WO2005044849A2 - Lp mammalian proteins; related reagents - Google Patents

Lp mammalian proteins; related reagents Download PDF

Info

Publication number
WO2005044849A2
WO2005044849A2 PCT/US2004/019099 US2004019099W WO2005044849A2 WO 2005044849 A2 WO2005044849 A2 WO 2005044849A2 US 2004019099 W US2004019099 W US 2004019099W WO 2005044849 A2 WO2005044849 A2 WO 2005044849A2
Authority
WO
WIPO (PCT)
Prior art keywords
polypeptide
sequence
protein
antibody
polynucleotide
Prior art date
Application number
PCT/US2004/019099
Other languages
French (fr)
Other versions
WO2005044849A3 (en
Inventor
Jai Pal Singh
Asavari Prasad Wagle
Original Assignee
Eli Lilly And Company
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Eli Lilly And Company filed Critical Eli Lilly And Company
Publication of WO2005044849A2 publication Critical patent/WO2005044849A2/en
Publication of WO2005044849A3 publication Critical patent/WO2005044849A3/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N9/00Enzymes; Proenzymes; Compositions thereof; Processes for preparing, activating, inhibiting, separating or purifying enzymes
    • C12N9/14Hydrolases (3)
    • C12N9/48Hydrolases (3) acting on peptide bonds (3.4)
    • C12N9/50Proteinases, e.g. Endopeptidases (3.4.21-3.4.25)
    • C12N9/64Proteinases, e.g. Endopeptidases (3.4.21-3.4.25) derived from animal tissue
    • C12N9/6421Proteinases, e.g. Endopeptidases (3.4.21-3.4.25) derived from animal tissue from mammals
    • C12N9/6424Serine endopeptidases (3.4.21)
    • C12N9/6437Coagulation factor VIIa (3.4.21.21)
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12YENZYMES
    • C12Y304/00Hydrolases acting on peptide bonds, i.e. peptidases (3.4)
    • C12Y304/21Serine endopeptidases (3.4.21)
    • C12Y304/21021Coagulation factor VIIa (3.4.21.21)

Definitions

  • FVIIa is a key initiator of the proteolytic cascade that leads to the formation of blood clot via the intrinsic pathway.
  • the primary function of FVIIa is the proteolytic activation of factor IX and factor X (FX) after complexation with the cell associated tissue factor (TF).
  • Activated FX (factor Xa) converts prothrombin to thrombin which in turn induces the formation of fibrin from fibrinogen (Martin, et al. 1998 Thromb. Res. 90, 1-25; Craner, et al. 1996 Thromb. Res. 81, 1-41).
  • FVIIa has been shown to act as a ligand for triggering TF mediated signaling pathway.
  • FVIIa-TF cellular pathway plays an important role in inflammation (Cunningham, et al. 1999 Blood 94, 3413-3420) and angiogenesis (Shoji, et al. 1998 Am. J. Pathol. 152, 399-411; Carmeliet, et al., Nature 383, 73-75).
  • FVIIa-TF pathway is critical for blood vessel development. Gene knock out studies have demonstrated that the mutant mice lacking TF have fragile vessels (Carmeliet, et al., Nature 383, 73-75).
  • FVIIa-TF in tumor angiogenesis is corroborated by high level expression of TF in tumors that correlates with metastatic state (Fernandez, and Rickles, 2002 Curr. Opin. Hematol. 9. 401-406). Consequently, there is a need in the fields of hematopoiesis, inflammation, and angiogenesis for novel compositions that can modulate and/or influence conditions or states relating to these fields.
  • the present invention satisfies such a need by disclosing a novel truncated FVIIa, designated LP FVIIa, which is generated by deletion of 38 amino acid residues from an N-terminad portion of native Factor Vila.
  • LP FVIIIa is devoid of Factor X activation activity and is a significantly more potent inducer of proangiogenic cytokines than native Factor Vila.
  • the present invention is based in part upon the discovery of novel Factor Vila LP compositions (herein designated as LP FVIIa compositions including without limitation, LPs disclosed with sequence identifiers and in a Table herein and variants thereof).
  • LP FVIIa compositions including without limitation, LPs disclosed with sequence identifiers and in a Table herein and variants thereof.
  • the invention provides substantially pure, isolated, and/or recombinant protein or polypeptide exhibiting identity over a length of at least about 12 contiguous amino acids to a corresponding sequence of a Table described herein; a fusion protein comprising a sequence as described herein.
  • an LP portion is at least about: 12, 15, 20, 25, 30, 35, 40, 45, 50, 55, 60, 65, 70, 75, 80, 85, 90, 95, 100, 110, 120, 130, 140, 150, 160, 170, 180, 190, 200, 210, 220, 230, 240, 250, 260, 270, 280, 290, 300, 310, 320, 330, 340, 350, 360, 370, 380, 390, 400, or 405 contiguous amino acid residues in length of a sequence described herein, or of SEQ ID NO: X or Y, or an LP sequence of Table 1.
  • an LP (e.g., LPFVII, or variants thereof) comprises a mature sequence (or a variant sequence) of Table 1; protein or peptide: is from a warm blooded animal selected from a mammal, including a primate; comprises at least one polypeptide segment of Table 1, exhibits a plurality of portions exhibiting identity to polypeptide segments of Table 1; is a natural allelic variant of the LPFVII; has a length at least about 30, 35, 40, 45, 50, 55, 60, 65, 70, 75, 80, 85, 90, 95, 100, 110, 120, 130, 140, 150, 160, 170, 180, 190, 200, 210, 220, 230, 240, 250, 260, 270, 280, 290, 300, 310, 320, 330, 340, 350, 360, 370, 380, 390, 400, or 405 contiguous amino acid residues exhibiting identity to a polypeptide segment of Table 1; exhibits at least
  • compositions comprising: a sterile LP FVIIa protein or peptide and a carrier, wherein the carrier is: an aqueous compound, including water, saline, and/or buffer; and/or formulated for oral, rectal, nasal, topical, or parenteral administration.
  • the invention further provides a binding compound comprising an antigen binding portion from an antibody, which specifically binds to a natural LP FVIIa protein or polypeptide, wherein: the protein or polypeptide is a primate protein; the binding compound is an Fv, Fab, or Fab2 fragment; the binding compound is conjugated to another chemical moiety; or the antibody: is raised against a peptide sequence of a mature polypeptide comprising sequence of Table 1, is raised against a mature LPFVII; is immunoselected; is a polyclonal antibody; binds to a denatured LPFVII; exhibits a Kd to antigen of at least 30 mM; is attached to a solid substrate, including a bead or plastic membrane; is in a ster
  • the present invention further provides an isolated or recombinant LP nucleic acid encoding a protein or peptide or fusion protein described above, wherein: the LP protein and/or polypeptide is from a mammal, including a primate; or the LP nucleic acid: encodes an antigenic peptide sequence from an LP FVIIa (or variant thereof) of Table 1; encodes a plurality of antigenic peptide sequences from an LP FVIIa of Table 1 , exhibits identity to a natural cDNA encoding the segment; is an expression vector; further comprises an origin of replication; is from a natural source; comprises a detectable label; comprises synthetic nucleotide sequence; is less than 6 kb, preferably less than 3 kb; is from a mammal, including a primate; comprises a natural full length coding sequence; is a hybridization probe for a gene encoding an LP family protein; or is a PCR primer, PCR product, or mutagenesis primer
  • LP FVIIa nucleic acid embodiments include those which: hybridize under wash conditions of at least 42°C, 45°C, 47°C, 50°C, 55°C, 60°C, 65°C, or 70°C and less than about 500 mM, 450 mM, 400 mM, 350 mM, 300 mM, 250 mM, 200 mM, 100 mM, to an LP nucleic acid sequence of Table 1) that exhibit identity over a stretch of at least about 30, 32, 34, 36, 38, 39, 40, 42, 44, 46, 48, 49, 50, 52, 54, 56, 58, 59, 75, or at least about 150 contiguous nucleotides to an LPFVII; in further embodiments, an LP nucleic acid embodiment which: hybridizes under wash conditions of at least 42°C, 45°C, 47°C, 50°C, 55°C, 60°C, 65°C, or 70°C and less than about 500 mM, 450 mM,
  • the wash conditions are at 55° C and/or 300 mM salt; 60° C and/or 150 mM salt; the identity is over a stretch is at least 35, 40, 45, 50, 55, 60, 65, 70, 75, 80, 85, 90, 95, 100, 110, 120, 130, 140, 150, 160, 170, 180, 190, or 200 nucleotides.
  • the invention provides a method of modulating physiology or development of a cell or tissue culture cells comprising introducing into such cell an agonist or antagonist of an LPFVII.
  • FIGURES Figure 1 Factor Vila induces cytokine expression in THP-1 cells:
  • Inset figure IC shows the time course of TF expression in THP-1 cells after the addition of PMA.
  • Cells extracts were prepared immediately (open bars), 24 hours (hatched bares) and 48 hours (solid bars) after treatment with PMA.
  • control or PMA treated cells were incubated without or with 50 nM FVIIa for 0 hour (open bars), 24 hours hatched bars or 48 hours (solid bars).
  • IL-6 was determined after incubation for the indicated interval (C).
  • THP-1 cells were treated with 10 nM PMA for 24 hours as in figure 1. Hirudin (lOOnM) or FXa (5nM) was then added. After 10 min, cells were treated with 50 nM FVIIa. IL-6 in culture supernatant collected after 24 hour treatment with FVIIa was determined as before.
  • FIG. 3 Factor X to Xa converting activity and IL-6 inducing activity of FVIIa:
  • FVIIa lug protein
  • A Coomassie Gelcode staining solution
  • PVD membrane for western blotting using a polyclonal FVIIa antibody.
  • Lanes indicate molecular weight standards (MW), 1, 2, 3 are preparation 1, 2 and 3.
  • B Shows the N- terminal amino acid sequence of the 52 kD protein band.
  • C Shows the N-terminal amino acid sequence of the light chain of the 46 kD protein. The protease domain N-terminal was same as in figure B.
  • D Shows the complete amino acid sequence of FVIIa with N- terminal of the light chain starting with H2N-ANAFLEE ) and the N-terminal of the protease domain (153 -H2N-IVGGKVC— ).
  • Preparation 2 containing the native and the truncated FVIIa was fractionated using a mono S anion exchange FPLC column.
