09SI| The characteristics observed in these animals suggests that healing in this model may be similar to the healing observed in human diabetes (Grcenhalgh, el al. Am. J. of Pathol. /J |0')S2| Genetically diabetic female C57BL/KsJ (db+/db+) mice and their non-diabetic (db+/+m) heterozygous littermates are used in this study (Jackson laboratories). The animals are purchased at 6 weeks of age and are 8 weeks old at the beginning of the study. Animals are individually
housed and received tood ana water ad libitum. All manipulations are performed using aseptic techniques. The experiments are conducted
according to the rules and guidelines of Human Genome Sciences, Inc. Institutional Animal Care and Use Committee and the Guidelines for the
Care and Use of Laboratory Animals.
|0983| Wounding protocol is performed according to previously reported methods (Tsuboi, R. and Rifkin, D.B., J. Exp. Meii. 772:245-251
(1990)). Briefly, on the day of wounding, animals are anesthetized with an intraperitoneal injection of Avertin (0.01 mg/mL), 2,2,2-tnbromocthanol
and 2-rncthyl-2-butanol dissolved in deionized water. The dorsal region of the animal is shaved and the skin washed with 70% ethanol solution and
iodine. The surgical area is dried with sterile gauze prior to wounding. An 8 mm full-thickness wound is then created using a Kcyes tissue punch.
Immediately following wounding, the surrounding skin is gently stretched to eliminate wound expansion. The wounds are left open for the duration
of the experiment. Application of the treatment is given topically for 5 consecutive days commencing on the day of wounding. Prior to treatment,
wounds are gently cleansed with sterile saline and gauze sponges.
|0984] Wounds are visually examined and photographed at a fixed distance at the day of surgery and at two day intervals thereafter. Wound
closure is determined by daily measurement on days 1-5 and on day 8. Wounds are measured horizontally and vertically using a calibrated Jameson
caliper. Wounds are considered healed if granulation tissue is no longer visible and the wound is covered by a continuous epithelium.
[0985] An albumin fusion protein of the invention is administered using at a range different doses, from 4mg to SOOmg per wound per day for
8 days in vehicle. Vehicle control groups received 50mL of vehicle solution.
[0986] Animals are euthanized on day 8 with an intraperitoneal injection of sodium pentobarbital (300mg/kg). The wounds and surrounding
skin are then harvested for histology and immnnohistochemistry. Tissue specimens are placed in 10% neutral buffered formalin in tissue cassettes
between biopsy sponges for further processing.
[0987) Three groups of 10 animals each (5 diabetic and 5 non-diabetic controls) are evaluated: 1) Vehicle placebo control, 2) untreated group,
and 3) treated group.
|0988) Wound closure is analyzed by measuring the area in the vertical and horizontal axis and obtaining the total square area of the wound.
Contraction is then estimated by establishing the differences between the initial wound area (day 0) and that of post treatment (day 8). The wound
area on day 1 is 64mm!, the corresponding size of the dermal punch. Calculations are made using the following formula:
a. [Open area on day 8] - [Open area on day 1 ] / [Open area on day 1 ]
[0989] Specimens arc fixed in 10% buffered formalin and paraffin embedded blocks are sectioned perpendicular to the wound surface (5mm) and cut using a Rcichert-Jung microtome. Routine hematoxylin-cosin (H&E) staining is performed on cross-sections of bisected wounds, liistologic examination of the wounds are used to assess whether the healing process and the morphologic appearance of the repaired skin is altered by treatment with an albumin fusion protein of the invention. This assessment included verification of the presence of cell accumulation, inflammatory cells, capillaries, fibroblasts, re-epithelialization and epidermal maturity (Greenhalgh, D.G. et al.. Am. J. Pathol. 136:\23S (1990)). A calibrated lens micrometer is used by a blinded observer.
|0990] Tissue sections are also stained immunohistochemically with a polyclonal rabbit anti-human keratin antibody using ABC Elite detection system. Human skin is used as a positive tissue control while non-immune IgG is used as a negative control. KeraTinocyte growth is determined by evaluating the extent of reepithelialization of the wound using a calibrated lens micrometer.
|0991] Proliferating cell nuclear antigen/cyclin (PCNA) in skin specimens is demonstrated by using anti-PCNA antibody (1:50) with an ABC Elite detection system. Human colon cancer served as a positive tissue control and human brain tissue is used as a negative tissue control. Each specimen included a section with omission of the primary antibody and substitution with non-immune mouse IgG. Ranking of these sections is ba^ed on the extent of proliferation on a scale of 0-8, the lower side of the scale reflecting slight proliferation to the higher side reflecting intense proliferation. [0992] Experimental data arc analyzed using an unpaired t test. A p value of Steroid Impaired Rat Model
|0993| The inhibition of wound healing by steroids has been well documented in various in vitro and in vivo systems (Wahl, Glucocorticoids and Wound healing. In: Anti-Inflammatory Steroid Action: Basic and Clinical Aspects. 280-302 (1989); Wahlef al., J. Immunol. /IS: 476-481 (1975), Werb el at., J. Ex/>. Mcd. 747:1684-1694 (1978)). Glucocorticoids retard wound healing by inhibiting angiogenesis, decreasing vascular permeability (Ebert et nl., An. Intern. Mcd. J7:701-705 (1952)), fibroblast proliferation, and collagen synthesis (Beck et al., Growth Factors. 5: 295-304 (1991); Haynes et al., J. Clin. Invest. 61: 703-797 (1978)) and producing a transient reduction of circulating monocytes (Haynes ct al., J. Clin. Invest. 61: 703-797 (1978); Wahl, "Glucocorticoids and wound healing", In: Antiinflammatory Steroid Action: Basic and Clinical Aspects, Academic Press, New York, pp. 280-302 (1989)). The systemic administration of steroids to impaired wound healing is a well establish phenomenon in rals (Heck et al., Growth Factor-:. 5: 295-304 (1991); Haynes et al., J. Clin. Invest. 61: 703-797 (1978); Wahl, "Glucocorticoids and wound healing", In: Antiinflammatory Steroid Action: Basic and Clinical Aspects, Academic Press, New York, pp. 280-302 (1989); Pierce ct nl., Prcic. Nail. Acad. Sci. USA 86: 2229-2233 (1989)).
|fl994J To demonsirale that an albumin fusion protein of the invention can accelerate Ihc healing process, the effects of multiple topical
applications of the fusion protein on tun thickness excisiona] skin wounds in rats in which healing has been impaired by the systemic administration
of methylprednisolone is assessed
|u995] V'oung adult male Sprague Dawlcy rats weighing 250-300 g (Charles River Laboratories) arc used in this example The animals are
purchased at 8 weeks of age and are 9 weeks old at the beginning of the study. The healing response of rats is impaired by the systemic
administration of methylprednisolone (I7mg/kg/rat intramuscularly) at the time of wounding. Animals are individually housed and received food
and water ad libitum. All manipulations are performed using aseptic techniques. This study is conducted according to the rules and guidelines of
Human Genome Sciences, Inc. Institutional Animal Care and Use Committee and the Guidelines for the Care and Use of Laboratory Animals.
(0996| The wounding protocol is followed according to that described above. On the day of wounding, animals are anesthetized with an
intramuscular injection of ketamine (50 mg/kg) and xylazine (5 mg/kg). The dorsal region of the animal is shaved and the skin washed with 70%
ethanol and iodine solutions. The surgical area is dried with sterile gauze prior to wounding. An 8 mm full-thickness wound is created using a
Kcycs tissue punch. The wounds are left open for the duration of the experiment. Applications of the testing materials are given topically once a
day for 7 consecutive days commencing on the day of wounding and subsequent to methylprednisolone administration. Prior to treatment, wounds
arc gently cleansed with sterile saline and gauze sponges.
[0997J Wounds are visually examined and photographed at a fixed distance at the day of wounding and at the end of treatment. Wound closure
is determined by daily measurement on days 1-5 and on day 8. Wounds are measured horizontally and vertically using a calibrated Jameson caliper.
Wounds are considered healed if granulation tissue is no longer visible and the wound is covered by a continuous epithelium.
|0998] 'Hie fusion protein of the invention is administered using at a range different doses, from 4mg to 500mg per wound per day for 8 days
in vehicle. Vehicle control groups received 50mL of vehicle solution.
|0999] Animals are euthanized on day 8 with an intraperitoneal injection of sodium pentobarbital (300mg/kg). The wounds and surrounding
skin are then harvested for histology. Tissue specimens are placed in 10% neutral buffered formalin in tissue cassettes between biopsy sponges for
further processing.
|1000| Three groups of 10 animals each (5 with methylprednisolone and 5 without glucocorticoid) are evaluated: I) Untreated group 2) Vehicle
placebo control 3) treated groups.
110011 Wound closure is analyzed by measuring the area in the vertical and horizontal axis and obtaining the total area of the wound. Closure is
then estimated by establishing the differences between the initial wound area (day 0) and that of post treatment (day 8). The wound area on day 1 is
64mm2, the corresponding size of the dermal punch. Calculations arc made using the following formula:
b. [Open area on day 8J - [Open area on day I] / [Open area on day 1]
[1002| Specimens are fixed in 10% buffered formalin and paraffin embedded blocks are sectioned perpendicular to the wound surface (5mm) and cut using an Olympus microtome. Routine hematoxylm-eosin (H&K) staining is performed on cross-sections of bisected wounds. Histologic examination of the wounds allows assessment of whether the healing process and the morphologic appearance of the repaired skin is improved by treatment with an albumin fusion protein of the invention. A calibrated lens micrometer is used by a blinded observer to determine the distance of the wound gap. |1003| Experimental data are analyzed using an unpaired t test. A p value of EXAMPLE 22: Lymphedema Animal Model.
|1004| The purpose of this experimental approach is to create an appropriate and consistent lymphederna model for testing the therapeutic effects of an albumin fusion protein of the invention in lymphangiogenesis and re-establishment of the lymphatic circulatory system in the rat hind limb. Effectiveness is measured by swelling volume of the affected limb, quantification of the amount of lymphatic vasculature, total blood plasma protein, and histopathology. Acute lymphederna is observed for 7-10 days. Perhaps more importantly, the chronic progress of the edema is followed for up to 3-4 weeks.
[1005] Prior to beginning surgery, blood sample is drawn for protein concentration analysis. Male rats weighing approximately ~350g are dosed with Pentobarbital. Subsequently, the right legs are shaved from knee to hip. The shaved area is swabbed with gau2e soaked in 70% EtOH. Blood is drawn for serum total protein testing. Circumference and volumetric measurements arc made prior to injecting dye into paws after marking 2 measurement levels (0.5 cm above heel, at mid-pt of dorsal paw). The intradermal dorsum of both right and left paws are injected with 0.05 ml of 1% Evan's Blue. Circumference and volumetric measurements arc then made following injection of dye into paws.
|1006| Using the knee joint as a landmark, a mid-leg inguinal incision is made circumferentially allowing the femoral vessels to be located. Forceps and hemostals are used to dissect and separate the skin (laps. After locating the femora] vessels, the lymphatic vessel that runs along side and underneath the vessel(s) is located. The main lymphatic vessels in this area are then electrically coagulated or suture ligated. 11007] Using a microscope, muscles in back of the leg (near the semitendinosis and adductors) arc bluntly dissected. The popliteal lymph node is then located. The 2 proximal and 2 distal lymphatic vessels and distal blood supply of the popliteal node are then ligated by suturing. The popliteal lymph node, and any accompanying adipose tissue, is then removed by cutting connective tissues.
[10!)8| Care is taken to control any mild bleeding resulting from this procedure. After lymphatics arc occluded, the skin flaps are sealed by
using liquid skin (Vetbond) (AJ Buck). The separated skin edges are scaled to the underlying muscle tissue while leaving a gap of-0.5 cm around
the leg. Skin also may he anchored by suturing to underlying muscle when necessary.
11009] To avoid infection, animals are housed individually with mesh (no bedding). Recovering animals are checked daily through the optimal
edernatous peak, which typically occurred by day 5-7. The plateau edematous peak are then observed. To evaluate the intensity of the lymphedema,
the circumference and volumes of 2 designated places on each paw before operation and daily for 7 days are measured. The effect of plasma
proteins on lymphedema is determined and whether protein analysis is a useful testing perimeter is also investigated. The weights of both control
and edematous limbs are evaluated at 2 places. Analysis is performed in a blind manner.
11010| Circumference Measurements: Under brief gas anesthetic to prevent limb movement, a cloth tape is used to measure lirnb
circumference. Measurements are done at the ankle bone and dorsal paw by 2 different people and those 2 readings are averaged. Readings are
taken from both control and edematous limbs.
110111 Volumetric Measurements: On the day of surgery, animals are anesthetized with Pentobarbital and are tested prior to surgery. For daily
volumetrics animals are under brief halothane anesthetic (rapid immobilization and quick recovery), and both legs are shaved and equally marked
using waterproof marker on legs. Legs are first dipped in water, then dipped into instrument to each marked level then measured by Buxco edema
sofrware(Chen/Victor). Data is recorded by one person, while the other is dipping the limb to marked area.
[1012] Blood-plasma protein measurements: Blood is drawn, spun, and serum separated prior to surgery and then at conclusion for total protein
and Ca2* comparison.
|1013| Limb Weight Comparison: After drawing blood, the animal is prepared for tissue collection. The limbs are amputated using a quillitine,
then both experimental and control legs are cut at the ligature and weighed. A second weighing is done as the tibio-cacancal joint is disarticulated
and the foot is weighed.
(1014J Histological Preparations: The transverse muscle located behind the knee (popliteal) area is dissected and arranged in a metal mold,
filled with freezeGel, dipped into cold methylbutane, placed into labeled sample bags at - 80EC until sectioning. Upon sectioning, the muscle is
observed under fluorescent microscopy for lymphatics..
EXAMPLE 23: Suppression of 77V/1 alpha-Induced Adhesion Molecule Expression by an Albumin Fusion Protein of the
Invention.
The recruitment of lymphocytes to areas of inflammation and angiogenesis involves specific rcceptor-ligand interactions between cell
surface adhesion molecules (CAMs) on lymphocytes and the vascular cndothelium. The adhesion process, in both normal and pathological settings,
follows a multi-step cascade that involves intercellular adhesion molecule-1 (ICAM-I), vascular cell adhesion molecule-1 (VCAM-1), and
endothelial leukocyte adhesion molecule-1 (E-selcctin) expression on endothelial cells (EC). The expression of these molecules and others on the
vascular cndothelium determines the efficiency wilh which leukocytes may adhere to the local vasculature and extravasate into the local tissue
during the development of an inflammatory response. The local concentration of cytokmes and growth factor participate in the modulation of the
expression of these CAMs.
Tumor necrosis factor alpha (TNF-a), a potent proinflamrnatory cytokine, is a stimulator of all three CAMs on endothelial cells and may
be involved in a wide variety of inflammatory responses, often resulting in a pathological outcome.
|1017| The potential of an albumin fusion protein of the invention to mediate a suppression of TNF-a induced CAM expression can be examined. A modified EL1SA assay which uses EC's as a solid phase absorbent is employed to measure the amount of CAM expression on TNF-a treated KCs when co-stimulated with a member of the FGF family of proteins.
[1018] To perform the experiment, human umbilical vein endothelial cell (HUVEC) cultures are obtained from pooled cord harvests and maintained in growth medium (ECiM-2; Clonetics, San Diego, CA) supplemented with 10% PCS and 1% penicillin/streptomycin in a 37 degree C humidified incubator containing 5% CO>2 HUVECs are seeded in 96-well plates at concentrations of 1 x 10 cells/well in EGM medium at 37 degree C for 18-24 hrs or until confluent. The monolayers are subsequently washed 3 times with a serum-free solution of RPMI-1640 .supplemented wilh 100 U/ml penicillin and 100 nig/ml streptomycin, and treated with a given cytokine and/or growth factor(s) for 24 h at 37 degree C. Following incubation, the cells are then evaluated for CAM expression.
|1019] Human Umbilical Vein Endothelial cells (HUVECs) are grown in a standard 96 well plate to confluence, Growth medium is removed from the cells and replaced wilh 90 ul of 199 Medium (10% FBS). Samples for testing and positive or negative controls are added to the plate in triplicate (in 10 ul volumes). Plates are incubated at 37 degree C for either 5 h (selectin and integrin expression) or 24 h (integrin expression only). Plates are aspirated to remove medium and 100 /il of 0 1% paraformaldehyde-PBS(with Ca*~* and Mg") is added to each well. Plates are held at 4°C fur 30 niin.
[1020) Fixative is then removed from the wells and wells arc washed IX with PBS(-t-Ca,Mg)-H).5% BSA and drained. Do not allow the wells to dry. Add 10 /il of diluted primary antibody to the test and control wells. Anti-ICAM-1-Biotin, Ami-VCAM-1-Biotin and Anti-E-selectin-Biotin are used at a concentration of 10 /ig/ml (1:10 dilution of O.I mg/ml stock antibody). Cells are incubated at 37°C for 30 min. in a humidified
environment. Wells are washed X3 with PBS(-tCa,Mg)+0.5% BSA
[1021] Then add 20 /il of diluted ExtrAvidin-Alkaline Phosphotase (1:5,000 dilution) to each well and incubated at 37°C for 30 min. Wells are washed1 X3 with PBS(-t-Ca,Mg)-r0.5% BSA. 1 tablet of p-NitiophcnoI Phosphate pNPP is dissolved in 5 ml of glycine buffer (pH 10.4). 100 ;il of pNPP substrate in glycine buffer is added to each test well. Standard wells in triplicate are prepared from the working dilution of the ExtrAvidin-Alkaline Phosphotase in glycine buffer: 1:5,000 (10°) > 10'" > 10' > ID'15. 5 /il of each dilution is added to triplicate wells and the resulting AP content in each well is 5.50 ng, 1.74 ng, 0.55 ng, 0.18 ng. 100 /il of pNNP reagent must then be added to each of the standard wells. The plate must be incubated at 37°C for 4h. A volume of 50 /il of 3M NaOH is added to all wells. The results are quantified on a plate reader at 405 nm. The background subtraction option is used on blank wells filled with glycine buffer only. The template is set up to indicate the concentration of AP-conjugate in each standard well [ S.50 ng; 1.74 ng; 0.55 ng; 0.18 ng]. Results are indicated as amount of bound AP-conjugate in each sample.
EXAMPLE 24: Construction of GAS Reporter Construct.
|1022| One signal transduction pathway involved in the differentiation and proliferation of cells is called the Jaks-STATs pathway. Activated proteins in the Jaks-STATs pathway bind to gamma activation site "GAS" elements or interferon-sensitive responsive element ("1SRE"), located in the promoter of many genes. The binding of a protein to these elements alter the expression of the associated gene.
|1023| GAS and 1SRE elements are recognized by a class of transcription factors called Signal Transducers and Activators of Transcription, or "STATs." There arc six members of the STATs family. Stall and Stat3 are present in many cell types, as is Stat2 (as response to IFN-alpha is widespread). Stal4 is more restricted and is not in many cell types though it has been found in T helper class I, cells after treatment with IL-12. Stat5 was originally called mammary growth factor, but has been found at higher concentrations in other cells including myeloid cells. It can be activated in tissue culture cells by many cytokines.
|1024| The STATs are activated to translocate from the cytoplasm to the nucleus upon tyrosine phosphorylation by a set of kinases known as the Janus Kinase ("Jaks") family. Jaks represent a distinct family of soluble tyrosine kinases and include Tyk2, Jakl, Jak2, and Jak3. These kinases display significant sequence similarity and arc generally catalytically inactive in resting cells.
(1025) The Jaks are activated by a wide range of receptors summarized in the Table below. (Adapted from review by Schidler and Darnell, Ann. Rev. Biochem. 64:621-51 (1995)). A cytokine receptor family, capable of activating Jaks, is divided into two groups: (a) Class 1 includes receptors for IL-2, IL-3, IL-4,1L-A, 1L-7, IL-9, IL-11, IL-12, IL-15, Epo, PRL, GH, G-CSF, GM-CSF, LIF, CNTF, and thrombopoietin; and (b) Class 2 includes IFN-a, IFN-g, and IL-10. The Class 1 receptors share a conserved cysteine motif (a set of four conserved cysteines and one tryptophan) and a WSXWS motif (a membrane proximal region encoding Trp-Scr-Xaa-Trp-Scr (SEQ ID NO:53)).
|1026| Thus, on binding of a ligand to a receptor, Jaks are activated, which in turn activate STATs, which then translocate and bind to GAS elements. This entire process is encompassed in the Jaks-STATs signal transduction pathway. Therefore, activation of the Jaks-STATs pathway, reflected by the binding of the GAS or the ISRE clement, can be used to indicate proteins involved in the proliferation and differentiation of cells. For example, growth factors and cytokines are known to activate the Jaks-STATs pathway (See Table 5, below). Thus, by using GAS elements linked to reporter molecules, activators of the Jaks-STATs pathway can be identified.
(Table Removed)
|I027| To construct a synthetic GAS containing promoter element, which is used in the Biological Assays described in Examples 27-29, a PCR
based strategy is employed to generate a GAS-SV40 promoter sequence. The 5' primer contains four tandem copies of the GAS binding site found
in the IRF1 promoter and previously demonstrated to bind STATs upon induction with a range of cytokines (Rothman et al., Immunity 1:457-468
(1994).), although other GAS or ISRE elements can be used instead. The 5' primer also contains 18bp of sequence complementary to the SV40
early promoter sequence and is flanked with an Xhol site. The sequence of the 5' primer is:
S^GCGCCTCGAGAriTCCCCGAAATC'fAGAlTrCCCCGAAATGA'nTCCCCGAAATGATrTCCCCGAAATATCTGCCAlCrCAATTAG:
3' (SEQ ID NO:54)
|I028| The downstream primer is complementary to the SV40 promoter and is flanked with a Hind III site:
5':GCGGCAAGCTTTTTGCAAAGCCTAGGC:3' (SEQ ID NO:55)
11029| PCR amplification is performed using the SV40 promoter template present in the B-galpromoter plasmid obtained from Clontech. The
resulting PCR fragment is digested with XhoI/Hind III and subcloned into BLSK2-. (Stratagene.) Sequencing with forward and reverse primers
confirms that the insert contains the following sequence:
yXn'CGACiA'riTCCCCGAAA'I'CrAGA'ITrCCCCGAAA'rGATTTCCCCGAAA'l'GA'nTCCCCGAAA'rATCTGCCATCTCAAlTAGTCAGC
AACCATAGTCCCGCCCX'TAACTCCGCCCAirCCGCCCCTAACrCCGCCCAGTTCCGCCCATTCTCCGCCCCATGGCTGACTAATTTTTr
TTATl'rAI-GCAaAGGCCGAGGCCGCCTCGGCCTCTGAGCTATTCCAGAAGTAGTGAGGAGGCTTTTTTGGAGGCCTAGGCTrrrGCAA
AAAGCTT:3' (SRQ ID NO:S6)
11030) With this GAS promoter element linked to the SV40 promoter, a GAS:SEAP2 reporter construct is next engineered. Here, the reporter
molecule is a secreted alkaline phosphatase, or "SEAP." Clearly, however, any reporter molecule can be instead of SEAP, in this or in any of the
other Examples. Well known reporter molecules that can be used instead of SEAP include chloramphenicol acetyltransferase (CAT), luciferase,
alkaline phosphatase, B-galactosidasc, green fluorescent protein (GFP), or any protein detectable by an antibody.
(1031] The above sequence confirmed synthetic GAS-SV40 promoter element is subcloned into the pSEAP-Promoter vector obtained from
Clonlcch using Hindlll and Xhol, effectively replacing the SV40 promoter with the amplified GAS:SV40 promoter element, to create the GAS-
SEAP vector. However, this vector does not contain a neomycin resistance gene, and therefore, is not preferred for mammalian expression systems.
|1032| Thus, in order to generate mammalian stable cell lines expressing the GAS-SEAP reporter, the GAS-SEAP cassette is removed from the
GAS-SEAP vector using Sail and Noll, and inserted into a backbone vector containing the neomycin resistance gene, such as pGEP-1 (Clontech),
using these restriction sites in the multiple cloning site, to create the GAS-SEAP/Neo vector. Once this vector is transfccted into mammalian cells,
this vector can then be used as a reporter molecule for GAS binding as described in Examples 27-29.