  • the monoS column ( ⁇ x cm) was equilibrated with Tris-HCL buffer, pH 7.4. Twenty-five microliters of prep 2 was loaded to the column and then eluted with a linear gradient of 0.1M-0.5M NaCl in the equilibrating buffer. One ml fractions were collected. Column effluent was monitored for protein (absorption at 280nm) using a online detector (diamonds figure 5,). Aliquots from each fraction were subjected to SDS-PAGE and western blotting (A). (B) Each fraction was assayed for FX to Xa converting activity (triangles) and IL-8 induction in THP-1 cells (squares).
  • THP-1 cells expressing TF (24-hour treatment with PMA) were treated with the indicated concentrations of the compounds. Factor Xa activity was then determined.
  • THP-1 cells expressing TF were treated with the indicated concentrations of compounds for 10 min before addition of 50 nM FVIIa. IL-8 in the supernatants collected after 24- hour treatment with FVIIa was then determined using an ELISA.
  • references to “an organism” includes, e.g., one or more different organisms
  • reference to “a cell” includes, e.g., one or more of such cells
  • reference to “a method” include, e.g., reference to equivalent steps and methods known to a person of ordinary skill in the art, and so forth.
  • all technical and scientific terms used herein have the same meaning as commonly understood by a person of ordinary skill in the art to which this invention belongs. Although methods and materials similar or equivalent to those described herein can be used to practice or test the present invention, suitable methods and materials are described below. AU publications, patent applications, patents, and other references discussed herein are provided solely for their disclosure before the filing date of the present application.
  • LP FVIIa (SEQ ID NO: 2) is a primate polypeptide that is a novel N-terminad truncated factor Vila, which lacks factor X activating activity but acts as a potent stimulator of inflammatory cytokine expression.
  • LP FVIIa is generated by cleavage at amino acid residues Lys38-Leu39 leading to the deletion of the gama-carboxyglutamic acid (Gla) domain (See Table 1 below).
  • FVIIa is significantly more potent inducer of cytokine expression than the native factor Vila, suggesting that the N-terminal cleavage may serve as an activation mechanism for generating a potent proinflammatory/proangiogenic form of factor Vila.
  • the coagulation factor Vila (FVIIa) is a key initiator of the proteolytic cascade that leads to the formation of blood clot via the intrinsic pathway.
  • the primary function of FVIIa is the proteolytic activation of factor IX and factor X (FX) after complexation with the cell associated tissue factor (TF).
  • Activated FX converts prothrombin to thrombin which in turn induces the formation of fibrin from fibrinogen (Martin, et al.
  • FVIIa has been shown to act as a ligand for triggering TF mediated signaling pathway.
  • the FVIIa-TF cellular pathway plays an important role in inflammation (Cunningham, et al. 1999 Blood 94, 3413-3420) and angiogenesis (Shoji, et al. 1998 Am. J. Pathol. 152, 399-411; Carmeliet, et al., Nature 383, 73-75).
  • FVIIa induces a number of pro-angiogenic genes including VEGF, Egrl, CYR61, CTGF, FGF-5, IL-8, and MMP1, MMP13 (SNr, et al., 2000 J. Biol. Chem. 275, 6580-6585).
  • VEGF vascular endothelial growth factor
  • Egrl Egrl
  • CYR61 CYR61
  • CTGF FGF-5
  • IL-8 IL-8
  • MMP1 MMP13
  • PI3 kinase and Akt (Versteg, et al., 2000 J. Biol. Chem. 275, 28750-28756), a pathway consistent with angiogenesis activation.
  • the role of FVIIa-TF in tumor angiogenesis is corroborated by high level expression of TF in tumors that correlates with metastatic state (Fernandez, and Rickles, 2002 Curr. Opin. Hematol. 9. 401-406).
  • Tissue factor is also expressed at high levels in atherosclerotic plaques where it may play pathological role in thrombosis after plaque rupture Wilcox, et al., 1989 Proc. Natl. Acad. Sci. U.S.A.
  • FVIIa-TF IL-6, LIF, IL-8, IL-b, MMPs and CTGF
  • the growth factors produced in response to FVIIa may contribute to smooth muscle cell proliferation and intimal thickening (Sato, et al., 1997 Thromb. Haemost. 78, 1138-1141).
  • cytoplasmic domain of TF The role of cytoplasmic domain of TF was demonstrated using cells transfected with TF gene deficient of the cytoplasmic domain. Deletion of cytoplasmic domain abrogated TF- induced metastasis (Pendurthi & Rao 2002 Vitamins and Hormones 64, 323-355; Mueller, B., M., and Ruf, W. 1998 J. Clin. Invest. 101, 1372-1378). The cellular signaling involves recruitment of filamin upon phosphorylation of the cytoplasmic domain of TF (Pendurthi & Rao 2002 Vitamins and Hormones 64, 323-355). Thus, it is still not fully established whether the protease activity of FVIIa is essential or whether TF can act as a direct signaling receptor.
  • FVIIa induces cytokine expression in human macrophage THP-1 cells in a TF dependent manner.
  • the effect of FVIIa is independent of thrombin and FXa.
  • the coagulation activity (generation of factor Xa from factor X) is not required for cytokine induction by FVIIa.
  • the present invention discloses a novel truncated FVIIa, designated LP FVIIa, which is generated by deletion of 38 amino acid residues from an N-terminad portion of native Factor Vila.
  • This novel truncated Factor Vila is a novel composition that is devoid of Factor X activation activity and is a significantly more potent inducer of proangiogenic cytokines than the native Factor Vila.
  • LP FVIIa polynucleotide sequence (SEQ ID NO: 1) and corresponding polypeptide (SEQ ID NO: 2) .
  • the ORF for LP FVIIa is 1-1221 bp (with the start (ATG) and stop codons (TGA) identified in bold typeface and underlined in case numbering is misidentified one skilled in the art could determine the open reading frame without undue experimentation) .
  • IVGGKVCPKGECP QVLLLVNGAQLCGGTLINTIWVVSAAHCFDKIKN RNLIAVLG ⁇ HDLSEHDGDEQSRR VAQVIIPSTYVPGTTNHDIALLRLHQPWLTDHWPLCLP ⁇ RTFSERTLAFVRFSLVSG GQLLDRGATALE LMVLNVPRL TQDCLQQSRKVGDSPNITEYMFCAGYSDGS DSCKGDSGGPHATHYRGT YLTGIVS GQGC ATVGHFGVYTRVSQYIEWLQKLMRSEPRPGVLLRAPFP indicated below in italics.
  • Deleted portion of LP FVIIIa is indicated by underlining. 1 [ - • . . : 50
  • LP polynucleotide refers to a molecule comprising a nucleic acid sequence contained in a Table herein, in a sequence of SEQ ID NO:X or Y (where X or Y are general placeholder representations for any specific DNA or amino acid sequence identifier).
  • the polynucleotide can contain the nucleotide sequence of the full length cDNA sequence, including the 5' and 3' untranslated sequences, the coding region, with or without the signal sequence, the secreted protein coding region, as well as fragments, epitopes, domains, and variants of the nucleic acid sequence.
  • an "LP polynucleotide” also encompasses, e.g., those polynucleotides that stably hybridize, under stringent hybridization conditions, to a sequence contained in SEQ ID NO:X, or the complement thereof.
  • an LP polynucleotide sequence is at least 15, at least 30, at least 50, at least 100, at least 125, at least 500, or at least 1200 contiguous nucleotides but are less than or equal to 300 kb, 200 kb, 100 kb, 50 kb, 15 kb, 10 kb, 7.5 kb, 5 kb, 2.5 kb, 2.0 kb, or 1 kb, in length.
  • an LP polynucleotide sequence comprises a portion of a coding sequence, as disclosed herein, but does not comprise all or a portion of any intron.
  • the polynucleotides comprising coding sequences do not contain coding sequences of a genomic flanking gene (i.e., 5' or 3' to the sequence of interest in the genome).
  • an LP polynucleotide sequence do not contain the coding sequence of more than 1000, 500, 250, 100, 50, 25, 20, 15, 10, 5, 4, 3, 2, or 1 genomic flanking gene(s).
  • An LP polynucleotide sequence can be composed of any polyribonucleotide or polydeoxribonucleotide, which may be unmodified RNA or DNA or modified RNA or DNA.
  • polynucleotides can be composed of single- and double-stranded DNA, DNA that is a mixture of single- and double-stranded regions, single- and double-stranded RNA, and RNA that is mixture of single- and double-stranded regions, hybrid molecules comprising DNA and RNA that may be single-stranded or, more typically, double-stranded or a mixture of single-and double-stranded regions.
  • polynucleotide can be composed of triple-stranded regions comprising RNA or DNA or both RNA and DNA.
  • a polynucleotide may also contain one or more modified bases or DNA or RNA backbones modified for stability or for other reasons.
  • Modified bases can include, e.g., for example, tritylated bases and unusual bases such as inosine.
  • tritylated bases can include, e.g., for example, tritylated bases and unusual bases such as inosine.
  • a variety of modifications can be made to DNA and RNA; thus, the term "polynucleotide” embraces chemically, enzymatically, or metabolically modified forms.
  • altered nucleic acid sequences encoding LP include those sequences with deletions, insertions, or substitutions of different nucleotides, resulting in a polypeptide the same as LP or a polypeptide with at least one functional characteristic of LP. Included within this definition are polymorphisms which may or may not be readily detectable using a particular oligonucleotide probe of the polynucleotide encoding LP, and improper or unexpected hybridization to allelic variants, with a locus other than the normal chromosomal locus for the polynucleotide sequence encoding LP.
  • “Substantial similarity" in a nucleic acid sequence comparison context means either that the segments, or their complementary strands, when compared, are identical when optimally aligned, with appropriate nucleotide insertions or deletions, in at least about 50% of the nucleotides, generally at least 56%, more generally at least 59%, ordinarily at least 62%, more ordinarily at least 65%, often at least 68%, more often at least 71%, typically at least 74%, more typically at least 77%, usually at least 80%, more usually at least about 85%, preferably at least about 90%, more preferably at least about 95 to 98% or more, and in particular embodiments, as high at about 99% or more of the nucleotides.
  • substantial similarity exists when the segments will hybridize under selective hybridization conditions, to a strand, or its complement, typically using a sequence derived from SEQ ID X.
  • selective hybridization will occur when there is at least about 55% similarity over a stretch of at least about 30 nucleotides, preferably at least about 65% over a stretch of at least about 25 nucleotides, more preferably at least about 75%, and most preferably at least about 90% over about 20 nucleotides. See Kanehisa (1984) Nuc. Acids Res. 12:203-213.
  • sequence comparison typically one sequence acts as a reference sequence, to which test sequences are compared.
  • test and reference sequences are input into a computer, subsequent coordinates are designated, if necessary, and sequence algorithm program parameters are designated.
  • sequence comparison algorithm then calculates the percent sequence identity for the test sequence(s) relative to the reference sequence, based on the designated program parameters.
  • Optical alignment of sequences for comparison can be conducted, e.g., by the local homology algorithm of Smith and Waterman (1981) Adv. Appl. Math. 2:482, by the homology alignment algorithm of Needlman and Wunsch (1970) J. Mol. Biol. 48:443, by the search for similarity method of Pearson and Lipman (1988) Proc. Nat'l Acad. Sci.
  • PILEUP creates a multiple sequence alignment from a group of related sequences using progressive, pairwise alignments to show relationship and percent sequence identity. It also plots a tree or dendogram showing the clustering relationships used to create the alignment. PILEUP uses a simplification of the progressive alignment method of Feng and Doolittle (1987) J. Mol. Evol. 35:351-360.
  • the method used is similar to the method described by Higgins and Sharp (1989) CABIOS 5:151-153.
  • the program can align up to 300 sequences, each of a maximum length of 5,000 nucleotides or amino acids.
  • the multiple alignment procedure begins with the pairwise alignment of the two most similar sequences, producing a cluster of two aligned sequences. This cluster is then aligned to the next most related sequence or cluster of aligned sequences. Two clusters of sequences are aligned by a simple extension of the pairwise alignment of two individual sequences. The final alignment is achieved by a series of progressive, pairwise alignments.
  • the program is run by designating specific sequences and their amino acid or nucleotide coordinates for regions of sequence comparison and by designating the program parameters.
  • This algorithm involves first identifying high scoring sequence pairs (HSPs) by identifying short words of length W in the query sequence, which either match or satisfy some positive- valued threshold score T when aligned with a word of the same length in a database sequence. T is referred to as the neighborhood word score threshold (Altschul, et al., supra). These initial neighborhood word hits act as seeds for initiating searches to find longer HSPs containing them. The word hits are then extended in both directions along each sequence for as far as the cumulative alignment score can be increased.
  • HSPs high scoring sequence pairs
  • Extension of the word hits in each direction are halted when: the cumulative alignment score falls off by the quantity X from its maximum achieved value; the cumulative score goes to zero or below, due to the accumulation of one or more negative-scoring residue alignments; or the end of either sequence is reached.
  • the BLAST algorithm parameters W, T, and X determine the sensitivity and speed of the alignment.
  • the BLAST program uses as defaults a word length (W) of 11, the
  • the BLAST algorithm also performs a statistical analysis of the similarity between two sequences (see, e.g., Karlin and Altschul (1993) Proc. Nat'l Acad. Sci. USA 90:5873-5787).
  • One measure of similarity provided by the BLAST algorithm is the smallest sum probability (P(N)), which provides an indication of the probability by which a match between two nucleotide or amino acid sequences would occur by chance.
  • a nucleic acid is considered similar to a reference sequence if the smallest sum probability in a comparison of the test nucleic acid to the reference nucleic acid is less than about 0.1, more preferably less than about 0.01, and most preferably less than about 0.001.
  • Many proteins (and translated DNA sequences) contain regions where the amino acid composition is highly biased toward a small subset of the available residues.
  • membrane spanning domains and signal peptides typically contain long stretches where Leucine (L), Valine (V), Alanine (A), and Isoleucine (I) predominate.
  • Poly-Adenosine tracts (polyA) at the end of cDNAs appear in forward translations as poly-Lysine (poly-K) and poly-Phenylalanine (poly-F) when the reverse complement is translated. These regions are often referred to as "low complexity" regions. Such regions can cause database similarity search programs such as BLAST to find high-scoring sequence matches that do not imply true homology. The problem is exacerbated by the fact that most weight matrices (used to score the alignments generated by BLAST) give a match between any of a group of hydrophobic amino acids (L,V and I) that are commonly found in certain low complexity regions almost as high a score as for exact matches.
  • BLASTX.2 version 2.0 aSMP-WashU
  • filters designated “seg” and “xnu”
  • These filters parse the sequence for such regions, and create a new sequence in which the amino acids in the low complexity region have been replaced with the character "X”.
  • This is then used as the input sequence (sometimes referred to herein as "Query” and/or "Q") to the BLASTX program. While this regime helps to ensure that high-scoring matches represent true homology, there is a negative consequence in that the BLASTX program uses the query sequence that has been masked by the filters to draw alignments.
  • a polypeptide is typically substantially identical to a second polypeptide, for example, where the two peptides differ only by conservative substitutions.
  • Another indication that two nucleic acid sequences are substantially identical is that the two molecules hybridize to each other under stringent conditions, as described below. "Homologous" polynucleotide sequences, when compared, exhibit significant similarity (e.g., sequence identity at the nucleotide level).
  • hybridization refers to the process by which a polynucleotide strand anneals with a complementary strand through base pairing under defined hybridization conditions.
  • Specific hybridization is an indication that two nucleic acid sequences share a high degree of similarity and/or identity. Specific hybridization complexes form under permissive annealing conditions and remain hybridized after "washing.” Washing is particularly important in determining the stringency of the hybridization process, typically, with more stringent conditions allowing less non-specific binding (e.g., binding between polynucleotide sequences that demonstrate less sequence identity or similarity). Permissive conditions for annealing of nucleic acid sequences are routinely determinable by one of ordinary skill in the art and may be consistent among hybridization experiments, whereas wash conditions may be varied among experiments to achieve a desired stringency, and therefore, a particular hybridization specificity.
  • Stringent conditions when referring to homology or substantial similarity and/or identity in the hybridization context, will be stringent combined conditions of salt, temperature, organic solvents, and other parameters, typically those controlled in hybridization reactions.
  • Stringent temperature conditions will usually include temperatures in excess of about 30°C, more usually in excess of about 37°C, typically in excess of about 40°C, characteristically in excess of about 42°C, routinely in excess of about 45°C, usually in excess of about 47°C, preferably in excess of about 50°C, more typically in excess of about 55°C, characteristically in excess of about 60°C, preferably in excess of about 65°C, and more preferably in excess of about 70°C.
  • the term "about” includes, e.g., a particularly recited temperature (e.g., 50°C), and/or a temperature that is greater or lesser than that of the stated temperature by, e.g., one, two, three, four, or five degrees Celsius (e.g., 49°C or 51°C).
  • Stringent salt conditions will ordinarily be less than about 500 mM, usually less than about 450 mM, even more usually less than about 400 mM, more usually less than about 350 mM, even more usually less than about 300 mM, typically less than about 250 mM, even more typically less than about 200 mM, preferably less than about 100 mM, and more preferably less than about 80 mM, even down to less than about 20 mM.
  • the term "about” includes, e.g., a particularly recited molarity (e.g., 400 mM), and/or a molarity that is greater or lesser than that of the stated molarity by, e.g., three, five, seven, nine, eleven or fifteen millimolar (e.g., 389 mM or 415 mM). It is to be remembered that the combination of parameters is more important than the measure of any single parameter (see, e.g., Wetmur and Davidson (1968) J. Mol. Biol. 31:349-370).
  • a nucleic acid probe that binds to a target nucleic acid under stringent conditions to form a stable hybridization complex is said to be specific for said target nucleic acid.
  • hybridization under stringent conditions should give a signal of at least 2-fold over background, more preferably a signal of at least 3 to 5 -fold over background or more.
  • a hybridization probe is more than 11 nucleotides in length and is sufficiently identical (or complementary) to the sequence of the target nucleic acid (over the region determined by the sequence of the probe) to bind the target under stringent hybridization conditions to form a detectable stable hybridization complex.
  • hybridization complex refers to a complex formed between two nucleic acid molecules by virtue of the formation of hydrogen bonds between complementary bases.
  • a hybridization complex may be formed in solution (e.g., C 0 t or R 0 t analysis) or formed • between one nucleic acid sequence present in solution and another nucleic acid sequence immobilized on a solid support (such as, e.g., without limitation, paper, plastic, a membrane, a filter, a chip, a pin, glass, or any other appropriate substrate to which cells or their nucleic acids can be complexed with either covalently or non-covalently).
  • a solid support such as, e.g., without limitation, paper, plastic, a membrane, a filter, a chip, a pin, glass, or any other appropriate substrate to which cells or their nucleic acids can be complexed with either covalently or non-covalently).
  • LP protein from other mammalian species can be cloned and isolated by cross- species hybridization of closely related species (as described, e.g., herein). Similarity and/or sequence identity may be relatively low between distantly related species, and thus hybridization of relatively closely related species is advisable. Alternatively, preparation of an antibody preparation that exhibits less species specificity may be useful in an expression cloning approach.
  • stringency of hybridization is expressed, in part, with reference to the temperature under which the wash step is carried out. Such wash temperatures are typically selected to be about 5°C to 20°C lower than the thermal melting point (T m ) for the specific sequence at a defined ionic strength and pH.
  • the T m is the temperature (under defined ionic strength and pH) at which 50% of the target sequence hybridizes to a perfectly matched probe.
  • An equation for calculating T m and conditions for nucleic acid hybridization are well known (see, e.g., Sambrook, et al. (1990) Molecular Cloning: A Laboratory Manual (cur. ed.) Vol. 1-3, Cold Spring Harbor Laboratory, Cold Spring Harbor Press, NY, which is incorporated herein by reference and hereinafter referred to as "Sambrook, et al.”).
  • a non-limiting example of a high stringency condition of the invention comprises including a wash condition of 68°C in the presence of about 0.2 x SSC and about 0.1% SDS, for 1 hour.
  • temperatures of about 67°C, 63°C, 61°C, 59°C, 57°C, 53°C, 51°C, 49°C, 47°C, 43°C, or 41°C may be used.
  • SSC concentration may be varied from about 0.1 to 2X SSC, with SDS being present at about 0.1%.
  • blocking reagents are used to block non-specific hybridization.
  • blocking reagents include, for instance, sheared, and denatured salmon sperm DNA at about 100-200 Dg/ml.
  • Organic solvent such as, e.g., formamide at a concentration of about 35-50% v/v, may also be used under particular circumstances, such as for a RNA:DNA hybridization.
  • Hybridization particularly under high stringency conditions, may be suggestive of evolutionary similarity between the nucleotides. Such similarity is indicative of a similar functional and/or biological role for the nucleotide sequence and its correspondingly encoded polypeptide sequence.
  • a stringent hybridization condition comprises, e.g., an overnight incubation at 42°C in a solution comprising 50% formamide, 5x SSC (750 mM NaCl, 75 mM trisodium citrate), 50 mM sodium phosphate (pH 7.6), 5x Denhardt's solution, 10% dextran sulfate, and 20 pg/ml denatured, sheared salmon sperm DNA, followed by washing the filters in 0. lx SSC at about 65°C.