[1033| Other constructs can be made using the above description and replacing GAS with a different promoter sequence. For example,
construction of reporter molecules containing EGR and NF-KB promoter sequences are described in Examples 27-31. However, many other
promoters can be substituted using ihe protocols described in these Examples. For instance, SRE, 1L-2, NEAT, or Osteocalcin promoters can be
substituted, alone or in combination (e.g., GAS/NF-KB/EGR, GAS/NE-K.B, ll-2/NFAT, or NF-KB/GAS). Similarly, other cell lines can be used to
lest reporter construct activity, such as HELA (epithelial), HUVEC (endothelial), Reh (B-cell), Saos-2 (osteoblast), HUVAC (aortic), or
Cardiomyocyte.
F.XAMPLK 25: Assay for SF.AP Activity.
|ll)34| As a reporter molecule for the assays described in examples disclosed herein, SEAP activity is assayed using the Tropix Phospho-light Kit (Cat. BP^OO) according to the following general procedure. The Tropix Phospho-light Kit supplies the Dilution, Assay, and Reaction Buffers used below.
|1035] Prime a dispenser with the 2.5x Dilution Buffer and dispense 15 ul of 2.5x dilution buffer into Optiplates containing 35 ul of a solution containing an albumin fusion protein of the invention. Seal the plates with a plastic sealer and incubate at 65 degree C for 30 min. Separate the Optiplates to avoid uneven heating. |1036| Cool the samples to room temperature for 15 minutes. Empty the dispenser and prime wilh the Assay Buffer. Add 50 ml Assay Buffer
and incubale at room temperature 5 min. Empty the dispenser and prime with the Reaction Buffer (see the Table below). Add 50 ul Reaction Butter and incubate at room temperature for 20 minutes. Since the intensity of the ehemiluminescent signal is time dependent, and it takes about 10 minutes to read 5 plates on a lummometer, Ihus one shnulrl treat (Table Removed)
EXAMPLE 26: Assay Identifying NeuronalActivity.
(1038] When cells undergo differentiation and proliferation, a group of genes are activated through many different signal transduction pathways. One of these genes, EGR1 (early growth response gene 1), is induced in various tissues and ceil types upon activation. The promoter of EGR1 is responsible for such induction. Using the EGR1 promoter linked to reporter molecules, the ability of fusion proteins of the invention to activate cells can be assessed.
11039] Particularly, the following protocol is used to assess ncuronal activity in PC12 cell lines. PC12 cells (rat phenochromocytoma cells) are known to proliferate and/or differentiate by activation with a number of mitogens, such as TPA (tetradecanoyl phorbol acetate), NGF (nerve growth factor), and EOF (epidermal growth factor). The HGR1 gene expression is activated during this treatment. Thus, by stably transfecting PC 12 cells with a construct containing an EGR promoter linked to SEAP reporter, activation of PC12 cells by an albumin fusion protein of the present invention can be assessed.
(1040) The EGR/SEAP reporter construct can be assembled by the following protocol. The EGR-1 promoter sequence (-633 to +l)(Sakamoto K ct al., Oncogcne 6:867-871 (1991)) can be PCR amplified from human genomic DNA using the following primers:
First primer: 5' GCGCTCGAGGGATGACAGCGATAGAACCCCGG-31 (SEQ ID NO.57)
Second primer: 5' GCGAAGCTTCGCGACTCCCCGGATCCGCCTC-3' (SEQ ID NO:58)
|1041| Using the GAS:SEAH/Neo vector produced in Example 24, EGR1 amplified product can then be inserted into this vector. Linearize the GAS:SEAP/Neo vector using restriction enzymes Xliol/Hindlll, removing the GAS/SV40 stuffer. Restrict the EGR1 amplified product with these same enzymes. Ligate the vector and the EGR I promoter.
|1042| To prepare 96 well-plates for cell culture, two mis of a coating solution (1:30 dilution of collagen type 1 (Upstate Biotech Inc. Cat#08-115) in 30%ethanol (filter sterilized)) is added per one 10cm plate or 50 ml per well ofthe96-well plate, and allowed to air dry for 2 hr. 11043] PC 12 cells are routinely grown in RPMI-1640 medium (Bio Whiltaker) containing 10% horse serum (JRH BIOSCIENCES, Cat. # 12449-78P), 5% heat-inactivated fetal bovine serum (FBS) supplemented with 100 units/ml penicillin and 100 ug/ml streptomycin on a precoated 10 cm tissue culture dish. One to four split is done every three to four days. Cells are removed from the plates by scraping and resuspcndcd with pipetting up and down for more than 15 times.
|I044| Transfect the EGR/SEAP/Nco conslrucl into PC12 using techniques known in the art. EGR-SEAP/PCI2 stable cells are obtained by growing the cells in 300 ug/ml G4I8. The G418-frce medium is used for routine growth but every one to two months, the cells should be re-grown in 300 ug/ml G418 for couple of passages
|1045| To assay for neuronal activity, a 10 cm plate with cells around 70 to 80% confluent is screened by removing the old medium. Wash the
ceils once w.th PUS (Fhospnate Dunered saline), Then starve the cells in low scrum medium (RPMM640 containing i% horse serum and 0.5%
FBS with antibiotics) overnight.
11046] The r*xt morning, remove the medium and wash the cells with PBS. Scrape off the cells from the plate, suspend the cells well in 2 ml
low serum medium. Count the cell number and add more low serum medium to reach final cell density as 5xl05 cells/ml.
|1047| Add 200 ul ofthc cell suspension to each well of 96-well plate (equivalent to U105 cells/well). Add a series of different concentrations
of an albumin fusion protein of the inventon, 37 degree C for 48 to 72 hr. As a positive control, a growth factor known to activate PC12 cells
through EGR can be used, such as SO ng/ul of Neuronal Growth Factor (NGF). Over fifty-fold induction of SEAP is typically seen in the positive
control wells. SEAP assay may be routinely performed using techniques known in the art anil/or as described in Example 25.
EXAMPLE 27: Assay for T-cell Activity.
|1048J The following protocol is used to assess T-cell activity by identifying factors, and determining whether an albumin fusion protein of the invention proliferates and/or differentiates T-cells. T-cell activity is assessed using the GAS/SEAP/Neo construct produced in Example 24. Thus, factors that increase SEAP activity indicate the ability to activate the Jaks-STATS signal transduction pathway. The T-cell used in this assay is Jurkat T-cells (ATCC Accession No. TIB-152), although Molt-3 cells (ATCC Accession No. CRL-1552) and Molt-4 cells (ATCC Accession No. CRL-1582) cells can also be used.
|I049| Jurkat T-cells are lymphoblastic CD4+ Thl helper cells. In order to generate stable cell lines, approximately 2 million Jurkat cells are transfected with the GAS-SEAP/neo vector using DMRIE-C (Life Technologies)(rransfection procedure described below). The transfected cells are seeded to a density of approximately 20,000 cells per well and transfectants resistant to 1 mgAnl genticin selected. Resistant colonies are expanded and then tested for their response to increasing concentrations of interferon gamma. The dose response of a selected clone is demonstrated. [lOSOj Specifically, the following protocol will yield sufficient cells for 75 wells containing 200 ul of cells. Thus, it is either scaled up, or performed in multiple to generate sufficient cells for multiple 96 well plates. Jurkat cells are maintained in RPMI + 10% serum with l%Pen-Strep. Combine 2.5 nils of OPTI-MEM (Life Technologies) with 10 ug of plasmid DNA in a T25 flask. Add 2.5 ml OPTI-MEM containing 50 ul of DMRIE-C and incubate at room temperature for \S-45 mins.
During the incubation period, count cell concentration, spin down the required number of cells (101 per transfection), and resuspend in
OPTI-MEM to a final concentration of 10' cells/ml. Then add 1ml of I x 107 cells in OPTI-MEM to T25 flask and incubate at 37 degree C for 6
hrs. After the incubation, add 10 ml of RPMI + 15% serum.
The Jurkat:GAS-SEAP stable reporter lines are maintained in RPMI+ 10% serum, 1 nig/ml Genticin, and l%Pen-Strep. These cells are
treated with varying concentrations of one or more fusion proteins of the present invention.
[1053) Ort the day of treatment with the fusion protein, the cells should be washed and resuspended in fresh RPMI + 10% serum to a density of
500,000 celts per ml. The exact number of cells required will depend on the number of fusion proteins and the number of different concentrations
effusion proteins being screened. For one 96 well plate, approximately 10 million cells (for 10 plates, 100 million cells) arc-required.
110541 The. well dishes containing Jurkat cells treated with the fusion protein are placed in an incubator for 48 hrs (note: this time is variable
between 48-72 hrs]. 35 ul samples from each well are then transferred to an opaque 96 well plate using a 12 channel pipette. The opaque plates
should be covered (using sellophene covers) and stored at -20 degree C until SEAP assays are performed according to Example 25. The plates
containing the remaining treated cells are placed at 4 degree C and serve as a source of material for repeating the assay on a specific well if desired.
(HISS) As a positive control, 100 Unil/ml interferon gamma can be used which is known to activate Jurkat T cells. Over 30 fold induction is
typically observed in the positive control wells.
[1056) The above protocol may be used in the generation of both transient, as well as, stable transfected cells, which would be apparent to those
of skill in the art.
K.KAMPLE2S: Assay far T-cell Activity.
|1057| NF-KB (Nuclear Factor KB) is a transcription factor activated by a wide variety of agents including the inflammatory cytokines 1L-I and TNF, CD30 and CD40, lymprioloxin-alpha and lymphotoxin-beta, by exposure to LPS or thrombin, and by expression of certain viral gene products. As a transcription factor, NF-K.H regulates the expression of genes involved in immune cell activation, control of apoptosis (NF- KB appears to shield cells from apoptosis), B and T-cell development, anti-viral and antimicrobial responses, and multiple stress responses.
|I058| In non-stimulated conditions, NF- KB i.s retained in the cytoplasm with I-KB (Inhibitor KB). However, upon stimulation, 1- KB is phosphorylated and degraded, causing NF- K3 to shuttle to the nucleus, thereby activating transcription of target genes. Target genes activated by NF- KB include U..-2, IL-6, GM-CSF, 1CAM-1 and class 1 MHC.
|1059| Due to its central role and ability to respond to a range of stimuli, reporter constructs utilizing the NF-KB promoter element are used to screen the fusion protein. Activators or inhibitors of NF-KB would be useful in treating, preventing, and/or diagnosing diseases. For example, inhibitors of NF-KB could be used to Ircat those diseases related to the acute or chronic activation of NF-KB, such as rheumatoid arthritis. |I060| To construct a vector containing the NF-KB promoter element, a PCR based strategy is employed. The upstream primer contains four
tandem copies cf the NF-KB binding site (GGGGACTITCCC) (SEQ ID N0:59), 18 bp of sequence complementary to the 5' end of the SV4Q early promoter sequence, and is flanked with an Xhol site:
V:utGGCCltGAGGGOAC rrrCCCGGGGACTTTCCGGGGACTTTCCGGGACTTTCCATCCTGCCATCTrA^rTAG^' (SEQ ID NO-.60) 110611 The downstream primer is complementary to the 3' end of ihe SV40 promoter and is flanked with a Hind III site: S':GCGGCAAGCTrnTGCAAAGCCTAGGC:3' (SEQ SD N0:55)
|1062| PCR amplification is performed using the SV40 promoter template present in the pB-galrpromoter plasmid obtained from Clontech. The resulting PCR fragment is digested with Xhol and Hind 111 and subcloned into BLSK2-. (Stratagene) Sequencing with the T7 and T3 primers confirms the insert contains the following sequence:
V:CTCGAOGGGACTTTCCCGGC,GAC.TTTC(XJGGGACTTirCGGGACTrrCCATCTGCCATCTCAATTAGTCAGCAACCATAGTCCCGC CXX^TAACTCCGCCCV\TCCCGCCCCTAACTCCGCCCAGTTCCGC;CCATTCTCCGCCCCATGGCTGACTAATrrTTTTTATTTATGCAGAG GCCGAGGCCGCCTCGGCCrCTGAGCTArrCCAGAAGTAGTGAGGAGGCTTITTrGGAGGCCTAGGCTTTTGCAAAAAGCrr:3' (SCQ IDNO:6I)
[1063| Next, replace the SV40 minimal promoter element present in the pSEAP2-promoter plasmid (Clontech) with this NF-KB/SV40 fragment
using Xhol and HindUl. However, this vector does not contain a neomycin resistance gene, and therefore, is not preferred for mammalian
expression systems.
11064] In order lo generate stable mammalian cell lines, the NF-K.B/SV4Q/SEAP cassette is removed from the above NF-KB/SEAP vector using
restriction enzymes Sal! and Nntl, and inserted into a vector containing neomycin resistance. Particularly, the NF-K.B/SV40/SEAP cassette was
inserted into pGFP-1 (Clontech), replacing the GFP gene, after restricting pGFP-1 with Sail and Notl.
|1065[ Once NF-KB/SV40/SEAP/Neo vector is created, stable Jurkal T-cclls are created and maintained according to the protocol described in
Example 25. Similarly, the method for assaying fusion proteins with these stable Jurkat T-cclls is also described in Example 25. As a positive
control, exogenous TNF alpha (0.1,1, 10 ng) is added to wells H9,1110, and H11, with a 5-10 fold activation typically observed.
EXAMPLE 29: Assay Identifying Myttoid Activity.
(1066J The following protocol is used to assess myeloid activity of an albumin fusion protein of the present invention by determining whether the fusion protein proliferates and/or differentiates myeloid cells- Myeloid cell activity is assessed using the GAS/SEAP/Neo construct produced in Example 24. Thus, factors that increase SEAP activity indicate the ability to activate the Jaks-STATS signal transduction pathway. The myetoid cell used in this assay is U937, a pre-monocyte cell line, although TF-I, HL60, or K.G1 can be used. |10671 To transiently transfect U937 cells wilh the OAS/SF.AP/Neo construct produced in Example 24, a DEAE-Dextran method (Kharbanda
et. a!., 1994, Cell Growth A Differentiation, 5:259-265) is used. First, harvest 2xl07 U937 cells and wash with PBS. The U937 cells are usually
grown in RPMI 1640 medium containing 10% heat-inactivated fetal bovine scrum (FDS) supplemented with 100 units/ml penicillin and 100 mg/ml
streptomycin.
[10681 Next, suspend the cells in 1 ml of 20 mM Tris-HCl (pH 7.4) buffer containing 0.5 mg/ml DEAE-Dextran, 6 ug GAS-SEAP2 plasmid
DMA, 140 mM NaCl, 5 mM KC1, 375 uMNa2HPO4.7H2O, 1 niM MgClj, and 675 uM CaClj. Incubate at 37 degrees C for 45 min.
|I069] Wash the cells with RPMI 1640 medium containing 10% FBS and then resuspend in 10 ml complete medium and incubate at 37 degree
C for 36 hr.
|1070] The GAS-SEAP/U937 stable cells are obtained by growing the cells in 400 ug/ml G418. The G418-free medium is used for routine
grcwth but every one to two months, the cells should be re-grown in 400 ug/ml G418 for couple of passages.
s |1011| These cells are tested by harvesting 1x10 cells (this is enough for ten 96-well plates assay) and wash with PBS. Suspend the cells in
200 ml above described growth medium, with a final density of 5x10* cells/rnl. Plate 200 ul cells per well in the 96-well plate (or lx]0s cells/well). |10721 Arid different concentrations of the fusion protein. Incubate it 37 degee C for 48 to 72 hr. As a positive control, 100 Unit/ml interfcron gamma can be used which is known In activate U937 cells. Over 30 fold induction is typically observed in the positive control wells. SGAP assay the supernatant according to methods known in the art and/or the protocol described in Example 25.
F.XA MPLE 30: Assay Identifying Changes in Small Molecule Concentration and Metnhrane Permeability.
j!073| Binding ofa ligand to a receptor is known to aller intracellular levels of small molecules, such as calcium, potassium, sodium, and pH, as well as alter membrane potential. These alterations can be measured in an assay to identify fusion proteins which bind to receptors ofa particular cell. Although the following protocol describes an assay for calcium, this protocol can easily be modified lo detect changes in potassium, sodium, pH, membrane potential, or any other small molecule which is detectable by a fluorescent probe.
[!074| The following assay uses FliJorometric Imaging Plate Reader ("FLIPR") to measure changes in fluorescent molecules (Molecular Probes) that bind small molecules, Cltarly, any fluorescent molecule detecting a small molecule can be used instead of the calcium fluorescent molecule, fluent (Molecular Probes, Inc.; catalog no. F-14202), used here. [1075) ':or adherent cells, seed the cells at 10,000 -20,000 cells/well in a Co-star black 96-well plate with clear bottom. The plate is incubated in
a iJUj mcuoator tor M nours. i ne iiuiierent cells are washed two times in Biolek washer with 200 ul of HBSS (Hank's Balanced Salt Solution)
leaving 100 ul of buffer after the final wash.
|1076| A stock solution of 1 mg/ml fiuo-4 is made in 10% pluronic acid DMSO. To load the cells with fluo-4, 50 ul of 12 ug/ml fluo-4 is
added to each well. The plate is incubated at 37 degrees C in a C02 incubator for 60 min. The plate is washed four times in the Biotek washer with
HBSS leaving 100 ul of buffer.
|1077) For non-adherent cells, the cells are spun down from culture media. Cells are re-suspended to 2-5xl06 cells/ml with HBSS in a 50-ml
conical tube. 4 ul of 1 mg/ml fluo-4 solution in 10% pluronic acid DMSO is added to each ml of cell suspension. The tube is then placed in a 37
degrees C water bath for 30-60 min. The cells are washed twice with HBSS, resuspended to IxlO6 cells/ml, and dispensed into a microplate, 100
ul/well. The plate is centrifuged at 1000 rpm for 5 min. The plate is then washed once in Denley Cell Wash with 200 ul, followed by an aspiration
step to 100 ul final volume.
11078| For a non-cell based assay, each well contains a fluorescent molecule, such as fluo-4 . The fusion protein of the invention is added to the
well, and a change in fluorescence is detected.
[1079J To measure the fluorescence of intracellular calcium, the FI.1PR is set for the following parameters: (1) System gain is 300-800 mW;
(2) Exposure time is 0.4 second; (3) Camera F/stop is F72; (4) Excitation is 488 nm; (5) Emission is 530 nm; and (6) Sample addition is 50 ul.
Increased emission at 530 nm indicates an extracellular signaling event caused by an albumin fusion protein of the present invention or a molecule
induced by an albumin fusion protein of the present invention, which has resulted in an increase in the intracellular Ca^"1" concentration. EXAMPLE 31: Assay Identifying Tvrosine Kinase Activity.
[1080] The Protein Tyrosine Kinases (PTK) represent a diverse group of transmembrane and cytoplasmic kinases. Within the Receptor Protein Tyrosine Kinase (RPTK) group are receptors fora range of mitogenic and metabolic growth factors including the PDGF, FGF, EOF, NGF, HGF and Insulin receptor subfamilies. In addition there are a large family of RPTKs for which the corresponding ligand is unknown. Ligands for RPTKs include mainly secreted small proteins, but also membrane-bound and extracellular matrix proteins.
[10811 Activation of RPTK by ligands involves ligand-mediated receptor dimerization, resulting in transphosphorylation of the receptor subimits and activation of the cytoplasmic tyrosine kinases. The cytoplasmic tyrosinc kinases include receptor associated tyrosinc kinases of the src-family (e.g., src, yes, Ick, lyn, fyn) and non-receptor linked and cytosolic protein tyrosine kinases, such as the Jak family, members of which mediate signal transduction triggered by the cytokine superfamily of receptors (e.g., the Interleukins, Interferons, GM-CSF, and Leptin).
11082] Because of the wide range of known factors capable of stimulating tyrosine kinase activity, identifying whether an albumin fusion protein of the present invention or a molecule induced by a fusion proetin of the present invention is capable of activating tyrosine kinase signal transduction pathways is of interest. Therefore, the following protocol is designed to identify such molecules capable of activating the tyrosine kinase signal transduction pathways.
|1083| Seed target cells (e.g., primary keratinocytes) at a density of approximately 25,000 cells per well in a 96 well Loprodyne Silent Screen Plates purchased from Nalge Nunc (Napcrviilc, 1L). The plates are sterilized with two 30 minute rinses with 100% ethanol, rinsed with water and dried overnight. Some plates are coated for 2 hr with 100 ml of cell culture grade type I collagen (50 mg/ml), gelatin (2%) or polylysine (50 mg/ml), all of which can be purchased from Sigma Chemicals (St. Louis, MO) or 10% Matrigel purchased from Becton Dickinson (Bedford.MA), or calf serum, rinsed with PBS and stored at 4 degree C. Cell growth on these plates is assayed by seeding 5,000 cells/well in growth medium and indirect quantitation of cell number through use of alamarBlue as described by the manufacturer Alamar Biosciences, Inc. (Sacramento, CA) after 48 hr. Falcon plate covers #3071 from Becton Dickinson (Bedford.MA) are used to cover the Loprodyne Silent Screen Plates. Falcon Microtcst III cell culture plates can also be used in some proliferation experiments.
[1084| To prepare extracts, A431 cells are seeded onto the nylon membranes of Loprodyne plates (20,000/200ml/well) and cultured overnight in complete medium. Cells arc quiesced by incubation in scrum-free basal medium for 24 hr. After 5-20 minutes treatment with EOF (60ng/ml) or a different concentrations of an albumin fusion protein of the invention, the medium was removed and 100 ml of extraction buffer ((20 mM HEPES pH 7.5, 0.15 M NaCl, 1% Triton X-100, 0.1% SDS.2 mM Na3V04, 2 mM Na4P2O7 and a cocktail of protease inhibitors (# 1836170) obtained
from Boeheringer Mannheim (Indianapolis, IN)) is added to each well and the plate is shaken on a rotating shaker for 5 minutes at 4°C. The plate is
then placed in a vacuum transfer manifold and the extract filtered through the 0.45 mm membrane bottoms of each well using house vacuum.
Extracts are collected in a 96-well catch/assay plate in the bottom of the vacuum manifold and immediately placed on ice. To obtain extracts
clarified by centrifugation, the content of each well, after detergent solubilization for 5 minutes, is removed and centrifuged for 15 minutes at 4
degree Cat 16,000 x g.
[1085] Test the filtered extracts for levels of tyrosine kinase activity. Although many methods of delecting tyrosine kinase activity are known,
one method is described here.
[ 1086) Generally, the tyrosine kinase activity of an albumin fusion prolcin of the invention is evaluated by determining its ability to
phosphorylatc a tyrosinc residue on a specific substrate (a biotinylatcd peptide). Biotinylated peptides that can be used for this purpose include
l-SM (corresponding to ammo acius u-20 of the cell division kinase cdc2-p34) and PSK2 (corresponding to amino acids 1-17 of gaslrin). Both peptides arc substrates for a range of tyrosine kinascs and are available from Boehringer Mannheim.
11087] Die tyrosine kinase reaction is set up by adding the following components in order. First, add lOul of SnM Hiotinylated Peptide, then lOul ATP/Mg2+ (5mM ATP/50mM MgCl2), then lOul of 5x Assay Buffer (40mM imidazole hydrochloride, pH7.3, 40 mM beta-glycerophosphate, ImM EGTA, lOOmM MgCl,. 5 mM MnCI2| 0.5 mg/ml BSA), then 5ul of Sodium Vanadate(lmM), and then 5ul of water. Mix the components
gently and preincubate the reaction mix at 30 degree C for 2 min. Initial (he reaction by adding lOul of the control enzyme or the filtered supernatant.
|1088| The tyrosine kinase assay reaction is then terminated by adding 10 ul of 120mm EDTA and place the reactions on ice.
11089) Tyrosine kinase activity is determined by transferring 50 ul aliquot of reaction mixture to a microtitcr plate (MTP) module and
incubating at 37 degree C for 20 min. This allows the streptavidin coated 96 well plate to associate with the biotinylated peptide. Wash the MTP
module with 300uiy\vell of PBS four times. Next add 75 ul of ami-phospotyrosine antibody conjugated to horse radish peroxidase(anti-P-Tyr-
POD(0.5u/ml)) to each well and incubate at 37 degree C for one hour. Wash the well as above.
|1090| Next add lOOul of peroxidase substrate solution (Boehringer Mannheim) and incubate at room temperature for at least 5 mins (up to 30
min). Measure the absorbance of the sample at 405 nm by using ELISA reader. The level of bound peroxidase activity is quantitated using an
ELISA reader and reflects the level of tyrosine kinase activity.