  • nucleic acid molecules that hybridize to an LP polynucleotide sequence at lower stringency hybridization conditions.
  • washes are performed following stringent hybridization at higher salt concentrations (e.g. 5X SSC).
  • blocking reagents include, e.g., Denhardt's reagent, BLOTTO, heparin, denatured salmon sperm DNA, and commercially available proprietary formulations.
  • the inclusion of specific blocking reagents may require modification of a hybridization conditions described herein.
  • a polynucleotide that hybridizes only to polyA+ sequences (such as any 3' terminal polyA+ tract of a cDNA of the invention), or to a complementary stretch of T (or U) residues, is not included, e.g., in the definition of an "LP polynucleotide” since such a polynucleotide would hybridize to any nucleic acid molecule containing a poly (A) stretch or the complement thereof (i.e., practically any double-stranded cDNA clone generated using oligo dT as a primer).
  • a stringent hybridization condition is one that employs, e.g.: low ionic strength and high temperature for washing (e.g., 15mM sodium chloride/1.5 mM sodium citrate/0.1% sodium dodecyl sulfate at 50°C); a denaturing agent (during hybridization) such as formamide (e.g., 50% (v/v) formamide with 0.1% bovine serum albumin/0.1% ficoll/0.1% polyvinylpyrrolidone/50 mM sodium phosphate buffer at pH 6.5 with 750 mM sodium chloride/75 mM sodium citrate at 42°C); or 50% formamide, 5X SSC (750 mM sodium chloride, 75 mM sodium citrate), 50 mM sodium phosphate (pH 6.8), 0.1% sodium pyrophosphate, 5X Denhardt's solution, sonicated salmon sperm DNA (50 ⁇ g/mL), 0.1% SDS, and 10% dextran s
  • formamide e
  • An LP polynucleotide sequence of the present invention can be composed of any polyribonucleotide or polydeoxribonucleotide, which may be unmodified RNA or DNA or modified RNA or DNA.
  • polynucleotides can be composed of single- and double-stranded DNA, DNA that is a mixture of single- and double-stranded regions, single- and double-stranded RNA, and RNA that is mixture of single- and double- stranded regions, hybrid molecules comprising DNA and RNA that may be single- stranded or, more typically, double-stranded or a mixture of single-and double-stranded regions.
  • polynucleotide can be composed of triple-stranded regions comprising RNA or DNA or both RNA and DNA.
  • a polynucleotide may also contain one or more modified bases or DNA or RNA backbones modified for stability or for other reasons.
  • Modified bases can include, e.g., for example, tritylated bases and unusual bases such as inosine.
  • tritylated bases can include, e.g., for example, tritylated bases and unusual bases such as inosine.
  • a variety of modifications can be made to DNA and RNA; thus, the term "polynucleotide” embraces chemically, enzymatically, or metabolically modified forms.
  • isolated nucleic acid molecule will usually contain homogeneous nucleic acid molecules, but, in some embodiments, it will contain nucleic acid molecules having minor sequence heterogeneity. Typically, this heterogeneity is found at the polymer ends or portions of the LP sequence that are not critical to a desired biological function or activity.
  • isolated does not refer to genomic or cDNA libraries, whole cell total or mRNA preparations, genomic DNA preparations (including those separated by electrophoresis and transferred onto blots), sheared whole cell genomic DNA preparations, or other compositions where the art demonstrates no distinguishing features of a LP polynucleotide sequence of the present invention.
  • a "recombinant" nucleic acid or polynucleotide sequence is defined either by its method of production or its structure. In reference to its method of production, e.g., a product made by a process, the process is use of recombinant nucleic acid techniques, e.g., involving human intervention in the nucleotide sequence, typically selection or production. Alternatively, it can be a nucleic acid made by generating a sequence comprising fusion of two fragments which are not naturally contiguous to each other, but is meant to exclude products of nature, e.g., naturally occurring mutants.
  • nucleic acids comprising sequence derived using any synthetic oligonucleotide process.
  • Such is often done to replace a codon with a redundant codon encoding the same or a conservative amino acid, while typically introducing or removing a sequence recognition site.
  • it is performed to join together nucleic acid segments of desired functions to generate a single engineered entity comprising a desired combination of functions not found in the commonly available natural forms.
  • Restriction enzyme recognition sites are often the target of such artificial manipulations, but other site-specific targets may be incorporated by design (such as, e.g., promoters, DNA replication sites, regulation sequences, control sequences, and other useful features).
  • LP protein shall encompass, when used in a protein context, a protein or polypeptide having an amino acid sequence shown in SEQ ID NO: Y or a significant fragment of such a protein or polypeptide, preferably a natural embodiment.
  • the invention also embraces polypeptides that exhibit similar structure to an LP polypeptide (e.g., one that interacts with an LP protein specific binding composition).
  • binding compositions e.g., antibodies
  • bind an LP protein with high affinity e.g., at least about 100 nM; usually, better than about 30 nM; preferably, better than about 10 nM; and more preferably, at better than about 3 nM.
  • polypeptide fragments of the invention include, e.g., a fragment comprising, or alternatively consisting of, from about amino acid number 1-20, 21-40, 41 -60, 61-80, 81- 100, 102-120, 121-140, 141-160, or 161 to the end of the coding region.
  • an LP polypeptide fragment can be about at least: 8, 10, 12, 14, 16, 18, 20, 22, 24, 26, 28, 30, 32, 34, 36, 38, 40, 42, 44, 46, 48, 50, 52, 54, 56, 58, 60, 62, 64, 66, 68, 70, 72, 74, 76, • 78, 80, 82, 84, 86, 88, 90, 92, 94, 96, 98, 100, 110, 120, 130, 140, or 150 contiguous amino acids in length.
  • polypeptide fragments include, e.g., the secreted protein as well as the mature form. Further preferred polypeptide fragments include, e.g., the secreted protein or the mature form having a continuous series of deleted residues from the amino or the carboxy terminus, or both.
  • a polypeptide having biological activity refers to biologically active fragments or polypeptides exhibiting activity similar, but not necessarily identical to, an activity of an LP polypeptide (or fragment thereof), including mature forms, as measured in a particular biological assay, with or without dose dependency.
  • dose dependency 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 ten-fold less activity, and most preferably, not more than about three-fold less activity relative to the polypeptide of the present invention.).
  • the biological activity of a fragment may include, e.g., an improved desired activity, or a decreased undesirable activity.
  • Polynucleotides encoding such polypeptide fragments are also encompassed by the invention. Any appropriate assay described herein or otherwise known in the art may routinely be applied to measure the ability of a polypeptide of the invention and a fragment, variant, derivative, and analog thereof to elicit related biological activity related to that of the polypeptide of the invention (either in vitro or in vivo). Other methods will be known to the skilled artisan and are within the scope of the invention.
  • the present invention also provides a polypeptide comprising, or alternatively, consisting of, a polypeptide sequence (or fragment thereof) of at least 12 contiguous amino acid residues of a mature polypeptide SEQ ID NO: Y and/or at least a 12 contiguous amino acid residue fragment of a mature polypeptide encoded by a cDNA contained in ATCC deposit as described herein.
  • Polynucleotides encoding a polypeptide comprising, or alternatively consisting of a polypeptide sequence of SEQ ID NO:Y and/or a polypeptide sequence encoded by a cDNA contained in ATCC deposit as described herein are also encompassed by the invention.
  • polypeptides encode such polypeptides are also encompassed by the invention.
  • a polynucleotide fragment of the invention encodes a polypeptide that demonstrates a functional activity.
  • demonstrating a "functional activity” is meant, a polypeptide having one or more known functional activities associated with a mature protein.
  • Such functional activities include, e.g., but are not limited to, biological activity; antigenicity (an ability to bind, or compete with a polypeptide of the invention for binding, to an antibody to a polypeptide of the invention); immunogenicity (an ability to stimulate the formation of a specific and/or selective antibody which binds to a polypeptide of the invention); an ability to form multimers with a polypeptide of the invention; and an ability to specifically and/or selectively bind a binding composition of a polypeptide of the invention.
  • a functional activity of a polypeptide of the invention can be assayed by various methods.
  • immunoassays known in the art can be used, including but not limited to, competitive and non-competitive assay systems using techniques such as radioimmunoassays, ELISA (enzyme linked immunosorbent assay), "sandwich” immunoassays, immunoradiometric assays, gel diffusion precipitation reactions, immunodiffusion assays, in situ immunoassays (e.g., using colloidal gold, enzyme or radioisotope labels), 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.)
  • agglutination assays e.g., gel agglutination assays, hemagglutination assays, complement fixation assays, immunofluorescence assays,
  • a primary antibody is detected, e.g., by detecting binding of a secondary antibody or reagent to the primary antibody.
  • the secondary antibody is labeled. Many means are' known in the art for detecting binding in an immunoassay and are within the scope of the present invention.
  • binding can be assayed, e.g., by any art known method (such as, e.g., reducing and non-reducing gel chromatography, protein affinity chromatography, and affinity blotting; see, e.g., generally, Phizicky, et al. (1995)
  • An LP polypeptide can be composed of amino acids joined to each other by peptide bonds or modified peptide bonds, i.e., peptide isosteres, and may contain amino acids other than the 20 gene-encoded amino acids.
  • the polypeptides may be modified by either natural processes, such as post-translational processing, or by chemical modification techniques that are well known in the art. Such modifications are well described in basic texts and in more detailed monographs, as well as in a voluminous research literature. Modifications can occur anywhere in a polypeptide, including the peptide backbone, the amino acid side-chains and the amino or carboxyl termini.
  • Modifications include, e.g., acetylation, acylation, ADP- ribosylation, amidation, covalent attachment of flavin, covalent attachment of a heme moiety, covalent attachment of a nucleotide or nucleotide derivative, covalent attachment of a lipid or lipid derivative, covalent attachment of phosphotidylinositol, cross-linking, cyclization, disulfide bond formation, demethylation, formation of covalent cross-links, formation of cysteine, formation of pyroglutamate, formylation, gamma-carboxylation, glycosylation, GPI anchor formation, hydroxylation, iodination, methylation, myristoylation, oxidation, pegylation, proteolytic processing, phosphorylation, prenylation, racemization, selenoylation, sulfation, transfer-RNA mediated addition of amino acids to proteins such as arginylation, and ubiquitin
  • Such moieties include, without limit, glutathione S-transferase (GST), maltose binding protein (MBP), thioredoxin (Trx), calmodulin binding peptide (CBP), 6-His, FLAG, c-myc, and hemagglutinin (HA).