EXAMPLE 32: Assay fdentifyine Phosphorylarion Activity.
|1091] As a potential alternative and/or complement to the assay of protein tyrosine kinase activity described in Example 31, an assay which detects activation (phosphorylation) of major intraccllular signal transduction intermediates can also be used. For example, as described below one particular assay can detect tyrosine phosphorylation of the Erk-1 and Erk-2 kinases. However, phosphorylation of other molecules, such as Raf, JNK, p38 MAP, Map kinase kinase (MEK.), MEK kinase, Src, Muscle specific kinase (MuSK), IRAK, Tec, and Janus, as well as any other phosphosehne, phosphotyrosine, or phosphothreonine molecule, can be detected by substituting these molecules for Erk-1 or Erk-2 in the following assay.
|1092] Specifically, assay plates are made by coating the wells of a 96-well ELISA plate with O.lml of protein G (lug/ml) for 2 hr at room temp, (RT). The plates arc then rinsed with PBS and blocked with 3% BSA/PBS for 1 hr at RT. The protein 0 plates are then treated with 2 commercial monoclonal antibodies (lOOng/well) against Erk-1 and Erk-2 (1 hr at RT) (Santa Cruz Biotechnology). (To detect other molecules, this step can easily be modified by substituting a monoclonal antibody detecting any of the above described molecules.) After 3-5 rinses with PBS, the plates are stored at 4 degree C until use.
[1093] A431 cells arc seeded at 20,000/well in a 96-well Loprodyne filterplate and cultured overnight in growth medium. The cells are then starved for 48 hr in basal medium (DMOM) and then treated with EOF (fing/well) or varying concentrations of the fusion protein of the invention for 5-20 minutes. The cells are then sulubilized and extracts filtered directly into the assay plate.
(1094| Afler incubation with the extract for I hr at RT, the wells are again rinsed. As a positive control, a commercial preparation of MAP kinase (lOng/well) is used in place of A431 extract. Plates arc then treated with a commercial polyclonal (rabbit) antibody (lug/ml) which specifically recognizes the phosphorylated epitope of the Erk-1 and Hrk-2 kinases (1 hr at RT). This antibody is biotinylated by standard procedures. The bound polyclonal antibody is then quantitated by successive incubations with Europium-streptavidin and Europium fluorescence enhancing reagent in the Wai lac DELF1A instrument (time-resolved fluorescence). An increased fluorescent signal over background indicates a phosphorylation by the fusion protein of the present invention or a molecule induced by an albumin fusion protein of the present invention.
EXAMPLE 33: Phosphorytation Assay.
11095] In order to assay for phosphorylation activity of an albumin fusion protein of the invention, a phosphorylation assay as described in U.S. Patent 5,958,405 (which is herein incorporated by reference) is utilized. Briefly, phosphorylation activity may be measured by phosphorylation of a protein substrate using gamma-labeled "P-ATP and quantitation of the incorporated radioactivity using a gamma radioisotope counter. The fusion portein of the invention is incubated with the prolein substrate, "P-ATP, and a kinase buffer. The "P incorporated into the substrate is then separated from free J2P-ATP by electrophoresis, and the incorporated 32P is counted and compared to a negative control. Radioactivity counts above the negative control are indicative of phosphorylation activity of the fusion prolein.
EXAMPLE 34: Petecliiin of Phosphorylntion Activity (Activation) of an Albumin fusion Protein of the Invention in the Presence of Polvnei>tide l.ivands.
11096] Methods known in the art or described herein may be used to determine the phosphorylation activity of an albumin fusion protein of the invention. A preferred method of determining phosphorylation activity is by the use of the tyrosine phosphorylation assay as described in US 5,817,471 (incorporated herein by reference).
EXAMPLE 35: Assay for the Stimulation of Bone Marrow CD34+ Cell Proliferation. |1097| This assay is based on the ability of human CD34+ to proliferate in'the presence of hcmatopoietic growth factors and evaluates the
ability ot lusion proteins ot the inventor) to stimulate proliferation of CD34+ cells.
110981 It has been previously shown that most mature precursors will respond to only a single signal. More immature precursors require at least two signals to respond. Therefore, to test the effect of fusion proteins of the invention on hematopoietic activity of a wide range of progenitor cells, the assay contains a given fusion protein of the invention in the presence or absence of hematopoietic growth factors. Isolated cells are cultured for 5 days in the presence of Stem Cell Factor (SCF) in combination with tested sample. SCF alone has a very limited effect on the proliferation of bone marrow (BM) cells, acting in such conditions only as a "survival" factor. However, combined with any factor exhibiting stimulatory effect on these cells (e.g., IL-3), SCF will cause a synergistic effect. Therefore, if the tested fusion protein has a stimulatory effect on hematopoietic progenitors, such activity can be easily detected. Since normal BM cells have a low level of cycling cells, it is likely that any inhibitory effect of a given fusion protein might not be detected. Accordingly, assays for an inhibitory effect on progenitors is preferably tested in cells that are first subjected to in vitro stimulation with SCF+IL+3, and then contacted with the compound that is being evaluated for inhibition of such induced proliferation.
11099] Briefly, CD34+ cells are isolated using methods known in the art. The cells are thawed and resuspended in medium (QBSF 60 serum-free medium with 1% L-glutamine (500ml) Quality Biological, Inc., Gaithersburg, MD Cat# 160-204-101). After several gentle centrifugation steps at 200 x g, cells are allowed to rest for one hour. The cell count is adjusted to 2.5 x 10s cells/ml. During this time, 100 ul of sterile water is added to the peripheral wells of a 96-well plate. The cytokines that can be tested with an albumin fusion protein of the invention in this assay is rhSCF (R&O Systems, Minneapolis, MN, Cat# 255-SC) at 50 ng/ml alone and in combination with rhSCF and rhlL-3 (R&D Systems, Minneapolis, MN, Cat# 203-ML) at 30 ng/ml. After one hour, 10 ul of prepared cytokines, varying concentrations of an albumin fusion protein of the invention, and 20 ul of diluted cells are added to the media which is already present in the wells to allow for a final total volume of 100 ul The plates are then placed in a 37°C/5% CO2 incubator for five days.
[1100] Eighteen hours before the assay is harvested, 0.5 nCi/well of [311] Thymidine is added in a 10 ul volume to each well to determine the proliferation rate. The experiment is terminated by harvesting the cells from each 96-well plate to a filtermat using the Tomtec Harvester 96. After harvesting, the filtermats are dried, trimmed and placed into OmniFilter assemblies consisting of one OmniFilter plate and one OmniFilter Tray. 60 ul Microscint is added to each well and the plate sealed with TopSeal-A press-on sealing film A bar code 15 sticker is affixed to the first plate for counting. The sealed plates are then loaded and the level of radioactivity determined via the Packard Top Count and the printed data collected for analysis. The level of radioactivity reflects the amount of cell proliferation.
(1101) The studies described in this example test the activity of a given fusion protein to stimulate bone marrow CD34+ cell proliferation. One skilled in the art could easily modify the exemplified studies to test the activity of fusion porteins and polynucleotides of the invention (e.g., gene therapy) as well as agonists and antagonists thereof. The ability of an albumin fusion protein of the invention to stimulate the proliferation of bone marrow CIJ34+ cells indicates that the albumin fusion protein and/or polynucleotides corresponding to the fusion protein are useful for the diagnosis and treatment of disorders affecting the immune system and hematopoiesis. Representative uses are described in the "Immune Activity and "Infectious Disease" sections above, and elsewhere herein.
EXAMPLE 36: Assay for Extracellular Matrix Enhanced Cell Response (KMECR).
[1102] The objective of the Extracellular Matrix Enhanced Cell Response (EMECR) assay is to evaluate the ability of fusion proteins of the invention to act on hematopoietic stem cells in the context of the extracellular matrix (ECM) induced signal.
[1103| Cells respond to the regulatory factors in the context of signal(s) received from the surrounding microenvironment. For example, fibroblasts, and endothelial and epithelial stem cells fail to replicate in the absence of signals from the ECM. Hematopoietic stem cells can undergo self-renewal in the bone marrow, but not in in vilro suspension culture. Die ability of stem cells to undergo self-renewal in vitro is dependent upon iheir interaction with the stromal cells and the ECM protein fibronectin (fn). Adhesion of cells to fn is mediated by the avPi and CL,.p, integrin receptors, which are expressed by human and mouse hematopoietie stem cells. The factor(s) which integrate with the ECM environment and are responsible for stimulating stem cell self-renewal havea not yet been identified. Discovery of such factors should be of great interest in gene therapy and bone marrow transplant applications
|1104| Briefly, polystyrene, nun tissue culture treated, 96-well plates are coated with fn fragment at a coating concentration of 0.2 ug/ cm2. Mouse bone marrow cells are plated (1,000 cells/well) in 0.2 ml of serum-free medium. Cells cultured in the presence ofIL-3 ( 5 ng/ml) + SCF ( 50 ng/ml ) would serve as the positive control, conditions under which little self-renewal but pronounced differentiation of the stem cells is to be ixpected. Albumin fusion proleins of the invention are tested with appropriate negative controls in the presence and absence of SCF(5.0 ng/ml), •vhere volume of the administed composition containing the albumin fusion protein of the invention represents 10% of the total assay volume. The elated cells arc then allowed to grow by incubating in a low oxygen environment ( 5% CO2, 7% 02, and 88% N2 ) tissue culture incubator for 7 .lays. The number of proliferating cells within the wells is then quantitated by measuring thymidmc incorporation inlo cellular DNA. Verification >f the positive hits in the assay will require phenotypic characterization of the cells, which can be accomplished by scaling up of the culture system md using appropriate antibody reagents against cell surface antigens and FACScan.
[1105| If a particular fusion protein of the present invention is found to be a stimulator of hematopoietic progenitors, the fusion protein and polynucleotides corresponding to the fusion protein may be useful for example, in the diagnosis and treatment of disorders affecting the immune system and hematopoiesis. Representative uses are described in the "Immune Activity" and "Infectious Disease" sections above, and elsewhere herein. The fusion protein may also be useful in the expansion of stem cells and committed progenitors of various blood lineages, and in the differentiation and/or proliferation of various cell types.
|1106| Additionally, the albumin fusion proteins of the invention and polynucleotides encoding albumin fusion proteins of the invention, may also be employed to inhibit the proliferation and differentiation of hematopoietic cells and therefore may be employed to protect bone marrow stem cells from chcmotherapeutic agents dunng chemotherapy. This antiprolifcrative effect may allow administration of higher doses of chemothcrapeutic agents and, therefore, more effective chemotherapeutic treatment.
[1107] Moreover, fusion proteins of the invention and polynucleotides encoding albumin fusion proteins of the invention may also be useful for the treatment and diagnosis of hematopoietic related disorders such as, anemia, pancytopenia, leukopcnia, thrombocytopenia or leukemia, since stromal cells are important in the production of cells of hematopoietic lineages. The uses include bone marrow cell ex-vivo culture, bone marrow transplantation, bone marrow rcconstitution, radiotherapy or chemotherapy of neoplasia.
EXAMPLE 37: Human Dermal Fibroblast and Aortic Smooth Muscle Cell Proliferation.
11108| An albumin fusion protein of the invention is added to cultures of normal human dermal fibroblasts (NHDF) and human aortic smooth muscle cells (AoSMC) and two co-assays are performed with each sample. The first assay examines the effect of the fusion protein on the proliferation of normal human dermal fibroblasts (NHDF) or aortic smooth muscle cells (AoSMC). Aberrant growth of fibroblasts or smooth muscle cells is a part of several pathological processes, including fibrosis, and restenosis. The second assay examines IL6 production by both NHDF and SMC. IL6 production is an indication of functional activation. Activated cells will have increased production of a number of cytokines and other factors, which can result in a proinflammatory or imrnunomodulatory outcome. Assays are run with and without co-TNFa stimulation, in order to check for costimulatory or inhibitory activity.
11109| Briefly, on day 1, 96-wcll black plates are set up with 1000 cells/well (NHDF) or 2000 cells/well (AoSMC) in 100 ul culture media. NHDF culture media contains: Clonetics FB basal media, Img/ml hFGF, 5mg/ml insulin, 50mg/ml gentamycin, 2%FBS, while AoSMC culture media contains Clonetics SM basal media, 0.5 (ig/ml hEGF, 5mg/ml insulin, lug/ml hFGF, 50mg/ml gentamycin, 50 ug/ml Amphotericin B, 5%1'BS. After incubation at 37°C for at least 4-5 hours culture media is aspirated and replaced with growth arrest media. Growth arrest media for NHDF contains fibrohlast basal media, 50mg/ml gentamycin, 2% FBS, while growth arrest media for AoSMC contains SM basal media, 50mg/ml gentamycin, 50/ig/ml Amphotericin B, 0.4% FBS. Incubate at 37 °C until day 2.
|1110| On day 2, serial dilutions and templates of an albumin fusion protein of the invention are designed such that they always include media controls and known-protein controls. For both stimulation and inhibition experiments, proteins are diluted in growth arrest media. For inhibition experiments, TNFa is added to a final concentration of 2ng/ml (NHDF) or 5ng/ml (AoSMC). Add 1/3 vol media containing controls or an albumin fusion protein of the invention and incubate at 37 degrees C/5% COj until day 5.
|1111| Transfer 60(J from each well to another labeled 96-well plate, cover with a plate-sealer, and store at 4 degrees C until Day 6 (for IL6 KL.1SA). To the remaining 100 ;tl in the cell culture plate, aseptically add Alamar Blue in an amount equal to 10% of the culture volume (lOul). Return plates to incubator for 3 to 4 hours. Then measure fluorescence with excitation at 530nm and emission at 590nm using the CytoFluor. This yields the growth stimulation/inhibition data.
11112) On day 5, the IL6 EL1SA is performed by coating a 96 well plate with 50-100 ul/well of Anti-Human IL6 Monoclonal antibody diluted in PBS, pH 7.4, incubate ON at room temperature.
|1113] On day 6, empty the plates into the sink and blot on paper towels. Prepare Assay Buffer containing PBS with 4% BSA. Block the plates with 200 (il/well of Pierce Super Block blocking buffer in PBS for 1-2 hr and then wash plates with wash buffer (PBS, 0.05% Twcen-20). Blot plates on paper towels. Then add 50 ul/well of diluted Anti-Human IL-6 Monoclonal, Biotin-labeled antibody at 0.50 mg/ml. Make dilutions of IL-6 stock in media (30, 10, 3, 1, 0.3, 0 ng/ml). Add duplicate samples to top row of plate. Cover the plates and incubate for 2 hours at RTon shaker. |l 114| Plates are washed wilh wash buffer and blotted on paper towels. Dilute EU-labeled Streptavidin 1:1000 in Assay buffer, and add 100 ul/wcll. Cover the plate and incubate 1 h at RT. Plates are again washed with wash buffer and blotted on paper towels.
|1115| Add 100 ^I/well of Enhancement Solution. Shake for 5 minutes. Read the plate on the Wallac DELF1A Fluorometer. Readings from triplicate samples in each assay were tabulated and averaged.
|1116| A positive result in this assay suggests AoSMC cell proliferation and that the albumin fusion protein may be involved in dermal fibroblast pioliferation and/or smooth muscle cell proliferation. A positive result also suggests many potential uses of the fusion protein and polynucleotides encoding the albumin fusion protein. For example, inflammation and immune responses, wound healing, and angiogencsis, as detailed throughout this specification. Particularly, fusion proteins may be used in wound healing and dermal regeneration, as well as the promotion of vasculogenesis, both of the blood vessels and lymphatics. The growth of vessels can be used in the treatment of, for example, cardiovascular
diseases. Additionally, fusion proteins showing antagonistic activity in this assay may be useful in treating diseases, disorders, and/or conditions which involve angiogenesis by acting as an anti-vascular agent (e.g., ami-angiogenesis). These diseases, disorders, and/or conditions are known in the ait and/or are described iicil!h, such as, for example, malignancies, solid turuois, benign tumor?, for example hfmangionrc--, acoustic neuromas, neurofibromas, trachomas, and pyogenic granulomas; artherosclcric plaques; ocular angiogenic diseases, for example, diabetic retinopathy, retinopathy of prematurity, macular degeneration, comeal graft rejection, neovascular glaucoma, retrolental fibroplasia, rubeosis, retinoblastoma, uvictis and Pterygia (abnormal blood vessel growth) of the eye; rheumatoid arthritis; psoriasis; delayed wound healing; endometriosis; vasculogenesis; granulations; hypertrophic scars (keloids); nonunion fractures; scleroderma; trachoma; vascular adhesions; myocardial angiogenesis; coronary collaterals; cerebral collaterals; arteriovenous malformations; ischemic limb angiogenesis; Osier-Webber Syndrome; plaque neovascularization; telangiectasia; hemophiliac joints; angiofibroma; fibromuscular dysplasia; wound granulation; Crohn's disease; and atherosclerosis. Moreover, albumin fusion proteins that act as antagonists in this assay may be useful in treating anti-hyperproliferative diseases and/or anti-inflammatory known in the art and/or dcscnbed herein.
EXAMPLE 38: Cellular Adhesion Molecule (CAM) Expression on Endothelial Cells.
|1117| The recruitment of lymphocytes to areas of inflammation and angiogenesis involves specific receptor-ligand interactions between cell surface adhesion molecules (CAMs) on lymphocytes and the vascular endothelium. The adhesion process, in both normal and pathological settings, follows a multi-step cascade that involves intercellular adhesion moleculc-l (1CAM-1), vascular cell adhesion molccule-1 (VCAM-1), and endothelial leukocyte adhesion molecule-l (E-selectin) expression on endothelial cells (EC). The expression of these molecules and others on the vascular endothelium determines the efficiency with which leukocytes may adhere to the local vasculature and extravasate into the local tissue during the development of an inflammatory response. The local concentration of cytokines and growth factor participate in the modulation of the expression of these CAMs.
|1118| Briefly, endothelia! cells (e.g., Human Umbilical Vein Endothelial cells (HUVECs)) are grown in a standard 96 well plate to confluence, growth medium is removed from the cells and replaced with 100 jjl of 199 Medium (10% fetal bovine serum (FBS)). Samples for testing (containing an albumin fusion protein of the invention) and positive or negative controls are added to the plate in triplicate (in 10 ftl volumes). Plates are then incubated at 37°C for either 5 h (selectin and integrin expression) or 24 h (integrin expression only). Plates are aspirated to remove medium and 100 /il of 0.1% paraformaldchyde-PBS(with Ca++ and Mg-H-) is added to each well. Plates are held at 4°C for 30 min. Fixative is removed from the wells and wells arc washed 1X with PBS(+Ca,Mg) + 0.5% BSA and drained. 10 fj! of diluted primary antibody is added to the test and control wells. Anti-lCAM-1-Biotin, Anti-VCAM-1-Biotin and Anti-E-selectin-Biotin are used at a concentration of 10 fig/ml (1:10 dilution of 0.1 mg/rnl stock antibody) Oils are incubated at 37°C for 30 min. in a humidified environment. Wells are washed three times with PBS(+Ca,Mg) + 0.5% BSA. 20 jl\ of diluted ExtrAvidin-Alkaline Phosphatase (1:5,000 dilution, referred to herein as the working dilution) are added to each well and incubated at 37°C for 30 min. Wells are washed three times with PBS(+Ca,Mg)+0.5% BSA. Dissolve 1 tablet of p-Nitrophenol Phosphate pNPP per 5 ml of glycine buffer (pH 10.4). 100 n\ of pNPP substrate in glycine buffer is added to each test well. Standard wells in triplicate are prepared from the working dilution of the ExtrAvidin-Alkaline Phosphotase in glycine buffer: 1:5,000 (10°) > 10"os > 10"' > 10"". 5/il of each dilution is added to triplicate wells and the resulting AP content in each well is 5.50 ng, 1.74 ng, 0.55 ng, 0.18 ng. 100/ilof pNNP reagent is then added to each of the standard wells. The plate is incubated at 37°C for 4h. A volume of 50 ft] of 3M NaOH is added to all wells. Tlie plate is read on a plate reader at 405 nm using the background subtraction option on blank wells filled with glycine buffer only. Additionally, the template is set up to indicate the concentration of AP-conjugatc in each standard well [ 5.50 ng; 1.74 ng; 0.55 ng; 0.18 ng]. Results are indicated as amount of bound AP-conjugatc in each sample.
EXAMPLE 39: Alomar Blue Endotlielial Cells Proliferation Assay.
[1119] This assay may be used to quantitatively determine protein mediated inhibition of bFGF-induced proliferation of Bovine Lymphatic Endolhelial Cells (LECs), Bovine Aortic Endothelial Cells (BAF.Cs) or Human Microvascular Uterine Myometrial Cells (UTMECs). This assay incorporates a fluorometric growth indicator based on detection of metabolic activity. A standard Alamar Blue Proliferation Assay is prepared in EGM-2MV with 10 ng /ml of bFGF added as a source of endothelial cell stimulation. This assay may be used with a variety of endothelial cells with slight changes in growth medium and cell concentration. Dilutions of protein batches to be tested are diluted as appropriate. Serum-free medium (G1BCO SFM) without bFGF is used as a non-stimulated control and Angiostatin orTSP-1 are included as a known inhibitory controls. 11120] Briefly, LEG, B AECs or UTMECs are seeded in growth media at a density of 5000 to 2000 cells/well in a 9(5 well plate and placed at 37 degreesC overnight. After the overnight incubation of the cells, the growth media is removed and replaced with G1BCO EC-SFM. The cells are treated with the appropriate dilutions of an albumin fusion protein of the invention or control protein sample(s) (prepared in SFM ) in triplicate wells with additional bFGF to a concentration of 10 ng/ ml. Once the cells have been treated with the samples, the plate(s) is/are placed back in the 37° C incubator for three days. After three days 10 rnl of stock alamar blue (Biosource Cat# DAL1100) is added to each well and the plate(s) is/are placed back in the 37°C incubator for four hours. '[Tie plate(s) are then read at 530nm excitation and 590nm emission using the CytoFIuor fluorescence reader. Direct output is recorded in relative fluorescence units.
|1121| Alamar blue is an oxidation-reduction indicator that both fluoresces and changes color in response to chemical reduction of growth
medium resulting from cell growth. As cells grow in culture, innate metabolic activity results in a chemical reduction of the immediate surrounding environment. Reduction related to growth causes the indicator to change from oxidized (non-fluorescent blue) form to reduced (fluorescent red) form (i.e., stimulated proliferation v/il! produce a stronger signal and inhibited proliferation will produce a weaker signal and the total signal is proportional to the total number of cells as well as their metabolic activity). The background level of activity is observed with the starvation medium alone. This is compared to the output observed from the positive control samples (bFGF in growth medium) and protein dilutions.
EXAMPLE 40: Detection of Inhibition of a Mixed Lymphocyte Reaction.
[1122] This assay can be used to detect and evaluate inhibition of a Mixed Lymphocyte Reaction (MLR) by fusion proteins of the invention. Inhibition of a MLR may be due to a direct effect on cell proliferation and viability, modulation of costimulatory molecules on interacting cells, modulation of adhesiveness between lymphocytes and accessory cells, or modulation of cytokine production by accessory cells. Multiple cells may be targeted by the albumin fusion proteins that inhibit MLR since the peripheral blood mononuclear fraction used in this assay includes T, B and natural killer lymphocytes, as well as monocytes and dendritic cells.
|1123] Albumin fusion proteins of the invention found to inhibit the MLR may find application in diseases associated with lymphocyte and monocyte activation or proliferation. These include, but are not limited to, diseases such as asthma, arthritis, diabetes, inflammatory skin conditions, psoriasis, eczema, systemic lupus erythematosus, multiple sclerosis, glomerulonephritis, inflammatory bowel disease, crohn's disease, ulccrative colitis, arteriosclerosis, cirrhosis, graft vs. host disease, host vs. graft disease, hepatitis, leukemia and lymphoma.
|1124| Briefly, PBMCs from human donors are purified by density gradient centrifugation using Lymphocyte Separation Medium (LSMS, density 1.0770 g/ml, Organon Teknika Corporation, West Chester, PA). PBMCs from two donors are adjusted to 2 x I06 cells/ml in RPMI-1640 (Life Technologies, Grand Island, NY) supplemented with 10% PCS and 2 mM glutamine. PBMCs from a third donor is adjusted to 2 x 10s cells/ml. Fifty microliters of PBMCs from each donor is added to wells of a 96-well round bottom microtiter plate. Dilutions of the fusion protein test material (50 p.1) is added in triplicate to microtiter wells. Test samples (of the protein of interest) are added for final dilution of 1:4; rhulL-2 (R&D Systems, Minneapolis, MN, catalog number 202-IL) is added to a final concentration of 1 ug/ml; anti-CD4 mAb (R&D Systems, clone 34930.11, catalog number MAB379) is added to a final concentration of 10 ug/ml. Cells are cultured for 7-8 days at 37°C in 5% CO2, and 1 uC of [JH] thymidine is added to wells for the last 16 hrs of culture. Cells are harvested and thymidinc incorporation determined using a Packard TopCount. Data is expressed as the mean and standard deviation of triplicate determinations.
(1125| Samples of the fusion protein of interest are screened in separate experiments and compared to the negative control treatment, anti-CD4 mAb, which inhibits proliferation of lymphocytes and the positive control treatment, IL-2 (either as recombinant material or supernatant), which enhances proliferation of lymphocytes.
EXAMPLE 41: Assays for Protease Activity.
|l I26| The following assay may be used to assess protease activity of an albumin fusion protein of the invention.
|1127] Gelatin and casein zymography are performed essentially as described (Heusen et z\.,Anal. Biochem., 102:196-202 (1980); Wilson et al., Journal of Urology, 149:653-658 (1993)). Samples are run on 10% polyacryamide/0.1% SDS gels containing 1% gelain orcasein, soaked in 2.5% triton at room temperature for I hour, and in 0.1 M glycine, pH 8.3 at 37°C 5 to 16 hours. After staining in amido black areas of proteolysis apear as clear areas agains the blue-black background. Trypsin (Sigma T8642) is used as a positive control.
|1128) Protease activity is also determined by monitoring the cleavage of n-a-benzoyl-L-arginine ethyl ester (BAEE) (Sigma B-4500. Reactions are set up in (25mMNaPO4,lmM EDTA, and ImM BAEE), pH 7.5. Samples are added and the change in adsorbance at 260nm is monitored on the Beckman DU-6 spectrophotometer in the time-drive mode. Trypsin is used as a positive control.
|1129| Additional assays based upon the release of acid-soluble peplides from casein or hemoglobin measured as adsorbance at 280 nm or colorimetrically using the Folin method arc performed as described in Bergmeyer, et al., Methods of Enzymatic Analysis, 5 (1984). Other assays involve the solubilization of chromogenic substrates (Ward, Applied Science, 251-317 (1983)).
EXAMPLE 42: IdentiMne Scrinc Protease Siihxtrate Specificity.
|l 130| Methods known in the art or described herein may be used to determine the substrate specificity of the albumin fusion proteins of the present invention having serine protease activity. A preferred method of determining substrate specificity is by the use of positional scanning synthetic combinatorial libraries as described in GB 2 324 529 (incorporated herein in its entirety).
EXAMPLE 43: Lieanil Binding Assays.
[11311 The following assay may be used to assess ligand binding activity of an albumin fusion protein of the invention.
[II32| Ligand binding assays provide a direct method for ascertaining receptor pharmacology and are adaptable to a high throughput format. The purified ligand for an albumin fusion protein of the invention is radiolabeled to high specific activity (50-2000 Ci/mmol) for binding studies. A determination is then made that the process of radiolabeling does not diminish the activity of the ligand towards the fusion protein. Assay conditions for buffers, ions, pH and other modulators such as nucleotides arc optimized to establish a workable signal to noise ratio for both membrane and whole cell polypeptide sources. For these assays, specific polypeptide binding is defined as total associated radioactivity minus the radioactivity
measured in the presence ot an excess of unlabeled competing ligand. Where possible, more than one competing ligand is used to define residual nonspecific binding.
hXA MPLF. 44: Functional As±av in Xtnopus Poetics.
I1133J Capped RNA transcripts from linearized plasmid templates encoding an albumin fusion protein of the invention is synthesized in vitro with RNA polymerases in accordance with standard procedures. In vitro transcripts are suspended in water at a final concentration of 0.2 mg/mi. Ovarian lobes are removed from adult female toads, Stage V defolliculated oocytes are obtained, and RNA transcripts (10 ng/oocytc) are injected in a 50 nl bolus using a microinjection apparatus. Two electrode voltage clamps are used to measure the currents from individual Xenopus oocytes in response fusion protein and polypeptide agonist exposure. Recordings are made in Ca2+ free Barth's medium at room temperature. The Xenopus system can be used to screen known ligands and tissue/cell extracts for activating ligands.
EXAMPLE 45: MicrophysiometricAssays.
(1134| Activation of a wide variety of secondary messenger systems results in extrusion of small amounts of acid from a cell. The acid formed is largely as a result of the increased metabolic activity required to fuel the intracellular signaling process. The pH changes in the media surrounding the cell are very small but are detectable by the CYTOSENSOR microphysiometer (Molecular Devices Ltd., Menlo Park, Calif). The CYTOSENSOR is thus capable of detecting the ability of an albumin fusion protein of the invention to activate secondary messengers that are coupled to an energy utilizing intracellular signaling pathway.
EXAMPLE 46: Extract/Cell Supernatant Screening.
[1135] A large number of mammalian receptors exist for which there remains, as yet, no cognate activating ligand (agonist). Thus, active ligands for these receptors may not be included within the ligands banks as identified to date. Accordingly, the albumin fusion proteins of the invention can also be functionally screened (using calcium, cAMP, microphysiomeler, oocyte electrophysiology, etc., functional screens) against tissue extracts to identify natural ligands for the Therapeutic protein portion and/or albumin protein portion of an albumin fusion protein of the invention. Extracts that produce positive functional responses can be sequentially subfractionated until an activating ligand is isolated and identified.
EXAMPLE 47: ATP-bindine assay.
|1136] The following assay may be used to assess ATP-binding activity of fusion proteins of the invention.
[1137] ATP-binding activity of an albumin fusion protein of the invention may be detected using the ATP-binding assay described in U.S. Patent 5,858,719, which is herein incorporated by reference in its entirety. Briefly, ATP-binding to an albumin fusion protein of the invention is measured via photoaffinity labeling with 8-azido-ATP in a competition assay. Reaction mixtures containing 1 mg/ml of ABC transport protein are incubated with varying concentrations of ATP, or the non-hydrolyzable ATP analog adenyl-5'-imidodiphosphate for 10 minutes at 4°C. A mixture of 8-azido-ATP (Sigma Chem. Corp., St. Louis, MO.) plus 8-37.ido-ATP ("P-ATP) (5 mCi/umol, ICN, Irvine CA.) is added to a final concentration of 100 uM and 0.5 ml aliquots are placed in the wells of a porcelain spot plate on ice. The plate is irradiated using a short wave 254 nm UV lamp at a distance of 2.5 cm from the plate for two one-minute intervals with a one-minute cooling interval in between. The reaction is stopped by addition of dithiothreitol to a final concentration of 2mM. The incubations are subjected to SDS-PAGE electrophorcsis, dried, and autoradiographed. Protein bands corresponding to the albumin fusion proteins of the invention are excised, and the radioactivity quantified. A decrease in radioactivity with increasing ATP or adenly-5'-imidodiphosphate provides a measure of ATP affinity to the fusion protein.
EXAMPLE 48: Identification Of Signal Transduction Proteins That Interact With An albumin fusion protein Of The Present Invention.
|1I3S| Albumin fusion proteins of the invention may serve as research tools for the identification, characterization and purification of signal transduction pathway proteins or receptor proteins. Briefly, a labeled fusion protein of the invention is useful as a reagent for the purification of molecules with which it interacts. In one embodiment of affinity purification, an albumin fusion protein of the invention is covalently coupled to a chromatography column. Cell-free extract derived from putative target cells, such as carcinoma tissues, is passed over the column, and molecules with appropriate affinity bind to the albumin fusion protein. The protein complex is recovered from the column, dissociated, and the recovered molecule subjected to N-terminal protein sequencing. This amino acid sequence is then used to identify the captured molecule or to design degenerate oligonuclcotide probes for cloning the relevant gene from an appropriate cDNA library.
EXAMPLE 49: IL-6 Rioassay.
|I139] A variety of assays arc known in the art for testing the proliferative effects of an albumin fusion protein of the invention. For example, one such asssay is ihe IL-6 Bioassay as described by Marz et til. (Proc. Natl. Acad. Sci., U.S.A., 95:3251-56 (1998), which is herein incorporated by reference). After 68 hrs. at 37°C, Ihc number of viable cells is measured by adding the tctrazolium salt thiazolyl blue (MTT) and incubating for a further 4 hrs. at 37°C. D9 cells are lysed by SDS and optical density is measured at 570 nm. Controls containing IL-6 (positive) and no cytokine (negative) are Briefly, IL~6 dependent B9 murine cells are washed three times in IL-6 free medium and plated at a concentration of 5,000 cells per well in 50 jil, and 50 jil of fusion protein of the invention is added, utilized. Enhanced proliferation in the test sample(s) (containing an albumin fusion protein of the invention) relative to the negative control is indicative of proliferative effects mediated by (he fusion protein.
h.KAMl'Lk >».• Support of Chicken Embryo Neuron Survival,
[11401 To test whether sympathetic neuronal cell viability is supported by an albumin fusion protein of the invention, the chicken embryo ncuronal survival assay of Senaldi cl al may be utilized (Proc. Nat! Acad. Sri. V.'i.A.. 96:\ 1458-63 (1998), which is herein incorporated by reference). Briefly, motor and sympathetic neurons are isolated from chicken embryos, resuspended in LI 5 medium (with 10% FCS, glucose, sodium selenite, progesterone, conalbumin, putrescine, and insulin; Life Technologies, Rockville, MD.) and Dulbecco's modified Eagles medium [with 10% FCS, glutarnine, penicillin, and 25 mM Hcpcs buffer (pH 7.2); Life Technologies, Rockville, MD.], respectively, and incubated at 37°C in 5% COj in the presence of different concentrations of the purified fusion protein of the invention, as well as a negative control lacking any cytokine. After 3 days, neuron survival is determined by evaluation of cellular morphology, and through the use of the colorimetric assay of Mosmann (Mosmann, T., J. Immunol. Methods, (55:55-63 (1983)). Enhanced neuronal cell viability as compared to the controls lacking cytokine is indicative of the ability of the albumin fusion protein to enhance the survival of neuronal cells.
EXAMPLE 51: Assay for Phosphalase Activity.
(11411 The following assay may be used to assess scrine/threonine phosphatase (PTPase) activity of an albumin fusion protein of the invention. 11142| In order to assay for serine/threonine phosphatase (PTPase) activity, assays can be utilized which are widely known to those skilled in the art. For example, the serine/threonine phosphatase (PSPase) activity of an albumin fusion protein of the invention may be measured using a PSPase assay kit from New England Biolabs, Inc. Myelin basic protein (MyBP), a substrate for PSPase, is phosphorylated on serine and threonine residues with cAMP-dependent Protein Kinase in the presence of ["PJATP. Protein serine/threonine phosphatase activity is then determined by measuring the release of inorganic phosphate from 32P-labeled MyBP.
EXAMPLE 52: Interaction ofSerineflhreonine Phosphatases with other Proteins.
|1143| Fusion proteins of the invention having serine/threonine phosphatase activity ( e.g., as determined in Example 51) arc useful, for example, as research tools for the identification, characterization and purification of additional interacting proteins or receptor proteins, or other signal transduction pathway proteins. Briefly, a labeled fusion protein of the invention is useful as a reagent for the purification of molecules with which it interacts. In one embodiment of affinity purification, an albumin fusion protein of the invention is covalently coupled to a chromatography column. Cell-free extract derived from putative target cells, such as neural or liver cells, is passed over the column, and molecules with appropriate affinity bind to the fusion protein. The fusion protein -complex is recovered from the column, dissociated, and the recovered molecule subjected to N-terminal protein sequencing. This amino acid sequence is then used to identify the captured molecule or to design degenerate oligonucleotide probes for cloning the relevant gene from an appropriate cDNA library.
EXAMPLE 5J: Assaying fur He.paranase Activity.
[I144J There a numerous assays known in the art that may be employed to assay for heparanase activity of an albumin fusion protein of the invention. In one example, hcparanase activity of an albumin fusion protein of the invention, is assayed as described by Vlodavsky et al., (Vlodavsky ct al., Nat. Med., 5:793-802 (1999)). Briefly, cell lysates, conditioned media, intact cells (1 x 10* cells per 35-mm dish), cell culture-supernatant, or purified fusion protein are incubated for 18 hrs at 37°C, pH 6.2-6.6, with 35S-labeled ECM or soluble ECM derived peak I proteoglycans. The incubation medium is centrifuged and the supernatant is analyzed by gel filtration on a Sepharose CL-6B column (0.9 x 30 cm). Fractions are eluted with PBS and their radioactivity is measured. Degradation fragments of heparan sulfate side chains are eluted from Sepharose 6tt at 0.5 EXAMPLE 54: Immobilization ofbiomoleciiles.
|1145] This example provides a method for the stabilization of an albumin fusion protein of the invention in non-host cell lipid hi layer constucts (sec, e.g., Dieri ct al., Nature Biotech 17:1105-1108 (1999), hereby incorporated by reference in its entirety herein) which can be adapted for the study effusion proteins of the invention in the various functional assays described above. Briefly, carbohydrate-specific chemistry for biotinylation is used to confine a biotin tag to an albumin fusion protein of the invention, thus allowing uniform orientation upon immobilization. A 50uM solution of an albumin fusion protein of the invention in washed membranes is incubated with 20 mM NalO4 and 1.5 mg/ml (4mM) BACH or 2 mg/ml (7.5mM) biotin-hydrazide for 1 hr at room temperature (reaction volume, 150ul). Then the sample is dialyzed (Pierce Slidealizer Cassetl, 10 kOa cutoff; Pierce Chemical Co , Rockford IL) at 4C first for 5 h, exchanging the buffer after each hour, and finally for 12 h against 500 ml buffer R (0.15 M NaCI, 1 mM MgC12, 10 mM sodium phosphate, pi 17). Just before addition into a cuvette, the sample is diluted 1:5 in buffer ROG50 (Buffer R supplemented with 50 mM octylglucoside).
EXAMPLE 55; Assays for Afetalloproteinase Activity.
|1146) Metalloproteinases are peptide hydrolases which use metal ions, such as Zn2*, as the catalytic mechanism. Metalloproteinase activity of an albumin fusion protein of the present invention can he assayed according to methods known in the art. The following exemplary methods arc provided:
Prateo/vsis ofalpha-2-macrnzIobuIin
|M47| To confirm protease activity, a purified fusion protein of the invention is mixed with the substrate alpha-2-macroglobulin (0.2 unit/ml;
boehnnger Mannneim, ucrmanyj in ix assay buffer (50 mM HEPES, pH 7.5, 0.2 M NaCI, 10 rnM CaCl2, 25 \iM ZnCI2 and 0.05% Brij-35) and incubated at 37°C for 1-5 days. Trypsin is used as positive control. Negative controls contain only alpha-2-macroglobulin in assay buffer. The samples arc collected and boiled in SDS PAGE sample buffer containing 5% 2-merraptoethanol for 5-min, then loaded onto 8% SDS-polyacrylamide gel. After electrophoresis the proteins arc visualized by silver staining. Proteolysis is evident by the appearance of lower molecular weight bands as compared to the negative control.
Inhibition ofalpha-2-nmcroelobulin proteolvsis bv inhibitors of metalloproleinases
111481 Known mctalloproteinase inhibitors (metal ehelators (EDTA, EGTA, AND HgCl2), peptide metalloproteinase inhibitors (T1MP-1 and TIMP-2), and commercial small molecule MMP inhibitors) may also be used to characterize the proteolytic activity of an albumin fusion protein of the invention. Three synthetic MMP inhibitors that may be used are: MMP inhibitor I, [ICso = 1.0 u.M against MMP-1 and MMP-8; ICso = 30 \M against MMP-9; IC!0 = 150 pjvl against MMP-3]; MMP-3 (stromelysin-1) inhibitor I [IC50 = 5 uM against MMP-3], and MMP-3 inhibitor II [Ki = 130 nM against MMP-3); inhibitors available through Calbiochcm, catalog # 444250, 444218, and 444225, respectively). Briefly, different concentrations of the small molecule MMP inhibitors are mixed with a purified fusion protein of the invention (50u,g/rnl) in 22,9 u.1 of Ix HEPES buffer (50 mM HEPES, pH 7.5, 0.2 M NaCI, 10 mM CaCl2> 25 uJvl ZnCl2 and 0.05%Bnj-35) and incubated at room temperature (24 °C) for 2-hr, then 7.1 ul of substrate alpha-2-macroglobulin (0.2 unit/ml) is added and incubated at 37°C for 20-hr. The reactions are stopped by adding 4x sample buffer and boiled immediately for 5 minutes. After SDS-PAGE, the protein bands are visualized by silver stain.
Synthetic Fluoro^enic Peptide Substrates Cleuvcixe Assay
|1149] The substrate specificity for fusion proteins of the invention with demonstrated mctalloproteinase activity may be determined using techniques knonw in the art, such as using synthetic fluorogenic peptide substrates (purchased from BACHEM Bioscience Inc). Test substrates include, M-1985, M-2225, M-2105, M-2110, and M-2255. The first four are MMP substrates and the last one is a substrate of tumor necrosis factor-cc (TNF-a) converting enzyme (TACE). These substrates are preferably prepared in 1:1 dimethyl sulfoxide (DMSO) and water. The stock solutions arc 50-500 uM. Fluorescent assays are performed by using a Perkin Elmer LS 508 luminescence spectrometer equipped with a constant temperature water bath. The excitation X is 328 nm and the emission X is 393 nm. Briefly, the assay is carried out by incubating 176 u.1 Ix HEPES buffer (0.2 M NaCI, 10 mM CaCl2, 0.05% Brij-35 and 50 mM HEPES, pH 7.5) with 4 ul of substrate solution (50 uM) at 25 "C for 15 minutes, and then adding 20 u.1 of a purified fusion protein of the invention into the assay cuvett. The final concentration of substrate is I uM. Initial hydrolysis rates are monitored for 30-min.
EXAMPl.K 56: Occurrence of Diabetes in NOD Mice.
[1150| Female NOD (non-obese diabetic) mice are characterized by displaying IDDM with a course which is similar to that found in humans, although the disease is more pronounced in female than male NOD mice. Hereinafter, unless otherwise stated, the term "NOD mouse" refers to a female NOD mouse. NOD mice have a progressive destruction of beta cells which is caused by a chronic autoimmune disease. Thus, NOD mice begin life with euglycemia, or normal blood glucose levels. By about 15 to 16 weeks of age, however, NOD mice start becoming hyperglycemic, indicating the destruction of the majority of their pancreatic beta cells and the corresponding inability of the pancreas to produce sufficient insulin. Thus, both the cause and the progression of the disease are similar to human IDDM patients.
|1151| In vivo assays of efficacy of the immunization regimens can be assessed in female NOD/LU mice (commercially available from The Jackson laboratory, Bar Harbor, Me.). In the literature, it's reported that 80% of female mice develop diabetes by 24 weeks of age and onset of insulitis begins between 6-8 weeks age. NOD mice are inbred and highly responsive to a variety of immunoregulatory strategies. Adult NOD mice (6-8 weeks of age) have an average mass of 20-25 g.
|1152| These mice can be either untreated (control), treated with the therapeutics of the subject invention (e.g., albumin fusion proteins of the invention and fragments and variants thereof), alone or in combination with other therapeutic compounds stated above. The effect of these various treatments on the progression of diabetes can be measured as follows:
|1I53| At 14 weeks of age, the female NOD mice can be phenotypcd according to glucose tolerance. Glucose tolerance can be measured with the intraperitoneal glucose tolerance test (IPGTT). Briefly, blood is drawn from the paraorbital plexus at 0 minutes and 60 minutes afler the intrapcritoncal injection of glucose (1 g/kg body weight). Normal tolerance is defined as plasma glucose at 0 minutes of less than 144 rng %, or at 60 minutes of less than 160 mg %. Blood glucose levels are determined with a Glucometcr Elite apparatus.
[1154] Based upon this phenotypic analysis, animals can be allocated to the different experimental groups. In particular, animals with more elevated blood glucose levels can be assigned to the impaired glucose tolerance group. The mice can be fed ad libitum and can be supplied with acidified water (pH 2.3).
(1155] The glucose tolerant and intolerant mice can be further subdivided into control, albumin fusion proteins of the subject invention, and albumin fusion proteins/therapeutic compounds combination groups. Mice in the control group can receive an interperitoneal injection of vehicle daily, six times per week. Mice in the albumin fusion group can receive an interperitoneal injection of the therapeutics of the subject invention (e.g., albumin fusion proteins of the invention and fragments and variants thereof) in vehicle daily, six times per week. Mice in the albumin fusion
proteins/therapeutic compounds combination group can receive both albumin fusion proteins and combinations of therapeutic compounds as
described above.
[11561 The level of urine glucose in the NOD mice can be determined on a bi-weekly basis using Ijhstix (Bayer Diapnostics, Hampshire,
England). Weight and fluid intake can also be determined on a bi-weekly basis. The onset of diabetes is defined after the appearance of glucosuria
on two consecutive determinations. After 10 weeks of treatment, an additional IPGTT can be performed and animals can be sacrificed the following
day.
[1157] Over the 10 week course of treatment, control animals in both the glucose tolerant and glucose intolerant groups develop diabetes at a
rate of 60% and 86%, respectively (see US patent No. 5,866,546, Gross et a!.). Thus, high rates of diabetes occur even in NOD mice which arc
initially glucose tolerant if no intervention is made.
|1158] Results can be confirmed by the measurement of blood glucose levels in NOD mice, before and after treatment. Blood glucose levels are
measured as described above in both glucose tolerant and intolerant mice in all groups described.
[1159] In an alternative embodiment, the therapeutics of the subject invention (e.g., specific fusions disclosed as SEQ ID NO:Y and fragments
and variants thereof) can be quantified using spectrometric analysis and appropriate protein quantities can be resuspended prior to injection in 50
.mil.I phosphate buffered saline (PBS) per dose. Two injections, one week apart, can be administered subcutaneously under the dorsal skin of each
mouse. Monitoring can be performed on two separate occasions prior to immunization and can be performed weekly throughout the treatment and
continued thereafter. Urine can be tested for glucose every week (Keto-Diastix.RTM.; Miles Inc., Kankakee, 111.) and glycosuric mice can be
checked for serum glucose (ExacTech.RTM., MediSense, Inc., Waltham, Mass.). Diabetes is diagnosed when fasting glycemia is greater than
2.5g/L.
EXAMPLE 57: Histolocical Examination of NOD Mice.
[1160| Histological examination of tissue samples from NOD mice can demonstrate the ability of the compositions of the present invention, and/or a combination of the compositions of the present invention with other therapeutic agents for diabetes, to increase the relative concentration of beta cells in the pancreas. The experimental method is as follows:
[11611 The mice from Example 56 can be sacrificed at the end of the treatment period and tissue samples can be taken from the pancreas. The samples can be fixed in 10% formalin in 0.9% saline and embedded in wax. Two sets of 5 serial 5 .mu.m sections can he cut for immunolabelling at a cutting interval of 150 .mu.m. Sections can be immunolabclled for insulin (guinea pig anti-insulin antisera dilution 1:1000, ICN Thames U.K.) and glucagon (rabbit anti-pancreatic glucagon antisera dilution 1:2000) and detected with peroxidase conjugated anti-guinea pig (Dako, High Wycombe, U.K.) or peroxidase conjugated anti-rabbit antisera (dilution 1:50, Dako).
|1162] The composition of the present invention may or may not have as strong an effect on the visible mass of beta cells as it does on the clinical manifestations of diabetes in glucose tolerant and glucose intolerant animals.
KXAMPLK 58: In vivo Mouse Model ofNIDDM.