  • GST, MBP, Trx, CBP, and 6- His moieties enable purification of their cognate fusion proteins on immobilized glutathione, maltose, phenylarsine oxide, calmodulin, and metal-chelate resins, respectively.
  • the FLAG, c-myc, and hemagglutinin (HA) moieties enable immunoaffinity purification of fusion proteins using commercially available monoclonal and polyclonal antibodies that specifically recognize these epitope tags.
  • "Solubility" of an LP protein or polypeptide is reflected by sedimentation measured in Svedberg units, which are a measure of the sedimentation velocity of a molecule under particular conditions. The determination of the sedimentation velocity was classically performed in an analytical ultracentrifuge, but is typically now performed in a standard ultracentrifuge (see, Freifelder (1982) Physical Biochemistry (2d ed.) W.H.
  • a soluble particle or polypeptide will typically be less than about 30S, more typically less than about 15S, usually less than about 10S, more usually less than about 6S, and, in particular embodiments, preferably less than about 4S, and more preferably less than about 3S. Solubility of a polypeptide or fragment depends upon the environment and the polypeptide.
  • the temperature at which the polypeptide is used ranges from about 4° C to about 65° C. Usually the temperature at use is greater than about 18° C and more usually greater than about 22° C. For diagnostic purposes, the temperature will usually be about room temperature or warmer, but less than the denaturation temperature of components in the assay. For therapeutic purposes, the temperature will usually be body temperature, typically about 37° C for humans, though under certain situations the temperature may be raised or lowered in situ or in vitro.
  • the size and structure of the polypeptide should generally be in a substantially stable state, and usually not in a denatured state.
  • the polypeptide may be associated with other polypeptides in a quaternary structure, e.g., to confer solubility, or associated with lipids or detergents in a manner which approximates natural lipid bilayer interactions.
  • the solvent will usually be a biologically compatible buffer, of a type used for preservation of biological activities, and will usually approximate a physiological solvent. Usually the solvent will have a neutral pH, typically between about 5 and 10, and preferably about 7.5.
  • a detergent will be added, typically a mild non- denaturing one, e.g., CHS (cholesteryl hemisuccinate) or CHAPS (3-[3- cholarnidopropyl)-dimethylammonio]-l -propane sulfonate), or a low enough concentration as to avoid significant disruption of structural or physiological properties of the protein.
  • CHS cholesteryl hemisuccinate
  • CHAPS 3-[3- cholarnidopropyl)-dimethylammonio]-l -propane sulfonate
  • the present invention encompasses "mature" forms of a polypeptide comprising a polypeptide sequence listed in a Table herein, a polypeptide sequence of SEQ ID NO: Y, or a polypeptide sequence encoded by a cDNA in a deposited clone.
  • Polynucleotides encoding a mature form are also encompassed by the invention.
  • cleavage of a signal sequence of a secreted protein is not uniform, resulting in more than one secreted species for a given protein (e.g., a cleavage variant).
  • cleavage variant LP polypeptides, and the polynucleotides encoding them are also encompassed by the present invention.
  • the signal sequence identified by the above analysis may not necessarily predict a naturally occurring signal sequence.
  • a naturally occurring signal sequence may be further upstream from a predicted signal sequence.
  • a predicted signal sequence will be capable of directing the secreted protein to the ER.
  • the present invention encompasses a mature LP polypeptide or protein produced by expression of a polynucleotide sequence listed in a Table herein, an LP polynucleotide sequence of SEQ ID NO: X, or an LP polynucleotide sequence contained in a cDNA of a deposited clone, in a mammalian cell (e.g., a COS cell, as described).
  • LP polypeptides (fragments thereof), and the polynucleotides encoding them are also encompassed by the present invention.
  • the present invention encompasses variants of an LP polynucleotide sequence disclosed in SEQ ID NO: X, the complementary strand thereto, and/or a cDNA sequence contained in a deposited clone.
  • the present invention also encompasses variants of a polypeptide sequence disclosed in SEQ ID NO: Y and/or encoded by a deposited clone.
  • variant refers to a polynucleotide or polypeptide differing from an LP polynucleotide sequence or an LP polypeptide of the present invention, but retaining essential properties thereof.
  • variants are closely similar overall in structural and/or sequence identity, and, in many regions, identical to an LP polynucleotide or polypeptide of the present invention.
  • the present invention encompasses nucleic acid molecules that comprise, or alternatively consist of, a polynucleotide sequence that is at least: 80%, 85%, 90%, 95%, 96%, 97%, 98%, or 99% identical to, e.g., a polynucleotide coding sequence of SEQ ID NO: X (or a strand complementary thereto); a nucleotide sequence contained in a deposited cDNA clone (or a complementary strand thereto); a nucleotide sequence encoding a polypeptide of SEQ ID NO: Y; a nucleotide sequence encoding a polypeptide encoded by the cDNA contained in a deposited clone; and/or polynucleotide fragments of any of these nucleic acid molecules (e.
  • Polynucleotides that stably hybridize to a polynucleotide fragment (as defined herein) under stringent hybridization conditions or lower stringency conditions, are also encompassed by the invention, as are polypeptides (or fragments thereof) encoded by these polynucleotides.
  • the present invention is also directed to polypeptides that comprise, or alternatively consist of, an amino acid sequence that is at least: 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99% identical to, e.g., a polypeptide sequence of SEQ ID NO: Y (or fragments thereof); a polypeptide sequence encoded by a cDNA contained in a deposited clone, and/or a polypeptide fragment of any of these polypeptides (e.g., those fragments as defined herein).
  • a polynucleotide sequence having at least some "percentage identity,” (e.g., 95%) to another polynucleotide sequence means that the sequence being compared (e.g., the test sequence) may vary from another sequence (e.g. the referent sequence) by a certain number of nucleotide differences (e.g., a test sequence with 95% sequence identity to a reference sequence can have up to five point mutations per each 100 contiguous nucleotides of the referent sequence).
  • test sequence for a test sequence to exhibit at least 95% identity to a referent sequence, up to 5% of the nucleotides in the referent may differ, e.g., be deleted or substituted with another nucleotide, or a number of nucleotides (up to 5% of the total number of nucleotides in the reference sequence) may be inserted into the reference sequence.
  • the test sequence may be: an entire polynucleotide sequence, e.g., as shown in Tables 1-18, the ORF (open reading frame), or any fragment, segment, or portion thereof (as described herein).
  • determining if a particular nucleic acid molecule or polynucleotide sequence exhibits at least about: 80%, 85%, 90%, 95%, 96%, 97%, 98% or 99% identity to an LP polynucleotide sequence can be accomplished using known computer programs.
  • one sequence acts as a reference sequence, to which test sequences are compared.
  • test and reference sequences are input into a computer, subsequent coordinates are designated, if necessary, and sequence algorithm program parameters are designated.
  • the sequence comparison algorithm then calculates the percentage sequence identity for a test sequence(s) relative to the reference sequence, based on the parameters of a designated program.
  • Optimal alignment of sequences for comparison can be conducted, e.g., by the local homology algorithm of Smith and Waterman (1981) Adv. Appl. Math. 2:482, by the homology alignment algorithm of Needlman and Wunsch (1970) J. Mol. Biol. 48:443, by the search for similarity method of Pearson and Lipman (1988) Proc. Nat'l Acad. Sci. USA 85:2444, by computerized implementations of these algorithms (GAP, BESTFIT, FASTA, and TFASTA in the Wisconsin Genetics Software Package, Genetics Computer Group, 575 Science Dr., Madison, WI), or by visual inspection (see generally, Ausubel, et al. supra).
  • a typical method for determining , a best overall match (also referred to as a global sequence alignment) between a test and a referent sequence can be determined using , e.g., the FASTDB computer program based on the algorithm of Brutlag, et al. (1990) Comp. App. Biosci. 6: 237-245.
  • the test and referent sequences are, e.g., both DNA sequences.
  • An RNA sequence can be compared by converting U's to T's. The result of a global sequence alignment is given in terms of a percentage identity.
  • the percentage identity is corrected by calculating the number of bases of the test sequence that are 5 ' and 3' of the subject sequence, which are not matched/aligned, as a percentage of the total bases of the test sequence. Whether a nucleotide is matched/aligned is determined by results of the FASTDB sequence alignment. This percentage is then subtracted from the percentage identity, calculated by the above FASTDB program using the specified parameters, to arrive at a final percentage identity score. The corrected score is what is used for the purposes of sequence identity for the present invention.
  • bases outside the 5' and 3' bases of the subject sequence, as displayed by the FASTDB alignment, which are not matched/aligned with the test sequence are calculated for the purposes of manually adjusting the percent identity score.
  • a 90 base referent sequence is aligned to a 100 base test sequence to determine percentage identity.
  • the deletions occur at the 5' end of the referent sequence and therefore, the FASTDB alignment does not show a matched/alignment of the first 10 bases at the 5' end.
  • the 10 unpaired bases represent 10% of the sequence (number of bases at the 5' and 3' ends not matched/total number of bases in the test sequence) so 10% is subtracted from the percentage identity score calculated by the FASTDB program.
  • a 90 base referent sequence is compared with a 100 base test sequence. This time the deletions are internal deletions so that there are no bases on the 5' or 3' of the subject sequence, which are not matched/aligned with the test. In this case, the percentage identity calculated by FASTDB is not manually corrected. Again, only bases 5' and 3' of the subject sequence that are not matched/aligned with the test sequence are manually corrected for. No other manual corrections are to made for the purposes of the present invention. Especially preferred are polynucleotide variants containing alterations, which produce silent substitutions, additions, or deletions, but do not alter the properties or activities of the encoded polypeptide.
  • Nucleotide variants produced by silent substitutions due to the degeneracy of the genetic code are preferred. Polynucleotide variants can be produced for a variety of reasons, e.g., to optimize codon expression for a particular host (change codons in the human mRNA to those preferred by a bacterial host such as E. coli).
  • a further indication that two nucleic acid sequences of polypeptides are substantially identical is that the polypeptide encoded by the first nucleic acid is immunologically cross reactive with the polypeptide encoded by the second nucleic acid, as described herein.
  • a polypeptide is typically substantially identical to a second polypeptide, for example, where the two peptides differ only by conservative substitutions.
  • a polypeptide exhibiting or having at least about, e.g., 95% "sequence identity" to another amino acid sequence may include, e.g., up to five amino acid alterations per each 100 amino acid stretch of the test amino acid sequence.
  • a first amino acid sequence that is at least 95% identical to a second amino acid sequence can have up to 5% of its total number of amino acid residues different from the second sequence, e.g., by insertion, deletion, or substitution of an amino acid residue.