[1163] Male C57BL/6J mice from Jackson Laboratory (Bar Harbor, ME) can be obtained at 3 weeks of age and fed on conventional chow or diets enriched in either fat (35.5% wfwt; Bioscrv.Frenchtown, NJ) or fructose (60% wt/wt; Harlan Teklad, Madison, WI). The regular chow is composed of 4.5% wt/wt fat, 23% wt/wt protein, 31.9% wt/wt starch, 3.7% wt/wt fructose, and 5.3% wt/wt fiber. The high-fat (lard) diet is composed of 35.5% wt/wt fat, 20% wt/wt protein, 36.4% wt/wt starch, 0.0% wt/wt fructose, and 0.1% wt/wt fiber. The high-fructose diet is composed of 5% wt/wt fat, 20% wt/wt protein, 0.0% wt/wt starch, 60% wt/wt fructose, and 9.4% wt/wt fiber. The mice may be housed no more than five per cage at 22° +/- 3°C temperature- and 50% +/- 20% humidity-controlled room with a 12-hour light (6 am to 6 pm)/dark cycle (Luo et al., 1998, Metabolism 47(6): 663-8, "Nongenetic mouse models of non-insulin-dependent diabetes mellitus"; Larsen et al., Diabetes 50(11): 2530-9 (2001), "Systemic administration of the long-acting GLP-I derivative NN2211 induces lasting and reversible weight loss in both normal and obese rats"). After exposure to the respective diets for 3 weeks, mice can be injected intraperitoneally with either streptozotocin, "STZ" (Sigma, St. Louis, MO), at 100 mg/kg body weight or vehicle (0.05 mol/L citric acid, pH 4.5) and kept on the same diet for the next 4 weeks. Under nonfasting conditions, blood is obtained 1, 2, and 4 weeks post-STZ by nipping the distal part of the tail. Samples arc used to measure nonfasting plasma glucose and insulin concentrations. Body weight and food intake are recorded weekly.
[1164] To directly determine the effect of the high-fat diet on the ability of insulin to stimulate glucose disposal, the experiments can be initiated on three groups of mice, fat-fed, chow-fed injected with vehicle, and fat-fed injected with STZ at the end of the 7-weck period described above. Mice can be fasted for 4 hours before Ihe experiments. In the first series of experiments, mice can be anesthetized with methoxyfluranc (i'ilman-Moor, Mundclein, IL) inhalation. Regular insulin (Sigma) can be injected intravenously ([IV] 0.1 U/kg body weight) through a tail vein, and blood can be collected 3, 6, 9, 12, and 15 minutes after the injection from a different tail vein. Plasma glucose concentrations can be determined on these samples, and the half-life (t'/i) of glucose disappearance from plasma can be calculated using WinNonlin (Scientific Consulting, Apex, NC), a pharmacokinctics/pharmacodynamics software program.
[1165| In the second series of experiments, mice can be anesthetized with intraperitoneal sodium pentobarbital (Sigma). The abdominal cavity is opened, and the main abdominal vein is exposed and catheterized with a 24-gauge IV catheter (Johnson-Johnson Medical, Arlington, TX). The
catheter is secured to muscle tissue adjacent to the abdominal vein, cut on the bottom of the syringe connection, and hooked to a prefilled PESO plastic tube, which in turn is connected to a syringe with infusion solution. The abdominal cavity is then sutured closed. With this approach, there would be no blockage of backflow of the blood from the lower part of the body. Mice can he infused continuously with glucose (24.1 mg/kg/min) and insulin (10 mU/kg/min) at an infusion volume of 10 uUmin. Retro-orbital blood samples (70 u,L each) can be taken 90, 105, 120, and 135 minutes after the start of infusion for measurement of plasma glucose and insulin concentrations. The mean of these four samples is used to estimate steady-state plasma glucose (SSF'G) and insulin (SSPI) concentrations for each animal.
|1166| Finally, experiments to evaluate the ability of the albumin fusion proteins, the therapeutic compositions of the instant application, either alone or in combination with any one or more of the therapeutic drugs listed for the treatment of diabetes mellitus, to decrease plasma glucose can be performed in the following two groups of "N1DDM" mice models that are STZ-injccted: (1) fat-fed C57BI76J, and (2) fructose-fed C57B176J. Plasma glucose concentrations of the mice for these studies may range from 255 to 555 mg/dL. Mice are randomly assigned to treatment with either vehicle, albumin fusion therapeutics of the present invention either alone or in combination with any one or more of the therapeutic drugs listed for the treatment of diabetes mellitus. A total of three doses can be administered. Tail vein blood samples can be taken for measurement of the plasma glucose concentration before the first dose and 3 hours after the final dose.
|1167| Plasma glucose concentrations can be determined using the Glucose Diagnostic Kit from Sigma (Sigma No. 315), an enzyme colorimetric assay. Plasma insulin levels can be determined using the Rat Insulin RIA Kit from Linco Research (#RI-13K; St. Charles, MO). EXAMPLE 59: In vitro H4lfe -SEAP Reporter Assays Establishing In volvement in Insulin Action. Tile Various H411e Reporters
|1I68] H4IIe/rMEP-SEAP: '[Tie malic enzyme promoter isolated from rat (rMEP) contains a PPAR-gamma element which is in the insulin pathway. This reporter construct is stably transfected into the liver H411e cell-line.
[1169| II4lle/SREBP-SEAF\ The sterol regulatory element binding protein (SRElJP-lc) is a transcription factor which acts on the promoters of a number of insulin-responsive genes, for example, fatty acid synthctase (PAS), and which regulates expression of key genes in fatty acid metabolism in fibroblasts, adipocytcs, and hepatocytcs. SREBP-Ic, also known as the adipocyte determination and differentiation factor 1 (ADO-1), is considered as the primary mediator of insulin effects on gene expression in adipose cells. It's activity is modulated by the levels of insulin, sterols, and glucose. This reporter construct is stably transfected into the liver H4IIe cell-line.
11170] H41le/FAS-SEAI'\ The fatty acid synthetase reporter constructs contain a minimal SREBP-rcsponsive FAS promoter. This reporter construct is stably Iransfected into the liver H4Ile cell-line.
|I171| H41le/PEPCK-SEAP: The phosphoenolpyruvate carhoxykinasc (PEPCK) promoter is the primary site of hormonal regulation of PEPCK gene transcription modulating PEPCK activity. PEPCK catalyzes a committed and rate-limiting step in hepatic gluconcogcnesis and must therefore be carefully controlled to maintain blood glucose levels within normal limits. This reporter construct is stably transfected into the liver H4IIe cell-line.
11172| These reporter constructs can also be stably transfected into 3T3-L1 fibroblasts and L6 myoblasts. These stable cell-lines are then differentiated into 3T3-L1 adipocytes and L6 myotubes as previously described in Example 13. The differentiated cell-lines can then be used in the SEAP assay described below.
Growth and Asstiy Medium
|1173| The growth medium comprises 10% Fetal Bovine Scrum (FBS), 10% Calf Serum, 1% NEAA, Ix penicillin/streptomycin, and 0.75 mg/ml. G418 (for M4lle/rFAS-SEAP and H4IIc/SRGBP-SBAP) or 0.50 mg/mL G4I8 (for H4IIe/rMEP-SEAP). For H4IIe/PEPCK-SEAP, the growth medium consists of 10% FBS, 1% penicillin/streptomycin, 15 mM HEPES buffered saline, and 0.50 mg/mL G418.
[1174] The assay medium consists of low glucose DMEM medium (Life Technologies), 1% NEAA, Ix penicillin/streptomycin for the H4Ilc/rFAS-SEAP, H41Ie/SREDP-SEAP, II41le/rMEP-SEAP reporters. The assay medium for H4IIe/PEPCK-SEAP reporter consists of 0.1% FBS, 1% penicillin/streptomycin, and 15 mM HEPES buffered saline. Me I hot!
|1175| The 96-well plates are seeded at 75,000 cells/well in 100 u.L/well of growth medium until cells in log growth phase become adherent. Cells are starved for 48 hours by replacing growth medium with assay medium, 200 uL/well. (For H4IIe/PEPCK-SEAP cells, assay medium containing 0.5 uJvl dexamcthasone is added at 100 uLAvell and incubated for approximately 20 hours). The assay medium is replaced thereafter with 100 uLAvell of fresh assay medium, and a 50 u.L aliquot of cell supernatant obtained from transfected cell-lines expressing the therapeutics of the subject invention (e.g., albumin fusion proteins of the invention and fragments and variants thereof) is added to the well. Supematants from empty vector transfected cell-lines are used as negative control. Addition of 10 nM and/or 100 nM insulin to the wells is used as positive control. After 48 hours of incubation, the conditioned media are harvested and SEAP activity measured (Phospha-Light System protocol, Tropix #BP2500) Briefly, samples are diluted 1:4 in dilution buffer and incubated at 65 °C for 30 minutes to inactivate the endogenous non-placental form of SEAP. An aliquot of 50 uLofthe diluted samples is mixed with 50 jiLof SEAP Assay Buffer which contains a mixture of inhibitors active against the non-155
iacemai MiAi' isoenzymcs anu 15 incubated for another 5 minutes. An aliquot of 50 uLof CSPD chetniluminescent substrate which is diluted 1: EXAMPLE 60: Transvcnic Animals.
|1176| The albumin fusion proteins of the invention can also be expressed in transgenic animals. Animals of any species, including, but not limited to, mice, rats, rabbits, hamsters, guinea pigs, pigs, micro-pigs, goats, sheep, cows and non-human primates, e.g., baboons, monkeys, and chimpanzees may be used to generate transgenic animals. In a specific embodiment, techniques described herein or otherwise known in the art, are used to express fusion proteins of the invention in humans, as part of a gene therapy protocol.
11177] Any technique known in the art may be used to introduce the polynucleotides encoding the albumin fusion proteins of the invention into animals to produce the founder lines of transgenic animals. Such techniques include, but are not limited to, pronuclear microinjection (Paterson et a!., Appl. Microbiol. Biotechnol. 40:691-698 (1994); Carver et al., Biotechnology (NY) 11:1263-1270 (1993); Wright et al., Biotechnology (NY) 9:830-834 (1991); and Hoppe ct al., U.S. Pat. No. 4,873,191 (1989)); retrovirus mediated gene transfer into germ lines (Van der Putten et al., Proc. Nail. Acad. Sci., USA 82:6148-6152 (1985)), blastocysts or embryos, gene targeting in embryonic stem cells (Thompson et al., Cell 56:313-321 (1989)); electroporation of cells or embryos (Lo, 1983, Mol Cell. Biol. 3:1803-1814 (1983)); introduction of the polynucleotides of the invention using a gene gun (see, e.g., Ulmer et al, Science 259:1745 (1993); introducing nucleic acid constructs into embryonic plcuripotent stem cells and transferring the stem cells back into the blastocyst; and sperm-mediated gene transfer (Lavitrano et al., Cell 57:717-723 (1989); etc. For a review of such techniques, see Gordon, "Transgenic Animals," Intl. Rev. Cytol. 115:171-229 (1989), which is incorporated by reference herein in its entirety. 11178] Any technique known in the art may be used to produce transgenic clones containing polynucleotides encoding albumin fusion proteins of the invention, for example, nuclear transfer into enucleated oocytes of nuclei from cultured embryonic, fetal, or adult cells induced to quiescence (Campell et al., Nature 380:64-66 (1996); Wilmut et al., Nature 385:810-813 (1997)).
|1179] The present invention provides for transgenic animals that carry the polynucleotides encoding the albumin fusion proteins of the invention in all their cells, as well as animals which carry these polynucleotides in some, but not all their cells, i.e.. mosaic animals or chimeric. The transgene may be integrated as a single transgene or as multiple copies such as in concatamers, e.g., head-to-head tandems or head-to-tail tandems. The transgene may also be selectively introduced into and activated in a particular cell type by following, for example, the teaching of Lasko et al. (Lasko ct al., Proc. Natl. Acad. Sci. USA 89:6232-6236 (1992)). The regulatory sequences required for such a cell-type specific activation will depend upon the particular cell type of interest, and will be apparent to those of skill in the art. When it is desired that the polynucleotide encoding the fusion protein of the invention be integrated into the chromosomal site of the endogenous gene corresponding to the Therapeutic protein portion or ablumin portion of the fusion protein of the invention, gene targeting is preferred. Briefly, when such a technique is to be utilized, vectors containing some nucleotide sequences homologous to the endogenous gene are designed for the purpose of integrating, via homologous recombination with chromosomal sequences, into and disrupting the function of the nucleotide sequence of the endogenous gene. The transgene may also be selectively introduced into a particular cell type, thus inactivating the endogenous gene in only that cell type, by following, for example, the teaching of Gu et al. (Gu et al., Science 265:103-106 (1994)). The regulatory sequences required for such a cell-type specific inactivation will depend upon the particular cell type of interest, and will be apparent to those of skill in the art.
|1180| Once transgenic animals have been generated, the expression of the recombinarit gene may be assayed utilizing standard techniques. Initial screening may be accomplished by Southern blot analysis or PCR techniques to analyze animal tissues to verify that integration of the polynucleotide encoding the fsuion protien of the invention has taken place. The level of mRNA expression of the polynucleotide encoding the fusion protein of the invention in the tissues of the transgenic animals may also be assessed using techniques which include, but are not limited to, Northern blot analysis of tissue samples obtained from the animal, in situ hybridization analysis, and reverse transcriptase-PCR (rt-PCR). Samples of fusion protein-expressing tissue may also be evaluated immunocytochemically or immunohistochemically using antibodies specific for the fusion protein.
[1181] Once the founder animals are produced, they may be bred, inbred, outbred, or crossbred to produce colonies of the particular animal. Examples of such breeding strategies include, but are not limited to: outbrceding of founder animals with more than one integration site in order to establish separate lines; inbreeding of separate lines in order to produce compound transgenics that express the transgene at higher levels because of the effects of additive expression of each transgene, crossing of heterozygous transgenic animals to produce animals homozygous for a given integration site in order to both augment expression and eliminate the need for screening of animals by DNA analysis; crossing of separate homozygous lines to produce compound heterozygous or homozygous lines; and breeding to place the transgene (i.e., polynucleotide encoding an albumin fusion protein ot the invention) on a distinct background that is appropriate for an experimental model of interest.
Transgenic animals of the invention have uses which include, but are not limited to, animal model systems useful in elaborating the biological function of fusion proteins of the invention and the Therapeutic protein and/or albumin component of the fusion protein of the invention, studying conditions anoVor disorders associated with aberrant expression, and in screening for compounds effective in ameliorating such conditions and/or disorders.
EXAMPLE 61: Method ofTreatmenl Using f!fne Therapy-Ex KiVo.
(1I82| Une method ot gene tncrapy transplants iibroblasts, which are capable of expressing an albumin fusion protein of the present invention,
onto a patient. Generally, fibrublasts arc obtained from a subject by skin biopsy. The resulting tissue is placed in tissue-culture medium and
separated into small pieces. Small chunks of the tissue are placed on a wt-t surface of a tissue culture flask, approximately ten pieces are placed in
each flask. The flask is turned upside down, closed tight and left at room temperature over night. After 24 hours at room temperature, the flask is
inverted and the chunks of tissue remain fixed to the bottom of the flask and fresh media (e.g., Ham's F12 media, with 10% FBS, penicillin and
streptomycin) is added. The flasks are then incubated at 37 degree C for approximately one week.
|1183) At this time, fresh media is added and subsequently changed every several days. After an additional two weeks in culture, a monolayer
of fibroblasts emerge. The monolayer is trypsinized and scaled into larger flasks.
[1184] pMV-7 (Kirschmcicr, P.T. et al., DNA, 7:219-25 (1988)), Tanked by the long terminal repeats of the Moloney murine sarcoma virus, is
digested with KeoKI and HindUl and subsequently treated with calf intestinal phosphatase. The linear vector is fractionated on agarose gel and
purified, using glass beads.
|1185] Polynucleotides encoding an albumin fusion protein of the invention can be generated using techniques known in the art amplified using
PCR primers which correspond to the 5' and 3' end sequences and optionally having appropriate restriction sites and initiation/stop codons, if
necessary. Preferably, the 5' pnmer contains an licoRl site and the 3' primer includes a Kindlll site. Equal quantities of the Moloney murine
sarcoma virus linear backbone and the amplified EcoRl and Hindlll fragment are added together, in the presence of T4 DNA ligase. The resulting
mixture is maintained under conditions appropriate for ligation of Ihe two fragments. The ligation mixture is then used lo transform bacteria
HB101, which are then plated onto agar containing kanamycin for the purpose of confirming that the vector has the gene of interest properly
inserted.
|1186) The amphotropic pA317 or GP+aml2 packaging cells are grown in tissue culture to confluent density in Dulbecco's Modified Eagles
Medium (DMEM) with 10% calf serum (CS), penicillin and streptomycin. The MSV vector containing the gene is then added to the media and the
packaging cells transduced with the vector. The packaging cells now produce infectious viral particles containing the gene (the packaging cells are
now referred to as producer cells).
J1187J Fresh media is added lo the transduced producer cells, and subsequently, the media is harvested from a 10 cm plate of confluent
producer cells. The spent media, containing the infectious viral particles, is filtered through a millipore filter to remove detached producer cells and
this media is then used to infect tibroblast cells. Media is removed from a sub-confluent plate of fibroblasts and quickly replaced with the media
from the producer cells. This media is removed and replaced with fresh media. If the liter of virus is high, then virtually all fibroblasts will be
infected and no selection is required. If the titer is very low, then it is necessary to use a retroviral vector that has a selectable marker, such as neo or
his. Once the fibroblasts have been efficiently infected, the fibroblasts are analyzed lo determine whether the albumin fusion protein is produced.
)1188] The engineered fibroblasts are then transplanted onto the host, either alone or after having been grown to confluence on cytodex 3
microcarrier beads.
EXAMPLE 62: Method of Treatment Using Gene Therapy- in Vivo.
|1189] Another aspect of the present invention is using in vivo gene therapy methods to treat disorders, diseases and conditions. The gene therapy method relates to the introduction of naked nucleic acid (DNA, R.NA, and antisense DNA or RNA) sequences encoding an albumin fusion protein of the invention into an animal. Polynucleotides encoding albumin fusion proteins of the present invention may be opcratively linked to (i.e., associated with) a promoter or any other genetic elements necessary for the expression of the polypcptide by the target tissue. Such gene Iherapy and delivery techniques and methods are known in the art, see, for example, WO90/11092, WO98/11779; U.S. Patent NO. 5693622. 5705151, 5580859, Tabata et al., Cardiovasc. Res. 35(3):470-479 (1997); Chao et al., Pharmacol. Res. 35(6):517-522 (1997); Wolff, Neuromuscul. Disord. 7(5):314-318 (1997), Schwartz et al.. Gene Ther. 3(5):405-411 (1996); Tsurumi et al.. Circulation 94(12):3281-3290 (1996) (incorporated herein by reference).
[1190| The polynucleotidc constructs may be delivered by any method that delivers injectable materials to the cells of an animal, such as, injection inlo the interstitial space of tissues (heart, muscle, skin, !ur.g, liver, intestine and the like). The polynucleotide constructs can be delivered in a pharmaceutical ly acceptable liquid or aqueous carrier.
|l 191) The term "naked" polynucleotide, DNA or RNA, refers to sequences that are free from any delivery vehicle that ads to assist, promcte, or facilitate entry into the ceil, including viral sequences, viral particles, liposomc formulations, lipofectin or precipitating agents and the like. However, polynucleolities encoding albumin fusion proteins uf the present invention may also be delivered in liposome formulations (such as those taught in Feigner P.I... et al. (1995) Ann. NY Acad. Sci. 772:126-139 and Abdallah B. ct al. (1995) Biol. Cell 85(l);l-7) which, can be prepared by methods weli known to those skilled in the art.
]1I92] The polynucleotide vector constructs used in the gene therapy method arc preferably constructs that will not integrate into the host genome nor will they contain sequences that allow for replication. Any strong promoter known to those skilled in the art can be used for driving the expression of DNA. 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 ceils to
provide production 01 me uesiicu poiypcptide for periods of up to six months.
|1I93| 'Hie polynucleotide construct can be delivered to the interstitial space of tissues within an animal, including muscle, skin, brain, lung, liver, spleen, hone 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 cnsheathing 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 vim muscle cells are particularly competent in their ability to take up and express polynucleotidcs.
(1194] For the naked polynucleotide injection, an effective dosage amount of DNA or RNA will be in the range of from about 0.05 g/kg body weight to about 50 mg/kg body weight. Preferably the dosage will be from about 0.005 mg/kg to about 20 mg/kg and more preferably from about 0.05 mg/kg to about 5 mg/kg. Of course, as the artisan of ordinary skill will appreciate, this dosage will vary according to the tissue site of injection. The appropriate and effective dosage of nucleic acid sequence can readily be determined by those of ordinary skill in the art and may depend on the condition being treated and the route of administration. The preferred route of administration is by the parenteral route of injection into the interstitial space of tissues. 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 polynucleotide constructs can be delivered to arteries during angioplasty by the catheter used in the procedure.
[1195] The dose response effects of injected polynucleotide in muscle in vivo is determined as follows. Suitable template DNA for production of mRNA coding for polypcptide of the present invention is prepared in accordance with a standard recombinant DNA methodology. The template DNA, which may be either circular or linear, is either used as naked DNA or complexed with liposomcs. The quadriceps muscles of mice are then injected with various amounts of the template DNA.
[1196] Five to six week old female and male Balb/C mice are anesthetized by intraperitoneal injection with 0.3 ml of 2.5% Avertin. A 1.5 cm incision is made on the anterior thigh, and the quadriceps muscle is directly visualized. The template DNA is injected in 0,1 ml of carrier in a I cc syringe through a 27 gauge needle over one minute, approximately 0.5 cm from the distal insertion site of the muscle into the knee and about 0.2 cm deep. A suture is placed over the injection site for future localization, and the skin is closed with stainless steel clips.
|1197| After an appropriate incubation time (e.g., 7 days) muscle extracts are prepared by excising the entire quadriceps. Every fifth 15 urn cross-section of the individual quadriceps muscles is histochemically stained for protein expression. A time course for fusion protein expression may be done in a similar fashion except that quadriceps from different mice are harvested at different times. Persistence of DNA in muscle following injection may be determined by Southern blot analysis after preparing total cellular DNA and HIKT supematants from injected and control mice. The results of the above experimentation in mice can be used to extrapolate proper dosages and other treatment parameters in humans and other animals using naked DNA.
EXAMPLE 63: Biological Effects affusion Proteins of the Invention.
Astrocvle and Neuronal Assays.
II1981 Albumin fusion proteins of the invention can be tested for activity in promoting the survival, ncurite outgrowth, or phenotypic differentiation of cortical neuronal cells and for inducing the proliferation of glial fibrillary acidic protein immunopositive cells, astrocytes. The selection of cortical cells for the bioassay is based on the prevalent expression of FGF-1 and FGF-2 in cortical structures and on the previously reported enhancement of cortical neuronal survival resulting from FGF-2 treatment. A thymidine incorporation assay, for example, can be used to elucidate an albumin fusion prutcin of the invention's activity on these cells.
II1991 Moreover, previous reports describing the biological effects of FGF-2 (basic FGF) on cortical or hippocarnpal neurons in vitro have demonstrated increases in both neuron survival and neurite outgrowth (Walicke et al., "Fibroblast growth factor promotes survival of dissociated hippocampal neurons and enhances neurite extension." Proc, Nail Acad. Sci. USA 83:1012-3016. (1986), assay herein incorporated by reference in its entirety). However, reports from experiments done on PC-12 cells suggest that these two responses are not necessarily synonymous and may depend on not only which FGF is being tested but also on which receptor(s) are expressed on the target cells. Using the primary cortical neuronal culture paradigm, the ability of an albumin fusion protein of the invention to induce neurite outgrowth can be compared to the response achieved with FGF-2 using, for example, a thymidine incorporation assay.
nbrobldst and endothelial cell assays.
|1200| Human lung fibroblasts are obtained from Clonetics (San Diego, CA) and maintained in growth media from Clonetics. Dermal
microvascmar cnaotncaai ccmis are ootained from Cell Applications (San Diego, CA). For proliferation assays, the human lung fibroblasts and dermal microvascular endothelial cells can be cultured at 5,000 cells/well in a 96-well plate for one day in growth medium. The cells are then incubated far one day in 0.1% BSA basal medium. After replacing tlie medium with fresh 0.!% BSA medium, the cells are incubated with the test fusion protein of the invention proteins for 3 days. Alamar Blue (Alamar Bioscicnces, Sacramento, CA) is added to each well to a final concentration of 10%. The cells arc incubated for 4 hr. Cell viability is measured by reading in a CytoFluor fluorescence reader. For the PGE; assays, the human lung fibroblasts are cultured at 5,000 cells/well in a 96-well plate for one day. After a medium change to 0-1% BSA basal medium, the cells are incubated with FGF-2 or fusion protein of the invention with or without IL-lot for 24 hours. The supematants are collected and assayed for PGE2 by EIA kit (Cayman, Ann Arbor, Ml). For the IL-6 assays, the human lung fibroblasts are cultured at 5,000 cells/well in a 96-well plate for one day. After a medium change to 0.1% BSA basal medium, the cells are incubated with FGF-2 or with or without an albumin fusion protein of the invention and/or IL-la for 24 hours. The supematants are collected and assayed for IL-6 by ELISA kit (Endogen, Cambridge, MA).