  • a preferred method for determining the best overall match uses the FASTDB algorithm of Brutlag, et al. (1990) Comp. App. Biosci. 6:237-245.
  • the result of such a global sequence alignment is given as a percentage of sequence identity, e.g., with 100% representing complete sequence identity.
  • the percent identity is corrected by calculating the number of residues of the test 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 test 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 percentage- identity score. This final percentage identity score is what is used for the purposes of the present invention.
  • variants encompassed by the present invention may contain alterations in the coding regions, non-coding regions, or both. Moreover, variants in which 1-2, 1-5, or 5-10 amino acids are substituted, deleted, or added in any combination are also preferred. Naturally occurring variants encompassed herein are "allelic variants," which refer to one of several alternate forms of a gene occupying a given locus on a chromosome of an organism.
  • Allelic variants can vary at either the polynucleotide and/or polypeptide level and both types of variants are encompassed by the present invention.
  • non-naturally occurring variants may be produced by mutagenesis techniques or by direct synthesis using known methods of protein engineering and recombinant DNA technology.
  • Such variants may be generated to improve or alter the characteristics of an LP polypeptide (or fragment thereof).
  • one or more amino acids can be deleted from the N-terminus or C- terminus of a secreted polypeptide of the invention (or fragment thereof) without a substantial loss of biological function.
  • antigenicity and/or immunogenicity can be retained (e.g., the ability of a deletion variant to induce and/or to bind antibodies that recognize a mature form of a polypeptide) when less than the majority of the residues of the secreted form are removed from the N-terminus or C-terminus.
  • a polypeptide lacking N- or C-terminal residues of a protein retains such activities can readily be determined by routine methods such as those described herein or known in the art.
  • the invention also encompasses, e.g., polypeptide variants that show biological activity such as, e.g., immunogenicity, or antigenicity.
  • Such variants include, e.g., deletions, insertions, inversions, repeats, and substitutions selected so as have little effect on activity using general rules known in the art.
  • teachings on making phenotypically silent amino acid substitutions are provided, e.g., by Bowie, et al. (1990) Science 247: 1306-1310
  • One technique compares amino acid sequences in different species to identify the positions of conserved amino acid residues since changes in an amino acid at these positions are more likely to affect a protein function.
  • the positions of residues where substitutions are more frequent generally indicates that amino acid residues at these positions are less critical for a protein function.
  • positions tolerating amino acid substitutions typically may be modified while still maintaining a biological activity of a protein.
  • a second technique uses genetic engineering to introduce amino acid changes at specific positions of a polypeptide to identify regions critical for a protein function. For example, site directed mutagenesis or alanme-scanning mutagenesis (the introduction of single alanine mutations at every residue in the molecule) can be used. (Cunningham and Wells (1989) Science 244: 1081-1085) A resulting mutein can subsequently be tested for a biological activity.
  • variants of the present invention include, e.g., but are restricted to, e.g., (i) substitutions with 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) substitution with one or more of amino acid residues having a substituent group, or (iii) fusion of the mature polypeptide with another compound, such as a compound to increase the stability and/or solubility of the polypeptide (e.g., polyethylene glycol), or (iv) fusion of the polypeptide with additional amino acids, such as, e.g., an IgG Fc fusion region peptide, or leader or secretory sequence, or a sequence facilitating purification.
  • additional amino acids such as, e.g., an IgG Fc fusion region peptide, or leader or secretory sequence, or a sequence facilitating purification.
  • polypeptide variants containing amino acid substitutions of charged amino acids with other charged or neutral amino acids may produce polypeptides with improved characteristics e.g., such as less aggregation. Aggregation of pharmaceutical formulations both reduces activity and increases clearance due to the aggregate's immunogenic activity (Pinckard, et al. (1967) Clin. Exp. Immunol. 2:331-340; Robbins, et al. (1987) Diabetes 36:838-845; Cleland, et al. (1993) Crit. Rev.
  • a further embodiment of the invention encompasses a protein that comprises an amino acid sequence of the present invention that contains at least one amino acid substitution, but not more than 50 amino acid substitutions, even more preferably, not more than 40 amino acid substitutions, still more preferably, not more than 30 amino acid substitutions, and still even more preferably, not more than 20 amino acid substitutions, nor more than 15 amino acid substitutions.
  • a peptide or polypeptide it is highly preferable for a peptide or polypeptide to have an amino acid sequence that comprises an amino acid sequence of the present invention, which contains at least: one, but not more than: 10, 9, 8, 7, 6, 5, 4, 3, 2, or 1 amino acid substitutions.
  • the number of additions, substitutions, and/or deletions in an polypeptide sequence of the present invention or fragments thereof is at least: 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 10-50, or 50-150; wherein conservative amino acid substitutions are more preferable than non-conservative substitutions.
  • LP Polynucleotide and LP Polypeptide Fragments The present invention is also directed to fragments of an LP polynucleotide.
  • LP polynucleotide "fragment” encompasses a short polynucleotide of a nucleic acid molecule, which is a portion of a sequence contained in a deposited clone, or encoding a polypeptide encoded by a cDNA in a deposited clone; or a portion of a polynucleotide sequence of SEQ ID NO: X or a complementary strand thereto, or a portion of a polynucleotide sequence encoding a polypeptide of SEQ ID NO: Y (or fragment thereof).
  • Polynucleotide fragments of the invention encompass a polynucleotide sequence that is preferably at least about 15 nucleotides, more preferably at least about: 20, 21, 22, 24, 26, or 29 nucleotides, favorably at least about: 30, 32, 34, 36, 38, or 39 nucleotides, and even more preferably, at least about: 40, 42, 44, 46, 48, or 49 nucleotides, desirably at least about: 50, 52, 54, 56, 58, or 59 nucleotides, particularly at least about 75 nucleotides, or at least about 150 nucleotides in length.
  • at least about includes, e.g., a specifically recited value (e.g., 20nt), and a value that is larger or smaller by one or more nucleotides (e.g., 5, 4, 3, 2, or 1), at either terminus or at both termini.
  • a polynucleotide fragment has use that includes without limit; e.g., diagnostic probes and primers as discussed herein.
  • the present invention also encompasses a polynucleotide that stably hybridizes to a polynucleotide fragment described herein under either stringent or lowered stringency hybridization conditions. Additionally incorporated are polypeptides encoded by a polynucleotide fragment or a hybridized polynucleotide stably bound to a polynucleotide fragment of the invention. Additionally encompassed by the invention is a polynucleotide encoding a polypeptide, which are specifically or selectively bound by an antibody directed to/or generated against a mature polypeptide of the invention (or fragment thereof), e.g., a mature polypeptide of SEQ ID NO: Y.
  • a "polypeptide fragment or segment” encompasses an amino acid sequence that is a portion of SEQ ID NO: Y or a polypeptide segment encoded by a cDNA contained in a deposited clone.
  • Protein and/or polypeptide fragments or segments may be "free-standing,” or they may comprise part of a larger polypeptide or protein, of which the fragment or segment forms a portion or region, e.g., a single continuous region of SEQ ID NO: Y connected in a fusion protein.
  • a polypeptide segment of the invention can have a length of contiguous amino acids of a polypeptide of the invention (or fragment thereof) that is at least about: 7, 8, 9, 10, 12, 14, 16, 18, 20, 22, 24, 26, 28, 30, 32, 34, 36, 38, 40, 42, 44, 46, 48, 50, 52, 54, 56, 58, 60, 62, 64, 66, 68, 70, 72, 74, 76, 78, 80, 82, 84, 86, 88, 90, 92, 94, 96, 98, 100, 110, 120, 130, 140, or 150 contiguous amino acids in length.
  • a plurality will be at least two, more usually at least three, and preferably at least: four, five, six, seven, eight, nine, or more. While minimum lengths of a segment are provided, maximum lengths of various sizes are also encompassed for any specific plurality of segments, e.g., a plurality of three segments in toto could have one segment of length 7 contiguous amino acids, and two additional non- overlapping segments, each of which has a length of 12.
  • Preferred polypeptide fragments include, e.g., the secreted protein, as well as the mature form.
  • polypeptide fragments include, e.g., the secreted protein or the mature form having a continuous series of deleted residues from the amino or the carboxy terminus, or both.
  • a number of amino acids, ranging from 1-30 can be deleted from the amino terminus of either the secreted polypeptide or the mature form.
  • a number of amino acids, ranging from 1-30 can be deleted from the carboxy terminus of the secreted protein or mature form.
  • any combination of the above amino and carboxy terminus deletions are preferred.
  • polynucleotides encoding these polypeptide fragments are also preferred.
  • polypeptide fragments or segments that characterize structural or functional domains, such as, fragments, or combinations thereof, that comprise e.g., alpha-helix, and alpha-helix forming regions, beta-sheet, and beta-sheet-forming regions, turn, and turn-forming regions, coil, and coil-forming regions, hydrophilic regions, hydrophobic regions, alpha amphipathic regions, beta amphipathic regions, flexible regions, loop regions, hairpin domains, beta-alpa-beta motifs, helix bundles, alpha/beta barrels, up and down beta barrels, jelly roll or swiss roll motifs, transmembrane domains, surface-forming regions, substrate binding regions, transmembrane regions, linkers, immunogenic regions, epitopic regions, and high antigenic index regions.
  • Polypeptide fragments of SEQ ID NO: Y falling within conserved domains are specifically encompassed by the present invention. Moreover, polynucleotides encoding these domains are also encompassed. Other preferred polypeptide segments are biologically active fragments.
  • Bioly active fragments are those exhibiting activity similar, but not necessarily identical, to an activity of an LP polypeptide (or fragment thereof).
  • the biological activity of the fragments may include, e.g., an improved desired activity, or a decreased undesirable activity.
  • Polynucleotides encoding these polypeptide fragments are also encompassed by the invention.
  • the polynucleotide fragments of the invention encode a polypeptide that demonstrates a functional activity.
  • the phrase "functional activity" encompasses a polypeptide segment that can accomplish one or more known functional activities associated with a full-length (complete) polypeptide of invention protein.
  • Such functional activities include, e.g., without limitation, biological activity, antigenicity [ability to bind (or compete with a polypeptide of the invention for binding) to an antibody to a polypeptide of the invention], immunogenicity (ability to generate antibody that binds to a polypeptide of the invention), ability to form multimers with a polypeptide of the invention, and the ability to bind to a receptor or ligand of a polypeptide of the invention.
  • the functional activity of a polypeptide of the invention can be assayed by various methods.
  • immunoassays known in the art can be used, including, e.g., without limitation, competitive and non-competitive assay systems using techniques such as radioimmunoassays, ELISA (enzyme linked immunosorbent assay), "sandwich” immunoassays, immunoradiometric assays, gel diffusion precipitation reactions, immunodiffusion assays, in situ immunoassays (using colloidal gold, enzyme or radioisotope labels, for example), western blots, precipitation reactions, agglutination assays (e.g., gel agglutination assays, hemagglutination assays, complement fixation assays, immunofluorescence assays, protein A assays, and immunoelectrophoresis assays, etc.)
  • agglutination assays e.g., gel agglutination assays, hemagglutination assays, complement fixation assays, immunofluorescence as
  • the primary antibody is detected by detecting binding of a secondary antibody or reagent to the primary antibody.
  • the secondary antibody is labeled.
  • Many means are known in the art for detecting binding in an immunoassay and are within the scope of the present invention.
  • binding can be assayed, e.g., by using reducing and non-reducing gel chromatography, protein affinity chromatography, and affinity blotting (see generally, Phizicky, et al. (1995) Microbial. Rev. 59:94-123).
  • physiological correlates of binding of a polypeptide of the invention to its substrates can be assayed with common techniques.
  • assays described herein see, e.g., the "Examples” section of the application), or otherwise known in the art, can routinely be applied to measure the ability of a polypeptide of the invention (its fragments, variants derivatives and analogs thereof) to elicit a related biological activity (either in vitro or in vivo).
  • the present invention encompasses a polypeptide comprising, or alternatively consisting of, an epitope of SEQ ID NO: Y, or an epitope of a polypeptide sequence encoded by a polynucleotide contained in cDNA in a clone deposited as described herein; or encoded by a polynucleotide that stably hybridizes to form a hybridization complex, under stringent hybridization conditions (or lower stringency hybridization conditions) as defined herein, to a complement of a sequence of SEQ ID NO: X or to a sequence contained in deposited cDNA clone as described herein.
  • the present invention further encompasses a polynucleotide sequence encoding an epitope of a polypeptide sequence of the invention (such as, e.g., the sequence disclosed in SEQ ID NO: X), a polynucleotide sequence of the complementary strand of a polynucleotide sequence encoding an epitope of the invention, and a polynucleotide sequence that stably hybridizes to a complementary strand under stringent hybridization conditions or lower stringency hybridization conditions as defined herein.
  • epitope refers to a portion of a polypeptide having antigenic or immunogenic activity in an animal, preferably a mammal, and most preferably in a human.
  • the present invention encompasses a polypeptide comprising an epitope, as well as the polynucleotide encoding this polypeptide.
  • An "immunogenic epitope,” as used herein, is defined as a portion of a protein or a linearized polypeptide (or fragment thereof) that elicits an antibody response in an animal, as determined by any art known method (e.g., by the methods for generating antibodies described herein or otherwise known, see, e.g., Geysen, et al. (1983) Proc. Natl. Acad. Sci. USA 308 1 :3998-4002).
  • an antibody is specific for epitope "A”
  • the presence of a polypeptide comprising the epitope A, or the presence of free unlabeled A, in a reaction containing free labeled A and the antibody will reduce the amount of labeled A that binds to the antibody.
  • the specified antibodies bind to a particular protein or polypeptide sequence and do not significantly bind other proteins or other polypeptide sequences that are present in the sample. Specific binding to an antibody under such conditions may require an antibody that is selected for its specificity and/or selectivity for a particular protein.
  • antibodies raised to the protein immunogen with an amino acid sequence depicted in SEQ ID NO: Y can be selected to obtain antibodies specifically immunoreactive with LP proteins or LP polypeptides and not with other proteins or polypeptides. These antibodies will also recognize proteins or polypeptide sequences that have an above average degree of similarity or identity to an LP protein or LP polypeptide sequence. Fragments that function as epitopes can be produced by any conventional means such as, e.g., (1985) Houghten, Proc. Natl. Acad. Sci. USA 82:5131- 5135, further described in U.S. Patent No. 4,631,211.
  • an antigenic or immunogenic epitope preferably contains a polypeptide sequence of at least four, at least five, at least six, at least seven, more preferably at least eight, at least nine, 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 40, at least 50, and, favorably, between about 15 to about 30 contiguous amino acids of a mature polypeptide of SEQ ID NO: Y or encoded by a polynucleotide contained in a cDNA in a clone deposited as described herein.
  • Preferred polypeptide fragments of contiguous amino acid residues of SEQ ID NO: Y comprising immunogenic or antigenic epitopes are at least 10, 15, 20, 25, 30, 35, 40, 45, 50, 55, 60, 65, 70, 75, 80, 85, 90, 95, or 100 contiguous amino acid residues in length.
  • Additional non-exclusive preferred antigenic epitopes include, e.g., the antigenic epitopes disclosed herein, as well as portions thereof.
  • Antigenic epitopes are useful, e.g., to generate antibodies, including monoclonal antibodies that specifically bind the epitope.
  • Preferred antigenic epitopes include, e.g., the antigenic epitopes disclosed herein, as well as any plurality thereof, e.g., at least: two, three, four, five or more of these antigenic epitopes in any combination or structural arrangement.
  • Antigenic epitopes can be used as the target molecules in immunoassays (see, e.g., Wilson, et al. (1984) Cell 37:767-778; Sutcliffe, et al. (1983) Science 219:660-666).
  • immunogenic epitopes can be used, e.g., to induce antibodies according to any known art method (see, for instance, Sutcliffe, et al. supra; Wilson, et al.
  • immunogenic epitopes include, e.g., an immunogenic epitope disclosed herein, as well as a plurality or any combination thereof, e.g., of at least two, three, four, five or more of these immunogenic epitopes including, e.g., repeats of a particular epitope.
  • a polypeptide comprising a plurality of epitopes may be used to elicit an antibody response with a carrier protein, such as, e.g., an albumin, to an animal system (such as, e.g., a rabbit or a mouse), or, if a polypeptide is of sufficient length (e.g., at least about 25 amino acids), the polypeptide may be presented without a carrier.
  • a carrier protein such as, e.g., an albumin
  • an animal system such as, e.g., a rabbit or a mouse
  • immunogenic epitopes comprising as few as 8 to 10 amino acids have also been shown to be sufficient to generate antibodies and to be useful since they are capable of binding to, e.g., linear epitopes in a denatured polypeptide such as in Western blotting.
  • Polypeptides or proteins bearing an epitope of the present invention may be used to generate antibodies according to known methods including, e.g., without limitation, in vivo immunization, in vitro immunization, and phage display methods (see, e.g., Sutcliffe, et al. supra; Wilson, et al. supra, and Bittle, et al. (1985) J. Gen. Virol. 66:2347-2354.
  • animals may be immunized with free peptide; however, anti-peptide antibody titer may be boosted by coupling the peptide to a macromolecular carrier, such as keyhole limpet hemacyanin (KLH) or tetanus toxoid.
  • KLH keyhole limpet hemacyanin
  • polypeptides containing cysteine residues may be coupled to a carrier using a linker such as maleimidobenzoyl- N-hydroxysuccinimide ester (MBS), while other polypeptides may be coupled to carriers using a more general linking agent such as glutaraldehyde.
  • Animals such as rabbits, rats and mice are immunized with either free or carrier- coupled peptides, for instance, by intraperitoneal and/or intradermal injection of emulsions containing about 100 ⁇ g of peptide or carrier protein and Freund's adjuvant or any other adjuvant known for stimulating an immune response.
  • booster injections may be needed, for instance, at intervals of about two weeks, to provide a useful titer of anti-peptide antibody that can be detected, e.g., by ELISA assay using free peptide adsorbed to a solid surface.
  • binding composition refers to molecules that bind with specificity and/or selectivity to an LP of the invention or fragment thereof (such as, e.g., in an antibody-antigen interaction).
  • other compositions e.g., antibodies, oligonucleotides, proteins (e.g., receptors), peptides, or small molecules
  • the LP may be used to screen for binding compositions that specifically and/or selectively bind an LP of the invention or fragment thereof (e.g., a binding composition can be a molecule, or part of one, that selectively and/or stoichiometrically binds, whether covalently or not, to one or more specific sites of an LP (or fragment thereof) such as, e.g., in an antigen-antibody interaction, a hormone-receptor interaction, a substrate- enzyme interaction, etc.). At least one and up to a plurality of test binding compositions can be screened for specific and or selective binding with the LP.
  • a binding composition can be a molecule, or part of one, that selectively and/or stoichiometrically binds, whether covalently or not, to one or more specific sites of an LP (or fragment thereof) such as, e.g., in an antigen-antibody interaction, a hormone-receptor interaction, a substrate- enzyme interaction, etc.).
  • a binding composition thus identified is closely related to a natural ligand of an LP (such as, e.g., a ligand or fragment thereof, a natural substrate, a structural or functional mimetic, or a natural binding partner; see, e.g., Coligan, et al. (1991) Current Protocols in rmmunology l(2):_Chapter 5.)
  • a binding composition can be closely related to a natural ligand to which a secreted LP (or fragment thereof) binds such as, e.g., to a receptor or to at least a fragment of the receptor (e.g., the ligand binding site).
  • a binding composition can be rationally designed using known techniques.
  • screening for binding compositions involves using an appropriate cell that expresses an LP, or fragment thereof (either as a secreted protein or complexed with a cell membrane for presentation).
  • Preferred cells include, e.g., mammalian, yeast, insect (e.g., Drosophila), or bacterial cells (e.g., E. coli).
  • an isolated LP (or fragment thereof) is immobilized on a solid phase (such as, e.g., a stable surface such as, e.g., a membrane, plastic, nylon, a pin, glass, etc.), by covalent or non-covalent attachments, to permit presentation of the LP to a test binding composition for a time sufficient to permit selective and/or specific binding to occur.
  • a test binding composition is contacted to a presented LP (or fragment thereof) and the interaction is subsequently analyzed (e.g., to determine the presence or absence of: binding, stimulation, inhibition, agonist or antagonist activity, etc., either of the LP or the test composition).
  • inhibitors of a binding interaction can be identified, e.g., screening for peptide or small molecule inhibitors or agonists of a binding interaction between the LP and a binding composition.
  • binding agentiLP complex refers to a complex of a binding agent and a LP (or fragment thereof) which is formed by specific and/or selective binding of the binding agent to the respective LP (or fragment thereof).
  • Specific and/or selective binding of the binding agent means that the binding agent has a specific and/or selective binding site that recognizes a site on the LP protein (or fragment thereof).
  • antibodies raised against a LP protein (or fragment thereof) that recognize an epitope on the LP protein (or fragment thereof) are capable of forming a binding agentLP complex by specific and/or selective binding.