1110II Human lung fibroblasts are cultured with FGF-2 or an albumin fusion protein of the invention for 3 days in basal medium before the addition of Alamar Blue to assess effects on growth of the fibroblasts. FGF-2 should show a stimulation at 10 - 2500 ng/ml which can be used to compare stimulation with the fusion protein of the invention.
Cell proliferation based on fiHIthvmictine incorporation
|I202| The following [3H]Thymidine incorporation assay can be used to measure the effect of a Therapeutic proteins, e.g., growth factor proteins, on the proliferation of cells such as fibroblast cells, epithelial cells or immature muscle cells.
(1203] Sub-confluent cultures are arrested in Gl phase by an 18 h incubation in serum-free medium. Therapeutic proteins are then added for 24 h and during the last 4 h, the cultures are labeled with [3H]thymidinc, at a final concentration of 0.33 p.M (25 Ci/mmol, Amersham, Arlington Heights, II.). The incorporated [3II]thymidine is precipitated with ice-cold 10% trichloroacetic acid for 24 h. Subsequently, the cells are rinsed sequentially with ice-cold 10% trichloroacetic acid and then with ice-cold water. Following lysis in 0.5 M NaOH, the lysates and PBS rinses (500 ml) are pooled, and the amount of radioactivity is measured.
Parkinson Models.
|1204| The loss of molor function in Parkinson's disease is attributed to a deficiency of striatai dopamine resulting from the degeneration of the nigrnstriatal dopaminergic projection neurons. An animal model for Parkinson's that has been extensively characterized involves the systemic administration of l-methyl-4 phenyl 1,2,3,6-tetrahydropyridine (MPTP). In the CNS, MPTP is takcn-up by astrocytes and catabolized by monoamine oxidasc B to l-mcthyl-4-phenyl pyridine (MPP*) and released. Subsequently, MPPf is actively accumulated in dopaminergic neurons by the high-affinity reuptake transporter for dopamine. MPP* is then concentrated in mitochondria by the electrochemical gradient and selectively inhibits nicotidamidc adenine disphosphate: ubiquinone oxidoreductionase (complex I), thereby interfering with electron transport and eventually generating oxygen radicals.
|1205| It has been demonstrated in tissue culture paradigms that FGF-2 (basic FGF) has trophic activity towards nigral dopamincrgic neurons (Ferrari et al., Dev. Biol. 1989). Recently, Dr. Unsicker's group has demonstrated that administering FGF-2 in gel foam implants in the stratum results in the near complete protection of nigral dopaminergic neurons from the toxicity associated with MPTP exposure (Otto and Unsickcr, J. Neuroscience, 1990).
|1206] Based on the data with FGF-2, an albumin fusion protein of the invention can be evaluated to determine whether it has an action similar to that of FGF-2 in enhancing dopaminergic ncurorial survival in vitro and it can also be tested in vivo for protection of dopamincrgic neurons in the stnatum from the damage associated with MPTP treatment. The potential effect of an albumin fusion protein of the invention is first examined in vitro in a dopaminergic neuronal cell culture paradigm. The cultures are prepared by dissecting the midbrain floor plate from gestation day 14 Wistar rat embryos. The tissue is dissociated with trypsin and seeded at a density of 200,000 cells/cm2 on polyorthinine-laminin coaled glass coverslips. The cells are maintained in Dulbecco's Modified Eagle's medium and F12 medium containing hormonal supplements (Nl). The cultures are fixed with paraformaldeliydc after 8 days in vitro and are processed for tyrosine hydroxylase, a specific marker for dopaminergic neurons, immunohistochcmical staining. Dissociated cell cultures are prepared from embryonic rats. The culture medium is changed every third day and the factors are also added at that time.
[1207] Since the dopaminergic neurons are isolated from animals at gestation day 14, a developmental time which is past the stage when the dopaminergic precursor cells are proliferating, an increase in the number of tyrosine hydroxylase immunopositive neurons would represent an increase in the number of dopaminergic neurons surviving in vitro. Therefore, if a therapeutic protein of the invention acts to prolong the survival of dopaminergic neurons, it would suggest that the fusion protein may be involved in Parkinson's Disease.
EXAMPLE 64: Pancreatic Reta-Cell Transplantation Combination Therapy.
|1208] Transplantation is a common form of treatment of autoimmune disease, especially when the target self tissue has been severely damaged.
l-or example, and not by way 01 nmuaiion, pancreas transplantation and islet cell transplantation arc common treatment options for IDDM (See, e.g., Stewart et al.. Journal of Clinical Endocrinology & Metabolism 86 (3): 984-988 (2001); Brunicardi, Transplant. Proc. 28: 2138^40 (1996); Kendall & Robertson, Diabetes Mctah. 22- 157-163 (1996); Hamano et al, Kobe J. Med. Sci. 42: 93-104 (1996); Larsen & Stratta, Diabetes Metab. 22: 139-146 (.1996); and Kinkhabwala, et al., Am. J. Siirg. 171: 516-520 (1996)). As with any transplantation method, transplantation therapies for autoimmune disease patienls include treatments to minimize the risk of host rejection of the transplanted tissue. However, autoimmune disease involves the additional, independent risk that the pre-existing host autoimmune response which damaged the original self tissue will exert the same damaging effect on the transplanted tissue. Accordingly, the present invention encompasses methods and compositions for the treatment of autoimmune pancreatic disease using the albumin fusion proteins of the subject invetion in combination with immunomodulators/immunosuppressanls in individuals undergoing transplantation therapy of the autoimmune disease.
(1209] In accordance with the invention, the albumin fusion-based compositions and formulations described above, are administered to prevent and treat damage to the transplanted organ, tissue, or cells resulting from the host individual's autoimmune response initially directed against the original self tissue. Administration may be carried out both prior and subsequent to transplantation in 2 to 4 doses each one week apart. [12101 The following immunomodulators/immunosuppressants including, but not limited to, AI-401, CDP-571 (anti-TNF monoclonal antibody), CG-1088, Diamyd (diabetes vaccine), ICM3 (anti-ICAM-3 monoclonal antibody), linomide (Roquinimex), NB1-6024 (altered peptide ligand), TM-27, VX-740 (HMR-3480), caspase 8 protease inhibitors, thalidomide, hOKTSgammal (Ala-ala) (anti-CD3 monoclonal antibody), Oral Intei feron-Alpha, oral lactobacillus, and LymphoStat-B™ can he used together with the albumin fusion therapeutics of the subject invention in islet cell or pancreas transplantation.
EXAMPLE 65:Iiicntification and Cloning of VH and VI. domains.
11211] One method to identfy and clone VH and VL domains from cell lines expressing a particular antibody is to perform PCR with VH and VI. specific primers on cDNA made from the antibody expressing cell lines. Briefly, RNA is isolated from the cell lines and used as a template for RT-PCR designed to amplify the VH and VL domains of the antibodies expressed by the EBV cell lines. Cells may be lysed in the TRIzol® reagent (Life Technologies, Rockville. MD) and extracted with one fifth volume of chloroform. After addition of chloroform, the solution is allowed to incubate at room temperature for 10 minutes, and the centrifuged at 14,000 rpm for 15 minutes at 4°C in a tabletop centrifuge. The supernatant is collected and RNA is precipitated using an equal volume of isopropanol. Precipitated RNA is pelleted by centrifuging at 14,000 rpm for 15 minutes at 4°C in a tabletop centrifuge. Following ccntrifugation, the supernatant is discarded and washed with 75% ethanol. Follwing washing, the RNA is centrifuged again at 800 rpm for 5 minutes at 4°C. The supernatant is discarded and the pellet allowed to air dry. RNA is the dissolved in DEPC water and heated to 60°C for 10 minutes. Quantities of RNA can determined using optical density measurements.
(1212] cDNA may be synthesized, according to methods well-known in the art, from 1.5-2.5 micrograms of RNA using reverse transciptase and random hexamer primers. cDNA is then used as a template for PCR amplification of VH and VL domains. Primers used to amplify VH and VI. genes arc shown in Table 7. Typically a PCR reaction makes use of a single 5' primer and a single 3' primer. Sometimes, when the amount of available RNA template is limiting, or for greater efficiency, groups of 5' and/or 3' primers may be used. For example, sometimes all five VH-5' primers and all JH3' primers are used in a single PCR reaction. The PCR reaction is carried out in a 50 microliter volume containing IX PCR buffer, 2rnM of each dNTP, 0.7 units of High Fidelity Taq polymerse, 5' primer mix, 3' primer mix and 7.5 microliters of cDNA. The 5' and 3" primer mix of both VH and VL can be made by pooling together 22 pmole and 28 pmole, respectively, of each of the individual primers. PCR conditions arc: 96°C for 5 minutes; followed by 25 cycles of 94°C for 1 minute, 50°C for 1 minute, and 72°C for 1 minute; followed by an extension cycle of 72°C for 10 minutes. After the reaction is completed, sample tubes are stored 4°C.
(Table Removed)
PCR samples are then electrophorescd on a 1.3% agarose gel. DNA bands of the expected sizes (-506 base pairs for VH domains, and 344 base pairs for VL domains) can be cut out of the gel and purified using methods well known in the art. Purified PCR products can be ligated into a PCR cloning vector (TA vector from Invitrogen Inc., Carlsbad, CA). Individual cloned PCR products can be isolated after transfection of E. coli and blue/while color selection. Cloned PCR products may then be sequcnccd using methods commonly known in the art.
[1213] The PCR bands containing the VH domain and the VL domains can also be used to create full-length Ig expression vectors. VH and VL domains can be cloned into vectors containing the nucleotide sequences of a heavy (e.g., human IgGl or human lgG4) or light chain (human kappa or human lambda) constant regions such that a complete heavy or light chain molecule could be expressed from these vectors when transfected into an appropriate host cell. Further, when cloned heavy and light chains are both expressed in one cell line (from either one or two vectors), they can assemble into a complete functional antibody molecule that is secreted into the cell culture medium. Methods using polynucleotides encoding VH and VL antibody domain to generate expression vectors that encode complete antibody molecules are well known within the art.
EXAMPLE 66: Preparation of HA-cytokine or HA~nron'lli factor fusion proteins (such as NGF, BDNFa. BDNb'b anil BDNFc). 11214) The cDNA for the cytokine or growth factor of interest, such as NGF, can be isolated by a variety of means including from cDNA libraries, by RT-PCR and by PCR using a scries of overlapping synthetic oligonucleotide primers, all using standard methods. The nucleotide sequences for all of these proteins are known and available. The cDNA can be tailored at the 5' and 3' ends to generate restriction sites, such that oligonucleotide linkers can be used, for cloning of the cDNA into a vector containing the cDNA for HA. This can be at the N or C-temiinus with or without the use of a spacer sequence. NGF (or other cytokine) cDNA is cloned into a vector such as pPPCOOOS (Figure 2), pScCHSA, pScNHSA , or pC4:HSA from which the complete expression cassette is then excised and inserted into the plasmid pSAC35 to allow the expression of the albumin fusion protein in yeast. The albumin fusion protein secreted from the yeast can then be collected and purified from the media and tested for its biological activity. For expression in mammalian cell lines, a similar procedure is adopted except that the expression cassette used employs a mammalian promoter, leader sequence and terminator (See Example I). This expression cassette is then excised and inserted into a plasmid suitable for the transfection of mammalian cell lines.
EXAMPLE 67: Preparation of HA-IFN fusion proteins (suc/i as IFNg)-
IT'lS) The rONA for the interferon of interest such as IFNa can be isolated by a variety of means including but not exclusively, from cDNA
libraries, by RT-PCR and by PCR using a scries of overlapping synthetic oligonucleotide primers, all using standard methods. The nucleotidc sequences for interfcrons, such as IFNct are known and available, for instance, in U.S. Patents 5,326,859 and 4,588,585, in EP 32 134, as well as in public databases such as GenBank. The cDNA can be tailored at the 5' and 3' ends to generate restriction sites, such that oligonucleotide linkers can be used to clone the cDNA into a vector containing the cDNA for HA. This can be at the N or C-terminus of the HA sequence, with or without the use of a spacer sequence. The IFNa (or other interferon) cDNA is cloned into a vector such as pPPCOOOS (Figure 2), pScCHSA, pScNHSA , or pC4:HSA from which the complete expression cassette is then excised and inserted into the plasmid pSAC35 to allow the expression of the albumin fusion protein in yeast. The albumin fusion protein secreted from the yeast can then be collected and purified from the media and tested for its biological activity. For expression in mammalian cell lines a similar procedure is adopted except that the expression cassette used employs a mammalian promoter, leader sequence and terminator (See Example 1). This expression cassette is then excised and inserted into a plasmid suitable for the transfection of mammalian cell lines.
Maximum protein recovery from viah
|1216| The albumin fusion proteins of the invention have a high degree of stability even when they are packaged at low concentrations. In addition, in spite of the low protein concentration, good fusion-protein recovery is observed even when the aqueous solution includes no other protein added to minimize binding to the vial walls. The recovery of vial-stored HA-1FN solutions was compared with a stock solution. 6 or 30 ug/ml HA-1FN solutions were placed in vials and stored at 4°C. After 48 or 72 hrs a volume originally equivalent to 10 ng of sample was removed and measured in an IFN sandwich ELISA. The estimated values were compared to that of a high concentration stock solution. As shown, there is essentially no loss of the sample in these vials, indicating that addition of exogenous material such as albumin is not necessary to prevent sample loss to the wall of the vials
In vivo stability and bioavailabilitv ofHA-a-IFN fusions
11217] To determine the in vivo stability and bioavailability of a HA-a-lFN fusion molecule, the purified fusion molecule (from yeast) was administered to monkeys. Pharmaceutical compositions formulated from HA-a-IFN fusions may account for the extended serum half-life and bioavailability. Accordingly, pharmaceutical compositions may be formulated to contain lower dosages of alpha-interferon activity compared to the native alpha-interferon molecule.
(1218] Pharmaceutical compositions containing HA-a-IFN fusions may be used to treat or prevent disease in patients with any disease or disease state that can be modulated by the administration of a-lFN. Such diseases include, but are not limited to, hairy cell leukemia, Kaposi's sarcoma, genital and anal warts, chronic hepatitis B, chronic non-A, non-B hepatitis, in particular hepatitis C, hepatitis D, chronic myelogenous leukemia, renal cell carcinoma, bladder carcinoma, ovaiian and cervical carcinoma, skin cancers, recurrent respirator papillomatosis, non Hodgkin's and cutaneous T-cell lyrnphomas, melanoma, multiple myeloma , AIDS, multiple sclerosis, gliobastoma, etc. (see Interfcron Alpha, In: AHFS Drug Information, 1997.
[1219] Accordingly, the invention includes pharmaceutical compositions containing a HA-ct-IFN fusion protein, polypcptidc or peptide formulated with the proper dosage for human administration. The invention also includes methods of treating patients in need of such treatment comprising at least the step of administering a pharmaceutical composition containing at least one HA-a-IFN fusion protein, polypeptide or peptide.
Bifunctional HA-a-IITN fusions
|1220] A HA-a-IFN expression vector may be modified to include an insertion for the expression of bifunctional HA-a-IFN fusion proteins. For instance, the cDNA for a second protein of interest may be inserted in frame downstream of the "rllA-lFN" sequence after the double stop cndon has been removed or shifted downstream of the coding sequence.
112211 In one version of a bifunctional HA-a-IFN fusion protein, an antibody or fragment against B-lymphocyte stimulator protein (GenBank Ace 4455139) or polypeptide may be fused to one end of the HA component of the fusion molecule. This bifunctional protein is useful for modulating any immune response generated by the a-IFN component of the fusion.
EXAMPLE 68: Preparation of HA-hormone fusion protein
|1222] The cDNA for the hormone of interest can be isolated by a variety of means including but not exclusively, from cDNA libraries, by RT-PCR and by PCR using a series of overlapping synthetic oligonucleotide primers, all using standard methods. The nucleotide sequences for all of these proteins are known and available, for instance, in public databases such as GenBank. The cDNA can be tailored at the 5' and 3' ends to generate restriction sites, such that oligonucleotide linkers can be used, for cloning of the cDNA into a vector containing the cDNA for HA. This can be at the N or C-tcrmmus with or without the use of a spacer sequence. The hormone cDNA is cloned into a vector such as pPPCOOOS (Figure 2), pScCHSA, pScNHSA , or pC4:HSA from which the complete expression cassette is then excised and inserted into the plasmid pSAC35 to allow the expression of the albumin fusion protein in yeast. The albumin fusion protein secreted from the yeast can then be collected and purified from the media and tested for its biological activity. For expression in mammalian cell lines a similar procedure is adopted except that the expression cassette used employs a mammalian promoter, leader sequence and terminator (See Example 1). This expression cassette is then excised arid inserted into a plasmid suitable for the transfection of mammalian cell lines.
£, |1223] The cDNA for the soluble receptor or binding protein of interest can be isolated by a variety of means including but not exclusively, from cDNA libraries, by RT-PCR and by PCR using a series of overlapping synthetic oligonucleotide primers, all using standard methods. The nucleotide sequences for all of these proteins arc known and available, for instance, in GenBank. The cDNA can be tailored at the 5' and 3' ends to generate restriction sites, such that oligonucleotide linkers can be used, for cloning of the cDNA into a vector containing the cDNA for HA. This can be at the N or C-terminus with or without the use of a spacer sequence. The receptor cDNA is cloned into a vector such as pPPCOOOS (Figure 2), pScCHSA, pScNHSA , or pC4:IISA from which the complete expression cassette is then excised and inserted into the plasmid pSAC35 to allow the expression of the albumin fusion protein in yeast. The albumin fusion protein secreted from the yeast can then be collected and purified from the media and tested for its biological activity. For expression in mammalian cell lines a similar procedure is adopted except that the expression cassette used employs a mammalian promoter, leader sequence and terminator (See Example 1). This expression cassette is then excised and inserted into a plasmid suitable for the transfection of mammalian cell lines.
EXAMPLE 70: Preparation of HA-crowth factors.
|1224| The cDNA for the growth factor of interest can be isolated by a variety of means including but not exclusively, from cDNA libraries, by RT-PCR and by PCR using a series of overlapping synthetic oligonucleotide primers, all using standard methods (see GcnBank Ace. No.NP_000609). The cDNA can be tailored at the 5' and 3' ends to generate restriction sites, such that oligonucleotide linkers can be used, for cloning of the cDNA into a vector containing the cDNA for HA. This can be at the N or C-terminus with or without the use of a spacer sequence. The growth factor cDNA is cloned into a vector such as pPPCOOOS (Figure 2), pScCHSA, pScNHSA , or pC4:HSA from which the complete expression cassette is then excised and inserted into the plasmid pSAC35 to allow the expression of the albumin fusion protein in yeast. The albumin fusion protein secreted from the yeast can then be collected and purified from the media and tested for its biological activity. For expression in mammalian cell lines a similar procedure is adopted except that the expression cassette used employs a mammalian promoter, leader sequence and terminator (See Example 1). This expression cassette is then excised and inserted into a plasmid suitable for the transfection of mammalian cell lines.
EXAMPLE 77; Preparation of HA-single chain antibody fusion proteins.
|1225] Single chain antibodies are produced by several methods including but not limited to: selection from phage libraries, cloning of the variable region of a specific antibody by cloning the cDNA of the antibody and using the flanking constant regions as the primer to clone the variable region, or by synthesizing an oligonucleotide corresponding to the variable region of any specific antibody. The cDNA can be tailored at the 5' and 3' ends to generate restriction sites, such that oligonucleotide linkers can be used, for cloning of the cDNA into a vector containing the cDNA for HA. This can be at the N or C-tcrminus with or without the use of a spacer sequence. The cell cDNA is cloned into a vector such as pPPCOOOS (Figure 2), pScCHSA, pScNHSA , or pC4:HSA from which the complete expression cassette is then excised and inserted into the plasmid pSAC35 to allow the expression of the albumin fusion protein in yeast.
11226] In fusion molecules of the invention, the V,, and VL can be linked by one of the following means or a combination thereof: a peptide linker between the C-temiinus of the VH and the N-terniinus of the Vu, a K.ex2p protease cleavage site between the VM and VLsuch that the two are cleaved apart upon secretion and then self associate; and cystine residues positioned such that the VH and VL can form a disulphide bond between them to link them together. An alternative option would be to place the VH at the N-terminus of HA or an HA domain fragment and the VL at the C-terrninus of the HA or HA domain fragment.
J1227] The albumin fusion protein secreted from the yeast can then be collected and purified from the media and tested for its activity. For expression in mammalian cell lines a similar procedure is adopted except that the expression cassette used employs a mammalian promoter, leader sequence and terminator (See Hxample 1). This expression cassette is then excised and inserted into a plasmid suitable for the transfection of mammalian cell lines. The antibody produced in this manner can be purified from media and tested for its binding to its antigen using standard immunochcmical methods.
EXAMPLE 72: Preparation of HA-cett adhesion molecule fusion proteins.
11228] The cDNA for the cell adhesion molecule of interest can be isolated by a variety of means including but not exclusively, from cDNA libraries, by RT-PCR and by PCR using a series of overlapping synthetic oligonucleotide primers, all using standard methods. . The nucleotide sequences for the known cell adhesion molecules are known and available, for instance, in GenBank. The cDNA can be tailored at the 5" and 3' ends to generate restriction sites, such that oligonucleotide linkers can be used, for cloning of the cDNA into a vector containing the cDNA for HA. This can be at the N or C-terminus with or without the use of a spacer sequence. The cell adhesion molecule cDNA is cloned into a vector such as pPPCOOOS (Figure 2), pScCHSA, pScNHSA , or pC4:HSA from which the complete expression cassette is then excised and inserted into the plasmid pSAC35 to allow the expression of the albumin fusion protein in yeast. The albumin fusion protein secreted from the yeast can then be collected and purified from the media and tested for its biological activity. For expression in mammalian cell lines a similar procedure is adopted except that the expression cassette used employs a mammalian promoter, leader sequence and terminator (See Example 1). This expression cassette is then excised and inserted into a plasmid suitable for the transfection of mammalian cell lines.
l:.\AMI'Lk Y.i: Preparation of inhibitory factors and peplides as HA fusion proteins (such as HA-antiviral, HA-antibiotic. HA-enzvme inhibitor and }IA-anti-alIereicproteins).
|P29) The cHNA for the peplide of interest such as an antibiotic peptide can be isolated by a variety of means including but not exclusively, from cDNA libraries, by RT-PCR and by PCR using a series of overlapping synthetic oligonucleotide primers, all using standard methods. The cDNA can be tailored at the 5' and 3' ends to generate restriction sites, such that oligonucleotide linkers can be used, for cloning of the cDNA into a vector containing the cDNA for HA. This can be at the N or C-terminus with or without the use of a spacer sequence. The peptide cDNA is cloned into a vector such as pPPCOOOS (Figure 2), pScCHSA, pScNHSA , or pC4:HSA from which the complete expression cassette is then excised and inserted into the plasmid pSAC35 to allow the expression of the albumin fusion protein in yeast. The albumin fusion protein secreted from the yeast can then be collected and purified from the media and tested for its biological activity. For expression in mammalian cell lines a similar procedure is adopted except that the expression cassette used employs a mammalian promoter, leader sequence and terminator (See Example I). This expression cassette is then excised and inserted into a plasmid suitable for the transfection of mammalian cell lines.
EXAMPLE 74: Preparation of targeted HA fusion proteins.
|1230| The cDNA for the protein of interest can be isolated from cDNA library or can be made synthetically using several overlapping oligonucleotides using standard molecular biology methods. The appropriate nucleotides can be engineered in the cDNA to form convenient restriction sites and also allow the attachment of the protein cDNA to albumin cDNA. Also a targeting protein or peptide cDNA such as single chain antibody or peptidcs, such as nuclear localization signals, that can direct proteins inside the cells can be fused to the other end of albumin. The protein of interest and the targeting peptide is cloned into a vector such as pPPCOOOS (Figure 2), pScCHSA, pScNHSA , or pC4:HSA which allows the fusion with albumin cDNA. In this manner both N- and C-tenninal end of albumin are fused to other proteins. The fused cDNA is then excised from pPPCOOOS and is inserted into a plasmid such as pSAC35 to allow the expression of the albumin fusion protein in yeast. All the above procedures can be performed using standard methods in molecular biology. The albumin fusion protein secreted from yeast can be collected and purified from the media and tested for its biological activity and its targeting activity using appropriate biochemical and biological tests.
EXAMPLE 75: Preparation of HA-enz\>mes fusions.
11231) The cDNA for the enzyme of interest can be isolated by a variety of means including but not exclusively, from cDNA libraries, by RT-PCR and by PCR using a series of overlapping synthetic oligonucleotide primers, all using standard methods. The cDNA can be tailored at the 5' and 3' ends to generate restriction sites, such that oligonucleotide linkers can be used, for cloning of the cDNA into a vector containing the cDNA for HA. This can be at the N or C-terminus with or without the use of a spacer sequence. The enzyme cDNA is cloned into a vector such as pPPCOOOS (Figure 2), pScCHSA, pScNHSA, or pC4:MSA from which the complete expression cassette is then excised and inserted into the plasmid pSAC35 to allow the expression of the albumin fusion protein in yeast. The albumin fusion protein secreted from the yeast can then be collected and purified from the media and tested for its biological activity. For expression in mammalian cell lines a similar procedure is adopted except that the expression cassette used employs a mammalian promoter, leader sequence and terminator (See Example 1). This expression cassette is then excised and inserted into a plasmid suitable for the transfection of mammalian cell lines.
EXAMPLE 76: Construct //> 2053, IFNb-HSA. Gem-ration.
|1232| Construct ID 2053, pEE12.1:!FNb.HSA, comprises DNA encoding an IFNb albumin fusion protein which has the full-length IFNb protein including the native IFNb leader sequence fused to the amino-terminus of the mature form of HSA in the NSO expression vector pEE12.1.
Cloning of IFNb cDNA
|1233) The polynucleotide encoding IFNb was PCR amplified using primers IFNb-l and IFNb-2, described below, cut with Bam HUCla 1, and ligaled into Bam \\\/Cl (1234) Two oligonucleotides suitable for PCR amplification of the polynucleotide encoding the full-length of IFNb, IFNb-l and IFNb-2, were synthesized:
IFNb-l: 5^GCGCGGAlCCGAATTCCGCCCCCyl7GACCAACAAGTGTCTCCTCCAAATTGCTCTCCTGrrGTGCTT
CTCCACTACAGCrCTrrCCATGAGCTACAACTTGCTTGG-3' (SEQ ID NO: 107)
IFNb-2. 5'-GCGCGCA_TCOATGAGCAACCTCACTCTTGTGTGCATCGTTTCGGA GGTAACCTGT-31 (SEQ ID NO: 108) (1235| The IFNb-l primer incorporates a Bain HI cloning site (shown underlined), an Eco RI cloning site, and a Kozak sequence (shown in italics), followed by 80 nucleotides encoding the first 27 amino acids of the full-length form of IFNb. In IFNb-2, the Cla I site (shown underlined) and the DNA following it are the reverse complement of DNA encoding the first 10 amino acids of the mature HSA protein (SEQ ID NO:]) and the last IS nucleotides are the reverse complement of DNA encoding the last 6 amino acid residues of iFNb (see Example 2). A PCR amplimer was generated using these primers, purified, digested with Bam HI and Cla 1 restriction enzymes, and cloned into the Bam HI and Cla I sites of the pC4;HSA vector. After the sequence was confirmed, an Eco RI fragment containing the IFNb albumin fusion protein expression cassette was subcloned into Kco RI digested pEE12.l.
[1236| Further, analysis of the N-terminus of the expressed albumin fusion protein by amino acid sequencing can confirm the presence of the expected IFNb sequence (see below).
|1237] IFNb albumin fusion proteins of the invention preferably comprise the mature form of HSA , i.e., Asp-25 to Leu 60, fused to either the N- or C- terminus of the mature form of IFNb, i.e., Met-22 to Asn-187. In one embodiment of the invention, IFNb albumin fusion proteins of the invention further comprise a signal sequence which directs the nascent fusion polypeptide in the secretory pathways of the host used for expression. In a further preferred embodiment, the signal peptide encoded by the signal sequence is removed, and the mature IFNb albumin fusion protein is secreted directly into the culture medium. IFNb albumin fusion proteins of the invention may comprise heterologous signal sequences including, but not limited to, MAP, INV, Ig, Fibulin B, Clusterin, Insulin-Like Growth Factor Binding Protein 4, variant HSA leader sequences including, but not limited to, a chimeric HSA/MAF leader sequence, or other heterologous signal sequences known in the art. In a preferred embodiment, IFNb albumin fusion proteins of the invention comprise the native IFNb. In further preferred embodiments, the IFNb albumin fusion proteins of the invention further comprise an N-tenninal methionine residue. Polynucleotides encoding these polypeptidcs, including fragments and/or variants, arc also encompassed by the invention.
Expression and Purification of Construct ID 2053. Expression in marine myeloma NSO cell-lines. [1238| Construct ID # 2053, pEG12.1:IFNb-HSA , was electroporated into NSO cells by methods known in the art (see Example 6).
Purification from NSO cell supernatant.
|1239] Purification of IFNb-HSA from NSO cell supernatant may involve Q-Sepharose anion exchange chromatography at pll 7.4 using a NaCl gradient from 0 to 1 M in 20 mM Tris-HCI, followed by Poros PI 50 anion exchange chromatography at pH 6.5 with a sodium citrate gradient from 5 to 40 mM, and diafiltrating for 6 DV into 10 mM citrate, pll 6.5 and 140 mM NaCl, the final buffer composition. N-terminal sequencing should yield the sequence MSYNLL which is the amino terminus of the mature form of IFNb. The protein has an approximate MW of 88.5 kDa. |1240| For larger scale purification, e.g., 50 L of NSO cell supernatant can be concentrated into ~8 to 10 L. The concentrated sample can then be passed over the Q-Sepharose anion exchange column (10 x 19 cm, 1.5 L) at pll 7.5 using a step clution consisting of 50 mM NaOAc, pll 6.0 and 150 mM NaCl. The cluted sample can then be virally inactivated with 0.75% Triton-X 100 for 60 min at room temperature. SDK-Reverse Phase chromatography (10 em x 10 cm, 0.8 L) can then he employed at pH 6.0 with 50 mM NaOAc and 150 mM NaCl, or alternatively, the sample can be passed over an SP-sepharose column at pH 4.8 using a step elution of 50 mM NaOAc, pH 6.0, and 150 mM NaCl. DV 50 filtration would follow to remove any viral content. Phenyl-650M chromatography (20 cm x 12 cm, 3.8 L) al pH 6.0 using a step clution consisting of 350 mM (NIU^SC^ and 50 mM NaOAc, or alternatively consisting of 50 mM NaOAc pll 6.0, can follow. Diafilrration for 6-8 DV will allow for buffer exchange into the desired final formulation buffer of cither 10 mM Na2HPO« + 58 mM sucrose + 120 mM NaCl, pH 7.2 or 10 mM citrate, pH 6.5, and 140 mM NaCl or 25 mM Na,HPO,, 100 mM NaCl, pH 7.2.
The activity of IFNb can be assayed using an in vitro ISRR-SEAP assay.
|1241| All type I Intcrferon proteins signal through a common receptor complex and a similar Jak/STAT signaling pathway that culminates in the activation of Interferon, "IFN", responsive genes through the Intcrferon Sequence Responsive Element, "ISRE". A convenient assay for type I 1FN activity is a promoter-reporter based assay system that contains multiple copies of the ISRE element fused to a downstream reporter gene. A stable HEIC293 cell-line can be generated and contains a stably integrated copy of an 1SRE-SEAP reporter gene that is extremely sensitive to type 1 IFNs and displays linearity over 5 logs of concentration.
Method nf Screening of IFNb-HSA NSO stable clones.
[1242] Construct 2053 was electroporated into NSO cells as described in Example 6. The NSO cells transfccted with construct ID # 2053 were screened for activity by testing conditioned growth media in the ISRE-SEAP assay. The ISRE-SEAP/293F reporter cells were plated at 3 x 104 cell/well in 96-well, poly-D-lysine coated, plates, one day prior to treatment. Reporter cells were treated with various dilutions (including but not limited to 1:500 and 1:5000) of conditioned supernatant or purified preparations of IFNb albumin fusion protein encoded by construct ID 2053 or rhlFNb as a control. The reporter cells were then incubated for 24 hours prior to removing 40 |iL for use in the SEAP Reporter Gene Chemiluminescent Assay (Roche catalog/* 1779842), Recombinant human Interfcron beta, "rhlFNb" (Biogen), was used as a positive control.
Result
|1243] The purified preparation of NSO expressed IFNb-HSA had a greater EC50 of 9.3 x 10' g/mL than rhlFNb (Biogen) which had an EC50 of 1.8 x lO-'Vml-(see Figure 4).
In vivo induction of OAS b\i an Interferon. Method
|1244| The OAS cnryme, 2'-5'- OligoAdenylate Synthetase, is activated at the transcriptional level by interferon in response to antiviral infection. The effect of interferon constructs can be measured by obtaining blood samples from treated monkeys and analyzing these samples for transcriptional activation of two OAS mRNA, p4l and p69. A volume of 0.5 mL of whole blood can be obtained from 4 animals per group at 7 different time points, day 0, day ', day 2, day 4, day 8, day 10, and day 14 per animal. The various groups may include injection of vehicle control,
intravenous and/or subcutaneous injection of either 30 |ig/kg and/or 300 ng/kg IFN albumin fusion protein on day 1, and subcutaneous injection of 40 ug/kg of Intcrfcron alpha (Schering-Plough) as a positive control on days 1,3, and 5. The levels of the p41 and the p69 mRNA transcripts can be determined by real-lime quantitative PCR (Taqman) using probes specific for p4! OAS :ind p6-OAS. OAS mP.NA levels nn be quantitated relative to 18S ribosomal RNA endogenous control.
In vivo induction of OAS by Jnterferon beta albumin fusion encoded by construct ID 2053. Method
11245] The activity of the HSA-IFNb fusion protein encoded by construct 2053 can be assayed in the in vivo OAS assay as previously described above under subsection heading, "In vivo induction of OAS by an Intcrferon".
EXAMPLE 77; Indications for IFNb albumin fusion proteins.
(1246| IFN beta albumin fusion proteins (including, but not limited to, those encoded by construct 2053) can be used to treat, prevent, ameliorate and/or detect multiple sclerosis. Other indications include, but are not limited to Viral infections including Severe Acute Respiratory Syndrome (SARS) and other coronavirus infections; filoviruses, including but not limited to Ebola viruses and Marburg virus; Arenaviruses, including but not limited to Pichende virus, Lassa virus, Junin virus, Machupo virus, Guanarito virus; and lyniphocytic choriomeningitis vims (LCMV); Bunyaviruses, including but not limited to Punla toro virus, Crimean-Congo hemorrhagic fever virus, sandfly fever viruses, Rift Valley fever virus, La Crosse virus, and hantaviruses; Flaviviruses, including but not limited to Yellow Fever, Banzi virus, West Nile virus, Dengue viruses, Japanese Encephalitis virus, Tick-borne 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, and Rubella virus; Orthopox viruses, including but not limited to Vaccinia, Covvpox, Smallpox, and Monkeypox; Herpesviruses; FluA/B; Respiratory Sincytial virus (RSV); paraflu; measles; rliinovimses; 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, Etnerg. Med. Clin. North. Am. 2002; 20(2):311-30 and Darling et al, Emerg. Med. Clin. North Am. 2002,20(2):273-309).
EXAMPLE 78: Construct ID 2249. IFNal-HSA. Generation.
11247] Construct ID 2249, pSAC35:IFNa2.HSA, comprises DNA encoding an IFNa2 albumin fusion protein which has the HSA chimeric leader sequence, followed by the mature form of lFNa2 protein, i.e., C1-E165, fused to the amino-terminus of the mature form of HSA in the yeast S. ccrevisiae expression vector pSAC35.
(Inning oflh'Nnl cDN4
|1248) The polynucleotide encoding !FNa2 was PCR amplified using primers !FNa2-l and !FNa2-2, described below. The PCR amplimer was cut with Sal \ICIa 1, and ligated into Xha l/Clii 1 cut pScCHSA. Construct ID #2249 encodes an albumin fusion protein containing the chimeric leader sequence of HSA, the mature form of IFNa2, followed by the mature HSA protein.
11249] Two oligonucleotides suitable for PCR amplification of the polynucleotide encoding the mature form of lFNa2, IFNa2-l and !FNa2-2, were synthesized:
IFNa2-l:5l.CGCGCGCGTCGACAAAAGATGTCiATCTCiCCTCAAACCCACA-3' (SEQ ID NO: 109)
IFNa2-2: S'-GCGCGCATCGA l'GAGCAACCTCACTCTrGTGTGCATCTrCCTTACTTC'lTAAACTTTCT-3' (SEQ ID NO: 11C) |1250| The !FNa2-l primer incorporates a Sal I cloning site (shown underlined), nucleotides encoding the last three amino acid residues of the chimeric HSA leader sequence, as well as 22 nucleotides (shown in bold) encoding the first 7 amino acid residues of the mature form of IFNa2. In IFNa2-2, the Cla I site (shown underlined) and the DNA following it are the reverse complement of DNA encoding the first 10 amino acids of the mature HSA protein and the last 22 nucleotides (shown in bold) are the reverse complement of DNA encoding the last 7 amino acid residues of !FNa2 (see Example 2). A PCR amplimer of IFNa2-HSA was generated using these primers, purified, digested with Sal 1 and Cla 1 restriction enzymes, and cloned into the Xlio I and Cla I sites of the pScCHSA vector. After the sequence was confirmed, the expression cassette encoding this IKNa2 albumin fusion protein was subcloned into Not 1 digested pSAC35.
[1251] Further, analysis of the N-terminus of the expressed albumin fusion protein by amino acid sequencing can confirm the presence of the expected !FNa2 sequence (see below)
11252] Other !FNa2 albumin fusion proteins using different leader sequences have been constructed by methods known in the art (see Example 2). Examples of the various leader sequences include, but are not limited to, inverlasc "INV" (constructs 2343 and 2410) and mating alpha factor "MAF" (construct 2366). These IFNa2 albumin fusion proleins can be- subcloned into mammalian expression vectors such as pC4 (constructs 2382) and pEE12.1 as described previously (see Example 5). !FNa2 albumin fusion proteins with the therapeutic portion fused C-terminus to HSA can also be constructed (construct 2381).
|1253] IFNa2 albumin fusion proteins of the invention preferably comprise the mature form of HSA , i.e., Asp-25 to Leu-609, fused to either the N- or C- terminus of the mature form of !FNa2, i.e., Cys-1 to Glu-165. In one embodiment of the invention, IFNa2 albumin fusion proteins of the invention further comprise a signal sequence which directs the nascent fusion polypeptide in the secretory pathways of the host used for expression.
In a further preterred embodiment, me signal pcptide encoded by the signal sequence is removed, and the mature IFNa2 albumin fusion protein is secreted directly into the culture medium. IFNa2 albumin fusion proteins of the invention may comprise heterologous signal sequences including, hut not limited to, MAP, INV, Ig, Fibulin B, Clusterin, Insulin-Like Growth Factor Binding Protein 4, variant HSA leader sequences including, but not limited to, a chimenc HSA/MAF leader sequence, or other heterologous signal sequences known in the art. In a preferred embodiment, !FNa2 albumin fusion proteins of the invention comprise the native !FNa2. In further preferred embodiments, the IFNa2 albumin fusion proteins of the invention further comprise an N-temiinal methioninc residue. Polynuclcotides encoding these polypeptides, including fragments and/or variants, are also encompassed by the invention.
Expression and Purification of Construct ID 2249.
Expression in yeast S. cerevisiae.
(1254] Transformation of construct 2249 into yeast S. cerevisiae strain BXP10 was carried out by methods known in the art (see Example 3). Cells can be collected at stationary phase after 72 hours of growth. Supernatants are collected by clarifying cells at 3000g for 10 min. Expression levels are examined by immunoblot detection with anti-HSA serum (Kent Laboratories) or as the primary antibody. The IFNa2 albumin fusion protein of approximate molecular weight of 88.5 kDa can be obtained.
Purification from yeast S. cerevisiae cell supernatant.
[1255] The cell supernatant containing IFNa2 albumin fusion protein expressed from construct ID #2249 in yeast S. cerevisiae cells can be purified either small scale over a Dyax peptide affinity column (see Example 4) or large scale by following 5 steps: diafiltration, anion exchange chromatography using DEAE-Sepharose Fast Flow column, hydrophobic interaction chromatography (HIC) using Butyl 650S column, cation exchange chromatography using an SP-Sepharose Fast Flow column or a Blue-Sepharose chromatography, and high performance chromatography using Q-sepharose high performance column chromatography (see Example 4). The IFNa2 albumin fusion protein may elute from the DEAE-Sepharose Hast Flow column with 100 - 250 mM NaCI, from the SP-Sepharose Fast Flow column with 150 - 250 rnM NaCI, and from the Q-Sepharose High Performance column at 5 - 7.5 mS/cm. N-terminal sequencing should yield the sequence CDLPQ (SEQ ID NO.98) which corresponds to the mature form of IFNa2.
The (iclivily ofIFN(i2 can be assayed using fin in vitro ISRE-SFAP assay.
Method
11256| The IFNa2 albumin fusion protein encoded by construct ID If 2249 can be tested for activity in the ISRE-SEAP assay as previously described in Example 76. Briefly, conditioned yeast supernatants were tested at a 1:1000 dilution for their ability to direct ISRE signal transduction on the 1SRE-SEAP/293F reporter cell-line. The 1SRE-SEAP/293F reporter cells were plated at 3 x 104 cell/well in 96-well, poly-D-lysine coated, plates, one day prior to treatment. The reporter cells were then incubated for 18 or 24 hours prior to removing 40 uL for use in the SEAP Reporter Gene Chcmiluminescent Assay (Roche catalog # 1779842). Recombinant human Interferon beta, "rhIFNb" (Biogen), was used as a positive control.
Result
[1257] The purified preparation of !FNa2-HSA demonstrated a relatively linear increase in the ISRE-SEAP assay over concentrations ranging from 10"' to 10' ng/mL(sce Figure 5) or 10"'° to 10' ng/mL(see Figure 6).
In vivo induction of OAS hy Interferon alpha fusion encoded by constntct ID 2249.
Method
|1258| The OAS enzyme, 2'-5'- OligoAdenylate Synthetase, is activated at the transcriptional level by interferon in response to antiviral infection. The effect of interferon constructs can be measured by obtaining blood samples from treated monkeys and analyzing these samples for transcriptional activation of two OAS mRNA, p41 and p69. A volume of 0.5 mL of whole blood was obtained from 4 animals per group at 7 different time points, day 0, day 1, day 2, day 4, day 8, day 10, and day 14 per animal. The various groups include vehicle control, intravenous injection of 30 ug/kg HSA-1FN on day 1, subcutaneous injection of 30 ng/kg of HSA-1FN on day 1, subcutaneous injection of 300 u.g/kg of HSA-1FN on day 1, and subcutaneous injection of 40 ug/kg of Interferon alpha (Schering-Plough) as a positive control on days 1, 3, and 5. The levels of the p41 and the p69 mRNA transcripts were determined by real-time quantitative PCR (Taqman) using probes specific for p41-OAi> and p69-OAS. OAS mRNA levels were quantitated relative to I8S ribosomal RNA endogenous control. The albumin fusion encoded by construct 2249 can be subjected to similar experimentation.
Results
[1259] A significant increase in mRNA transcript levels for both p41 and p6(J OAS was observed in HSA-intcrfcron treated monkeys in contrast lo IFNa treated monkeys (see Figure 7 for p41 data). The effect lasted nearly 10 days.
KXAMPI.K 79: Indications for IFNal Albumin Fusion Proteins.
|1260I IFN alpha albumin fusion protein (including, but not limited to, those encoded by constructs 2249, 2343, 2410, 2366, 2382, and 2381) can be used to treat, prevent, ameliorate, and/or detect multiple sclerosis. Other indications include, but are not limited to viral infections including Severe Acute Respiratory Syndrome (SARS) and other coronavirus infections; filoviruses, including but not limited to Ebola viruses and Marburg
virus; Arenaviruses, including out not limited to Pichende virus, Lassa virus, Junin virus, Machupo virus, Guanarito virus; and l>Tnphoc>tic chonomeningitis virus (LCMV); Bunyaviruses, including but not limited to Punta toro virus, Crimean-Congo hemorrhagic fever virus, sandfly fever viruses. Rift Valley fever virus, La Crosse virus, and hantaviruses; Flaviviruses, including but not limited to Yellow Fever, Banzi virus, West Nile virus, Dengue viruses, Japanese Kncephalitis virus, Tick-home 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, and Rubella virus; Orthopox viruses, including but not limited to Vaccinia, Cowpox, Smallpox, and Monkeypox; Herpesviruses; FluA/B; Respiratory Sincytial virus (RSV); paraflu; measles; rhinoviruses; adenoviruscs; Semliki Forest virus; Viral Hemorrhagic fevers; Rhabdoviruscs; Paramyxoviruscs, including but not limited to Nipah virus and Hcndra 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, eg., Moran, Emerg. Med. Clin. North. Am. 2002; 20(2):311-30 and Darling et al., Kmcrg. Med. Clin. North Am. 2002;20(2):273-309).
[1261] Preferably, the IFNa-albumin fusion protein or 1FN hybrid fusion protein is administered in combination with a CCR5 antagonist, further in association with at least one of ribavirin, 1L-2, IL-12, pentafuside alone or in combination with an anti-HIV drug therapy, e.g., HAART, for preparation of a medicament for the treatment of HIV-1 infections, HCV, or HIV-1 and HCV co-infections in treatment-naive as well as treatment-experienced adult and pcdiatric patients.
Example SO: Construct ID ft 3691. BNP-HSA. Generation.
|1262] Construct ID #3691, pC4:SPCON.BNPI-32/HSA, comprises DNA encoding a BNP albumin fusion protein which has a consensus leader sequence, seerecon, followed by the processed, active BNP peptide (amino acids 1-32) fused to the ammo-terminus of the mature form of USA in the mammalian expression vector pC4.
Cloning afBNP cDNA for construct 3691
|1263) The polynucleotide encoding BNP was PCR amplified using primers BNP-1 and BNP-2, described below, cut with Sam Hl/C/a 1, and ligated into Ham HI/C/rt 1 cut pC4:HSA resulting in construct ID # 3691. Construct ID # 3691 encodes an albumin fusion protein containing a consensus leader sequence (SEQ ID No:l 11) and the processed, active form of BNP, followed by the mature HSA protein (see SEQ ID No:321 for construct 3691 in Table 2).
[1264] Two oligonuclcotides suitable for PCR amplification of the polynucleotide encoding the active, processed form of BNP, BNP-1 and BNP-2, were synthesized:
BNP-1: y-G\GCGCGGKTCCAAGCrTCCGCCA.TCATGTGGTGGCGCCTGTGGrGGCTGCTGCTGCTGCTGCTGCTGCTGTG
CCCC^rocrcrCCCCCAGCCCCAAGCTGGTGCAAGG -3' (SEQ ID NO:463)
BNP-2: 5'-AG'rCCC,VrCGAIGAGCAACCTCACTCTTGTGTGCATCATGCCGCCTCAGCACTTrGC-3' (SEQ ID NO:464). [1265| BNP-1 incorporates a Ham HI cloning site (underlined), polynuclcotides encoding a consensus leader sequence (SEQ ID Ncxlll) (italicized), and polynucleotides encoding the first seven amino acid sequence of BNP (bolded). In BNP-2, the underlined sequence is a Cla I site, and the polynuclcotides that follow it contains the reverse complement of DNA encoding the last 6 amino acids of BNP (bolded) and the first 10 amino acids of the mature USA protein. Using these two primers the BNP protein was PCR amplified. Annealing and extension temperatures and limes must be empirically determined for each specific primer pair and template.
112661 The PCR product was purified (for example, using Wizard PCR Preps DNA Purification System (Promega Corp)) and then digested with Bum HI and Cla I. After further purification of the Dam Hl-C/fl 1 fragment by gel clectrophorcsis, the product was cloned into Bam HI Ida \ digested pC4:HSA to produce construct ID # 3691. The expression construct was sequence verified. Expression and Purification of Construct ID 3691. Expression in 293F cells. [1267| Construct ID# 3691, pC4:SPCON.BNPl-32/HSA, was transfected into 293F cells by methods known in the art (see Example 6).
Purification from 293F cell supernatant.
[1268] Two liters of supernatant were collected 3 days post-transfection. The recombinant protein was captured by 5 ml Blue Sepharose CL-6B column (Amersham Biosciences, Piscataway, NJ, USA) and elutcd by 2 M NaCl. The material was bound to HiPrep 16/10 Phcnyl FF (high sub) column and eluted by 20 mM MES, pH 6.7. BNP-HSA was further purified by hydroxyapatite column chromatography in sodium phosphate buffer gradient (0-20 mS/cm in 200 ml) at pH 6.8. The final product was exchanged into PBS pH 7.2 by a HiPrep 26/10 desalting column (Amersham Bioscicnces).
The activity of BNP-HSA c.nn be assayed using an in vitro NPR-AfcGMP Assay.
|1269| Natriuretic peptide rcccptor-A (NPR-A) is the signaling receptor for BNP, and as such, is responsible for most of BNP's biological effects. DNP bioactivity is mediated by NPR-A guanylyl cyclase domain that converts GTP to cGMP upon activation. A convenient assay for BNP activity is to measure the BNP stimulation of a 293F cell line that stably over-expresses NPR-A. The cGMP production in the cells after exposure to BNP can he measured by cGMP EL1SA.
Method of Screening NPR-A 293 f stable clones.
[1270| The open reading frame of human NPR-A was constructed into pcDNA3.1 expression vector (Invitrogen). 293F cells were stably transacted with the plasmid DNA by Lipofectamine method and selected by 0.8 fig/ml G418. 293F/NPR-A stable clones were screened for best response to recombinant BNP.
Measurement ofcGUP derivation.
[1271] cGMP activation by BNP was carried out in 293F/NPK-A ceils and measured, by CatchPoint cyclic-GMP fluorescent assay kit (Molecular Devices, Sunnyvale, CA, USA). Briefly, 50,000 cells/well of 2931VNPR-A cells cultured in a 96-well plate were washed into 80 fil prestimulation buffer (Krebs-Ringer Bicarbonate Buffer with 10 mM glucose, pH 7.4, 15 nM sodium bicarbonate, and 0.75 mM 3-isobuty]-l-mcthylxanthine). BNP-HSA or recombinant BNP in 40 ul prestimulation buffer was added to the cells at 37°C for 10 min. The cells were lysed with 40 u.1 Lysis Buffer for 10 min with shaking. The amounts of cGMP in the lysates were quantitated as per the manufacturer's instruction.
Result
11272J The dose-response relationship of BNP-HSA and recombinant BNP were determined (see Figure 8). The maximal activities of Construct ID #3691 and recombinant BNP were similar (1.63 ± 0.016 vs. 1.80 ±0.016 pm, respectively), with F.C50 values of 28.4 ± 1.2, and 0.46 •t 1.1 nM, respectively.
BNP-HSA decreases blood pressure in vivo,
Method
[1I73| BNP reduces blood pressure by direct vasodilation as well as by suppression of renin/angiotensin/endothelin/aldosterone systems. The ability of BNP-HSA to decrease arterial blood pressure was tested in Ihree-month old male spontaneously hypertensive rats purchased from Taconic (Gennantown, NY, USA). Spontaneously hypertensive rats are genetically hypertensive with onset of high blood pressure after three months of age. BNP-HSA or recombinant BNP was reconstituted in 0.3 cc PBS per rat. The drugs were delivered via tail vein injection. Systolic and diaslolic blood pressures were recorded by cuff-tail method using XBP-1000 System (Kent Scientific, Torrington, CT, USA). For each blood pressure data point, 4-5 consecutive readings were taken and averaged. Mean arterial pressure (MAP) was calculated as 1/3 systolic pressure + 2/3 diastolic pressure. For dose-response determination, blood pressures were measured 20 h afterpC4:SPCON.BNPl-32/HSA administration at doses of 0.5, 2, 6, and 18 nmol/kg.
Result
(I274| The typical systolic pressure of spontaneously hypertensive rats was 180-200 mmHg before dosing. A single bolus of 6 nmol/kg BNP-I13A delivered via tail vein intravenous injection lowered both systolic and diastolic pressure, which accounted for more than 30 mmHg mean arterial pressure (MAP) reduction. The lowered blood pressure was steady and continued for a day and then gradually relumed to the baseline over several days (see Figure 9). In contrast, due to its instantaneous clearance, a single 6 nmol/kg bolus of recombinant BNP, produced only a very transient MAP decrease of about ~15 mmHg.
11275] In addition, the dose-response 20 hours post injection of a bolus of BNP-HSA was determined in four spontaneously hypertensive rats. 0.5 nmol/kg BNP-1ISA had an average of 7 mmHg MAP reduction, while 6 nmol/kg BNP-HSA had an average of 30 mmHg MAP reduction, and a high dose of 18 nmol/kg BNP-HSA only lowered the blood pressure slightly more than 6 nmol/kg.
In^vivo induction of plasma cfjMP by BNP-HSA.
Method
|1276| The intracellular cGMP activation by BNP results in its release from the cell to circulation. The plasma cGMP level correlates with BNP-induced cardiovascular and renal physiology. Plasma cGMP has been used as a biomarker for in vivo BNP action. To test the induction of plasma cfiMP by BNP-HSA in vivo, eleven- to 12-weck-old male C57/BL6 mice received a single bolus of recombinant BNP or BNP-HSA at a 6 nmol/kg dose via tail vein Plasma was prepared from the tail bleeds at 5, 10, 20, 40, and 80 min time points for the recombinant BNP dosing group and at additional 640, 1440, 2880, and 5760 min for the BNP-HSA group. Plasma samples from mice treated with PBS as the vehicle control were collected as the zero time points. cGMP levels were determined by Catch Point cyclic-GMP fluorescent assay kit according to the manufacture's instruction.
Result
11277] Following a single intravenous bolus of 6 nmol/kg recombinant BNP or BNP-HSA, peak plasma cGMP levels over the baseline were increased 3.9- or 5,6-fold, respectively (see Figure 10). In addition, the one-phase exponential decay half-life of cGMP following recombinant BNP treatment was 16 min (10 to 42 rnm, 95%CI), while the one-phase exponential decay half-life of cGMP following BNP-HSA administration was 1538 min (1017 to 3153 min, 95%CI).
In vivo Phfinnacokinctic fiiitilysis nfRNP albumin fusion encoded hy construct ID 3691.
Method
|I278| Eleven- to 12-week-old male C57/BL6 mice (obtained from Ace Animals, Boyertown, PA, USA) weighed 25.1 ± 0.12 g at the time of the study All animals were dosed at a volume of 10 ml/kg body weight. Predose animals were injected with PBS. Recombinant BNP was injected intravenously in the tail or subcutaneously in the mid-scapular region.
(Table Removed)
|1280] Blood was sampled from the inferior vena cava, placed into an EDTA-coated microtainer, and stored on ice. The samples were
centrifuged in a microcentrifuge at 14,000 rpm (16,000 *g) for 10 minutes at room temperature. The plasma was transferred into cluster tubes and
stored at -80°C.
|1281] BNP-HSA concentrations in plasma samples were determined using BNP ELA Kit (Phoenix Pharmaceutical, Belmont, CA, USA). The
standard curves were generated at the same time on the same plate with testing samples. The detection limit was 0.11 ng/mL for recombinant BNP.
The assay detects recombinant BNP and does not cross react to mouse BNP.
11282] Analysis was conducted by noncompartmental methods (WinNonlin; version 4.1; Pharsight Corp..Mountain View, CA, USA). The mean
plasma concentration at each time was used in the analysis. A linear up/log down trapezoidal method was used to calculate the AUCO ,.
Extrapolation to infinity AUC0^ was done by dividing the last observed concentration by the terminal elimination rate constant. Data were
uniformly weighted for these analyses.
Result
[1283] The mean baseline concentration of BNP-IISA in plasma as detected in the pre-dose samples was approximately 0.081-0.095 ng/ml. Following a single intravenous or subcutaneous injection, BNP-HSA had terminal elimination half-lives of 11.2 (intravenous delivery) or 19.3 h (subcutaneous delivery), while the half-life of recombinant BNP in mice was 3.1 min. Non compartmental analysis of BNP-HSA revealed that BNP-HSA had the following characteristics:
(Table Removed)
[1284] Five points at the terminal phase of the intravenous profile and four points at the terminal phase of the subcutaneous profile were selected for the terminal half-life calculation. The resulting AUC during this terminal phase was approximately 10% of the total AUC for the intravenous and subcutaneous profiles, respectively. This is compared to only 2% and 4% of the total AUC for the intravenous and subcutaneous profile, respectively, when the last three points were selected for the terminal half-life calculation.
EXAMPLE 81: Construct ID ft 3618. BNP(2X)-HSA. Generation.
|1285| Construct ID # 3618, pC4:SPCON.BNPl-32(2x)/HSA, comprises DMA encoding a BNP albumin fusion protein which has a consensus leader sequence, secreton, followed by two processed, active BNP pcptidcs (amino acids 1-32) in tandem fused to the ammo-terminus of the mature form of USA in the mammalian expression vector pC4.
Clonine of BNP cDNA (or construct 36! 8
J1286] The polynucleotide encoding the duplicate BNP moiety was first PCR amplified from the processed active form of BNP (amino acids I-32) using four primers BNP-1, BNP-2, BNP-3, and BNP-4, described below, to create two fragments A and B. Following amplification, two purified fragments (A and B) were mixed in an equal molar amount and used as a PCK template and amplified with primers BNP-5 and BNP-6, as
described below. the UNf(tx.) insert was then digested with Bnmlil and Cla\ and ligated into pC4HSA vector pre-digested with BamHl and Claresulting in construct 3618. Construct ID # 3618 encodes an albumin fusion protein containing a consensus leader sequence, secrecon (SEQ ID
No.l 1 0, and two, tandem copies of the processed, active form of BNP, followed by the mature HSA protein (see SEQ ID No:483 for construct
3618 in Table 2).
|I287| Four oligonucleotides suitable for PCR amplification of the polynucleotides encoding two fragments of BNP protein were first
.synthesized;
BNP-1 5'AGCCCCAAGATGGTGCAAGGGTCTGGCTGCTTTGGGAGGAAGATGGACCGGATCAGCTCCTCCAGTG
GCTGGGCT GCAAAGTGCTGAGGCGGCAT-3' (SEQ ID N0:486)
BNP-2 5'-CCrTGCACCATCTTGGGGCTATGCCGCCTCAGCACTTI'GC-3' (SEQ ID NO:487) HNP-3 5'-GCAAAGTGCTGAGGCGGCATAGCCCCAAGATGGTGCAAGG-3' (SEQ ID NO:488)
BNP-4 5'-AGTCCCATCGATGAGCAACCTCACTCTTGTGTGCATCATGCCGCCTC AGCACTTTGC-3' (SEQ ID N0:489) [1288] Using primer sets BNP-l/BNP-2 and BNP-3/BNP-4, two BNP proteins fragments (A and B, respectively) were PCR amplified. Annealing and extension temperatures and limes must be empirically determined for each specific primer pair and template. Fragments A and B were purified (for example, using Wizard PCR Preps DNA Purification System (Promega Corp)), mixed at equal molar amounts, and used as a template for PCR amplification using two additional oligonucleotides suitable for PCR amplification, BNP-5 and BNP-6:
BNP-5: y-GAGCCCGG^VCC\AGCT\~CCGCC\TCATGTGGTGGCGCCTGTGGTGGCTGCTGCTGCTGCTGCTGCTGCT
GrGGCCC/!?'GGrG7"GGGCCAGCCCCAAGCTGGTGCAAGG -3' (SEQ ID N0:463)
BNP-6: 5'-AGTCCCATCGATGAGCAACCTCACTCTTGTGTGCATCATGCCGCCTCAGCACTTTGC-3' (SEQ ID N0:464) |1289| BNP-5 incorporates a Sam HI cloning site (underlined), polynucleotides encoding a consensus leader sequence (SEQ ID No:lll) (italicized), and polynucleotides encoding the first seven amino acid sequence of BNP (bolded). In BNP-6, the underlined sequence is a Cla I site, and the polynucleotides that follow it contains the reverse complement of DNA encoding the last 6 amino acids of BNP (bolded) and the first 10 amino acids of the mature HSA protein. Using these two primers, a consensus leader sequence and two tandem copies of active BNP peptides were PCR amplified. Annealing and extension temperatures and times must be empirically determined for each specific primer pair and template. |1290| The PCR product was purified (for example, using Wizard PCR Preps DNA Purification System (Promega Corp)) and then digested with Bam HI and Cla I. After further purification of the Bam \\l-Cla I fragment by gel electrophoresis, the product was cloned into Bam HI /Cla I digested pC4:HSA to produce construct ID # 3618. The expression construct was sequence verified. Expression and Purification of Construct ID #3618. Expression in 293F cells. 11291] Construct ID # 3618, pC4:SPCON.BNPl-32(2x)/HSA, was transfected into 293F cells by methods known in the art (see Example 6).
Purification from 293F cell supernatant.
11292| pC4:SPCON.BNPl-32(2x)/HSA encoded by Construct ID # 3618 was purified as previously described above in Example 80 under subsection heading "Purification from 203F cell supernatant."
The Activity ofBNP{2X)-HSA can be assayed using an In Vitro NPR-A/cGMP Assay.
|1293] The activity of BNP(2X)-HSA encoded by Construct ID # 3618 can be assayed in vitro using an NPR-A/cGMP assay as previously described in Example 80 under subsection heading, "The activity of BNP-HSA can be assayed using an in vitro NPR-A/cGMP Assay," and "Method of Screening NPR-A 203F Stable Clones."
Result
|1294] The dose-response relationship of BNP(2X)-HSA and recombinant BNP were determined (see Figure 8). The maximal activities of BNP(2X)-HSA encoded by Construct ID #3618, and recombinant BNP were similar (1.68 ± 0.021, vs. 1.80 ± 0.016pm, respectively), with EC50 values of 9.8 ± 1.1, and 0.46 ± 1.1 nM respectively.
|I29S| The entire disclosure of each document cited (including patents, patent applications, patent publications, journal articles, abstracts, laboratory manuals, books, or other disclosures) as well as information available through Identifiers specific to databases such as GenBank, GeneScq, or the CAS Registry, referred to in this application are herein incorporated by reference in their entirety.
(1296] Furthermore, the specification and sequence listing of each of the following U.S. applications are herein incorporated by reference in their entirety: U.S. Application No. 60/542,274, filed February 4, 2005; U.S. Application No. 60/549,901, filed March 5, 2004; U.S. Application No. 60/556,906, filed March 29, 2004, and U.S. Application No. 60/636,603, filed December 17, 2004.
INDICATIONS RELATING TO DEPOSITED BIOLOGICAL MATERIAL
(PCTRulel3to)
The indications made below relate to the deposited biological material referred to in Table 3, page 25 of
(Table Removed)
The applicant requests that, until either a Canadian patent has been issued on the basis of an application or the application has been refused, or is abandoned and no longer subject to reinstatement, or is withdrawn, the Commissioner of Patents only authorizes the furnishing of a sample of the deposited biological material referred to in the application to an independent expert nominated by the Commissioner, the applicant must, by a written statement, inform the international Bureau accordingly before completion of technical preparations for publication of the international application.
NORWAY
The applicant hereby requests that the application has been laid open to public inspection (by the Norwegian Patent Office), or has been finally decided upon by the Norwegian Patent Office without having been laid open inspection, the furnishing of a sample shall only be effected to an expert in the art. The request to this effect shall be filed by the applicant with the Norwegian Patent Office not later than at the time when the application is made available to the public under Sections 22 and 33(3) of the Norwegian Patents Act. If such a request has been filed by the applicant, any request made by a third party for the furnishing of a sample shall indicate the expert to be used. That expert may be any person entered on the list of recognized experts drawn up by the Norwegian Patent Office or any person approved by the applicant in the individual case.
AUSTRALIA
The applicant hereby gives notice that the furnishing of a sample of a microorganism shall only be effected prior to the grant of a patent, or prior to the lapsing, refusal or withdrawal of the application, to a person who is a skilled addressee without an interest in the invention (Regulation 3.25(3) of the Australian Patents Regulations).
FINLAND
The applicant hereby requests that, until the application has been laid open to public inspection (by the National Board of Patents and Regulations), or has been finally decided upon by the National Board of Patents and Registration without having been laid open to public inspection, the furnishing of a sample shall only be effected to an expert in the art.

UNITED KINGDOM
The applicant hereby requests that the furnishing of a sample of a microorganism shall only he made available to an expert. The request to this effect must be filed by the applicant with the International Bureau before the completion of the technical preparations for the international publication of the application.
DENMARK
The applicant hereby requests that, until the application has been laid open to public inspection (by the Danish Patent Office), or has been finally decided upon by the Danish Patent office without having been laid open to public inspection, the furnishing of a sample shall only be effected to an expert in the art. The request to this effect shall be filed by the applicant with the Danish Patent Office not later that at the time when the application is made available to the public under Sections 22 and 33(3) of the Danish Patents Act. If such a request has been filed by the applicant, any request made by a third party for the furnishing of a sample shall indicate the expert to be used. That expert may be any person entered on a list of recognized experts drawn up by the Danish Patent Office or any person by the applicant in the individual case.
SWEDEN
The applicant hereby requests that, until the application has been laid open to public inspection (by the Swedish Patent Office), or has been finally decided upon by the Swedish Patent Office without having been laid open to public inspection, the furnishing of a sample shall only be effected to an expert in the art. The request to this effect shall be filed by the applicant with the International Bureau before the expiration of 16 months from the priority date (preferably on the Form PCT/RO/134 reproduced in annex Z of Volume I of the PCT Applicant's Guide). If such a request has been filed by the applicant any request made by a third party for the furnishing of a sample shall indicate the expert to be used. That expert may be any person entered on a list of recognized experts drawn up by the Swedish Patent Office or any person approved by a applicant in the individual case.
NETHERLANDS
The applicant hereby requests that until the date of a grant of a Netherlands patent or until the date on which the application is refused or withdrawn or lapsed, the microorganism shall be made available as provided in the 31F(1) of the Patent Rules only by the issue of a sample to an expert. The request to this effect must be furnished by the applicant with the Netherlands Industrial Property Office before the date on which the application is made available to the public under Section 22C or Section 25 of the Patents Act of the Kingdom of the Netherlands, whichever of the two dates occurs earlier.







We claim:
1. An albumin fusion protein comprising two or more tandemly oriented Glucagon-Like-Peptide 1 (GLP-1) polypeptides, wherein said GLP-1 polypeptides are selected from wild-type GLP-1, GLP-1 (9-36), GLP-1 (7-36), and GLP-1 (7-36(A8G)) fused to albumin wherein said albumin increases the serum plasma half-life of unfused GLP-1 polypeptides, and wherein said fusion protein has GLP-1 activity, and wherein the albumin fusion protein further comprises a modified HSA/Kex2 leader sequence.
2. The albumin fusion protein as claimed in claim 1, wherein said tandemly oriented GLP-1 polypeptides are selected from at least one wild-type GLP-1 sequence fused to at least one GLP-1 polypeptide selected from GLP-1 (9-36), GLP-1 (7-36), and GLP-1 (7-
36(A8G)).
3. The albumin fusion protein as claimed in claim 1, wherein said at least one GLP-1
polypeptide is selected from
(a) GLP-1 (9-36);
(b) GLP-1 (7-36); and
(c) GLP-1(7-36(A8G)).

4. The albumin fusion protein as claimed in claim 3, wherein said GLP-1 polypeptides are selected from two tandemly oriented GLP-1 (7-36(A8G)).
5. The albumin fusion protein as claimed in claim 1, produced from a host cell comprising a construct which expresses said albumin fusion protein, wherein said construct is selected from the following constructs defined in Table 2:

(a) Construct ID 3610; and
(b) Construct ID 3696.
6. The albumin fusion protein as claimed in claim 1 comprising two or more tandemly
oriented GLP-1 polypeptides fused to albumin, wherein said GLP-1 polypeptides
comprise at least one of an amino acid sequence selected from the group consisting of:
(a) SEQ ID NO:387; and
(b) SEQ ID NO:390.

7. The albumin fusion protein as claimed in claim 1 comprising two or more tandemly
oriented GLP-1 polypeptides fused to albumin, wherein said albumin fusion protein
comprises an amino acid sequence selected from the group consisting of:
(a) SEQ ID NO:319;and
(b) SEQ ID NO:322.

8. The albumin fusion protein as claimed in claim 1, wherein said at least two tandemly oriented GLP-1 polypeptides are fused at the N-terminus or at the C-terminus to said albumin.-
9. The albumin fusion protein as claimed in claim 8 wherein said albumin is human albumin.
10. The albumin fusion protein as claimed in claim 1 comprising SEQ ID NO: 1.
11. The albumin fusion protein as claimed in claim 1 wherein said two or more tandemly oriented GLP-1 polypeptides are genetically fused to said albumin.
12. A polynucleotide encoding the albumin fusion protein as claimed in claim 1.
13. A vector comprising the polynucleotide as claimed in claim 12.
14. An isolated host cell comprising the vector as claimed in claim 13.
15. The isolated host cell as claimed in claim 14, which is a yeast cell.
16. The isolated host cell as claimed in claim 15, wherein said yeast cell is S. cerevisiae, a K. lactis or a P. pastoris.
17. The isolated host cell as claimed in claim 15, wherein said yeast is glycosylation deficient.
18. The isolated host cell as claimed in claim 15, wherein said yeast is glycosylation and

protease deficient.
19. The albumin fusion protein as claimed in claim 1, which is expressed in yeast.
20. The albumin fusion protein as claimed in claim 19, wherein said yeast is S. cerevisiae, a K. lactis or a P. pastoris.
21. The albumin fusion protein as claimed in claim 19, wherein said yeast is glycosylation deficient.
22. The albumin fusion protein as claimed in claim 19, wherein said yeast is glycosylation and protease deficient.
23. The albumin fusion protein as claimed in claim 1, wherein the leader sequence is a modified HSA/kex2 leader sequence comprising amino acids of SEQ ID NO:l 12.
24. A composition comprising the albumin fusion protein as claimed in claim 1 and a pharmaceutically acceptable carrier such as mannitol, polysorbate 80, and phosphate
buffer at pH 7.2. .
25. A method of producing an albumin fusion protein comprising
(a) transforming a host cell comprising at least one vector as claimed in claim 13;
(b) culturing the host cell in suitable conditions for expression of the albumin fusion protein; and
(c) isolating the albumin fusion protein.
26. The method as claimed in claim 25, wherein the isolated albumin fusion protein undergoes further processing to remove the leader sequence.

Documents:


Patent Number 259504
Indian Patent Application Number 4362/DELNP/2006
PG Journal Number 12/2014
Publication Date 21-Mar-2014
Grant Date 14-Mar-2014
Date of Filing 28-Jul-2006
Name of Patentee HUMAN GENOME SCIENCES INC.
Applicant Address 14200 SHADY GROVE ROAD, ROCKVILLE, MARYLAND 20850,USA
Inventors:
# Inventor's Name Inventor's Address
1 CRAIG A. ROSEN 22400 ROLLING HILL LANE, LAYTONSVILLE, MARYLAND 20882, USA
2 ADAM BELL 13312 BURNT WOODS PLACE, GERMANTOWN, MARYLAND 20874, USA
3 DAVID LAFLLEUR 3142 QUESADA STREET, N.W., WASHINGTON, D 20015, USA
4 WILLIAM A. HASELTINE 3053 P. STREET, N.W. WAHINGTON, D 20007, USA
5 PAUL A. MOORE 7013 OLD GATE ROAD, NORTH BETHESDA, ,MARYLAND 20852, USA
6 JASON B. BOCK 14200 SECLUDED LANE, NORTH POTOMAC, MARYLAND 20878, USA
7 YANGGU SHI 710 SUFFIELD DRIVE, GAITHERSBURG, MARYLAND 20878, USA
PCT International Classification Number C07K1/00
PCT International Application Number PCT/US2005/004041
PCT International Filing date 2005-02-09
PCT Conventions:
# PCT Application Number Date of Convention Priority Country
1 60/542,274 2004-02-09 U.S.A.
2 60/636,603 2004-12-17 U.S.A.
3 60/549,901 2004-03-05 U.S.A.
4 60/556,906 2004-03-29 U.S.A.