  • the formation of a binding agentLP complex allows the measurement of LP protein (or fragment thereof) in a mixture of other proteins and/or biologies.
  • antibody:LP complex refers to an embodiment in which the binding agent, e.g., is an antibody.
  • the antibody may be monoclonal, polyclonal, or a binding fragment of an antibody (including, without limit, e.g., Fv, Fab, or F(ab)2 fragments; diabodies; linear antibodies (Zapata, et al, (1995) Protein Engin. 8(10): 1057-62); single- chain antibody molecules; and multispecific antibodies formed from antibody fragments).
  • the antibody is a polyclonal antibody.
  • K a is defined as the molar concentration of antibody:LP complex divided by the molar concentrations of free antigen and free antibody under equilibrium conditions.
  • K a association constant
  • the K a determined for a preparation of monoclonal antibodies, which are monospecific for a particular LP epitope, represents a specific measure of affinity.
  • High-affinity antibody preparations with K a ranging from about 10 9 to 10 12 L/mole are preferred for use in immunoassays in which the antibody:LP complex must withstand rigorous manipulations.
  • Low-affinity antibody preparations with K a ranging from about 10 6 to 10 7 L/mole are preferred for use in immunopurification and similar procedures which ultimately require dissociation of LP, preferably in active form, from the antibody (Catty, (1988) Antibodies, Volume I: A Practical Approach. IRL Press, Washington DC; Liddell, J.E. and A. Cryer (1991) A Practical Guide to Monoclonal Antibodies. John Wiley & Sons, New York NY).
  • polyclonal antibody preparations may be further evaluated to determine the quality and suitability of such preparations for certain downstream applications.
  • a polyclonal antibody preparation containing at least 1-2 mg specific antibody/ml preferably, 5-10 mg specific antibody/ml
  • Procedures for evaluating antibody selectivity, specificity, titer, and avidity, and guidelines for antibody quality and usage in various applications, are generally available (see, e.g., Catty, supra, and Coligan et al. supra).
  • a recombinant vector comprising a polynucleotide sequence comprising sequence encoding an LP binding composition can be administered using any appropriate known art method (e.g., by polynucleotide delivery) to modulate, treat, inhibit, ameliorate, or prevent a disease, syndrome, condition, or disorder associated with aberrant expression and/or activity of a polypeptide (or fragment thereof) of the invention.
  • the vector comprises polynucleotide sequence comprising sequence encoding an LP antibody, wherein the polynucleotide sequence is part of an expression vector that expresses the antibody, (or fragments, or chimeric proteins, or heavy or light chains thereof), in a suitable host.
  • polynucleotide sequences have promoters, operably linked to the antibody coding region, that can be either inducible or constitutive, and, optionally, e.g., tissue-specific, cell-specific or developmentally specific .
  • nucleic acid molecules comprising sequence in which the antibody coding sequences and any other desired sequences are flanked by regions that promote homologous recombination at a desired site, thus providing for targeted delivery and expression of the antibody (see, e.g., Koller and Smithies, Proc. Natl. Acad. Sci. USA 86:8932-8935 (1989); Zijlstra, et al, (1989) Nature 342:435-438.
  • the expressed antibody molecule is a single chain antibody or alternatively, the heterologous sequence includes, e.g., sequence encoding both heavy and light chains, or fragments thereof, of an antibody.
  • nucleic acid molecule or nucleic acid-carrying vectors Delivery of such sequences into a cell can either be direct, (in which case a cell is directly exposed to the nucleic acid molecule or nucleic acid-carrying vectors), or indirect, (in which a case a cell is first transformed in vitro, then transplanted into a mammalian host).
  • the two approaches are known, respectively, as in vivo or ex vivo polynucleotide delivery.
  • III. Nucleic Acids Primate LP proteins described herein are exemplary of larger classes of structurally and functionally related proteins. The preferred embodiments, as disclosed, are useful in standard procedures to isolate similar genetic sequences from different individuals or other species (e.g., warm blooded animals, such as birds and mammals).
  • Cross hybridization will allow isolation of related genes encoding proteins with substantially similar identity from individuals, strains, or species.
  • a number of different approaches are available to successfully isolate a suitable nucleic acid clone based upon the information provided herein.
  • Southern blot hybridization studies can qualitatively determine the presence of similar genetic sequences in human, monkey, rat, mouse, dog, cow, and rabbit genomes under specific hybridization conditions. Complementary sequences are useful as probes or primers. Based upon identification of the likely amino terminus, other peptides should be particularly useful, e.g., coupled with anchored vector or poly-A complementary PCR techniques or with complementary DNA of other peptides.
  • nucleic acid manipulation of genes encoding LP proteins such as subcloning nucleic acid sequences encoding polypeptides into expression vectors, labeling probes, DNA hybridization, and the like are described generally in Sambrook, et al.
  • Various methods of isolating DNA sequences encoding LP proteins can be utilized. For example, DNA is isolated from a genomic or cDNA library using labeled oligonucleotide probes having sequences identical or complementary to the sequences disclosed herein. Full-length probes may be used, or oligonucleotide probes may be generated by comparison of the sequences disclosed.
  • probes can be used directly in hybridization assays to isolate DNA encoding LP proteins, or probes can be designed for use in amplification techniques such as PCR, for the isolation of DNA encoding LP proteins.
  • amplification techniques such as PCR
  • cDNA is prepared from the mRNA and ligated into a recombinant vector. The vector is transfected into a recombinant host for propagation, screening, and cloning. Methods for making and screening cDNA libraries are well known. See Gubler and Hoffman (1983) Gene 25:263-269 and Sambrook, et al.
  • the DNA is extracted from tissue and either mechanically sheared or enzymatically digested to yield fragments of about 12-20 kb. The fragments are then separated by gradient centrifugation and cloned in bacteriophage lambda vectors. These vectors and phage are packaged in vitro, as described in Sambrook, et al. Recombinant phage are analyzed by plaque hybridization as described in Benton and Davis (1977) Science 196:180-182. Colony hybridization is carried out as generally described in e.g., Grunstein, et al. (1975) Proc. Natl Acad. Sci. USA ⁇ 72:3961-3965.
  • Oligonucleotides for use as probes are usually chemically synthesized according to the solid phase phosphoramidite triester method first described by Beaucage and Carruthers (1983) Tetrahedron Lett. 22(20):1859-1862, or using an automated synthesizer, as described in Needham-VanDevanter, et al. (1984) Nucleic Acids Res. 12:6159-6168. Purification of oligonucleotides is performed e.g., by native acrylamide gel electrophoresis or by anion-exchange HPLC as described in Pearson and Regnier (1983) J. Chrom. 255:137-149.
  • LP proteins of the invention exhibit limited similarity to portions other intracellular proteins.
  • beta-sheet and alpha-helix residues can be determined using, e.g., RASMOL program, see Sayle and Milner-White (1995) TIBS 20:374-376; or Gronenberg, et al. (1991) Protein Engineering 4:263-269; and other structural features are defined in Lodi, et al. (1994) Science 263:1762-1767.
  • This invention provides isolated DNA or polynucleotide fragments to encode an
  • this invention provides isolated or recombinant DNA that encodes a protein or polypeptide which is capable of hybridizing under appropriate conditions, e.g., high stringency, with the DNA sequences described herein.
  • Said biologically active protein or polypeptide can be an intact protein, or fragment, and have an amino acid sequence as disclosed in SEQ ID NO: Y (particularly natural embodiments), or as listed in a Table herein, or as encoded in a described deposited clone.
  • Preferred embodiments are full-length natural sequences.
  • this invention contemplates the use of isolated or recombinant DNA, or fragments thereof, which encode proteins that have sequence similarity (or identity) to an LP protein or which were isolated using cDNA encoding a LP protein as a probe.
  • the isolated DNA can have the respective regulatory sequences in the 5' and 3' flanks, e.g., promoters, enhancers, poly-A addition signals, and others. Also embraced are methods for making expression vectors with these sequences, or for making, e.g., expressing and purifying, protein products.
  • a DNA sequence that codes for an LP protein is particularly useful to identify genes, mRNA, and cDNA specie that code for related or similar proteins, as well as DNAs that code for homologous and/or proteins from different species that share sequence similarity or identity.
  • homologs e.g., orthologs and paralogs
  • similar sequences e.g., gene duplications
  • Various homologous LP proteins are encompassed herein. However, even proteins that have a more distant evolutionary relationship to an LP antigen can readily be isolated under appropriate conditions using these sequences if they are sufficiently structurally similar. Of particular interest, are primate LP proteins.
  • Recombinant clones derived from the genomic sequences, e.g., containing introns, will be useful for transgenic studies, including, e.g., transgenic cells and organisms, and for gene therapy (see, e.g., Goodnow (1992) "Transgenic Animals” in Roitt (ed.) Encyclopedia of Immunology, Academic Press, San Diego, pp. 1502-1504; Travis (1992) Science 256:1392-1394; Kuhn, et al.
  • Antibodies of the invention include, e.g., without limitation, polyclonal, monoclonal, multispecific, human, humanized or chimeric antibodies, single chain antibodies, Fab fragments, F(ab') fragments, fragments produced by a Fab expression library, anti-idiotypic (anti-Id) antibodies (including, e.g., anti-Id antibodies to antibodies of the invention), and an epitope-binding fragment of any of the above.
  • antibody refers to immunoglobulin compositions and immunologically active portions of immunoglobulin compositions, e.g., a molecule that contains an antigen binding site that specifically binds an antigen.
  • An immunoglobulin composition of the invention can be of any type (e.g., IgG, IgE, IgM, IgD, IgA, and IgY), class (e.g., IgGl, IgG2, IgG3, IgG4, IgAl, and IgA2) or a subclass of an immunoglobulin molecule.
  • an antibody is a human antigen-binding antibody fragment of the present invention such as, e.g., without limitation, Fab, Fab' and F(ab')2, Fd, 6 single- chain Fvs (scFv), single-chain antibodies, disulfide-linked Fvs (sdFv), and fragments comprising either a VL or VH domain.
  • Antigen-binding antibody fragments may comprise the variable region(s) alone or in combination with the entirety or a portion of the following: a hinge region, a CHI, a CH2, or a CH3 domain or combinations thereof. Also included in the invention is, e.g., without limitation, an antigen-binding fragment that also can comprise any combination of variable region(s) with a hinge region, e.g., such as a CHI, CH2, or a CH3 domain or combinations thereof.
  • An antibody of the invention may be from any animal origin including birds and mammals. Preferably, the antibodies are human, murine (e.g., mouse and rat), donkey, rabbit, goat, guinea pig, camel, horse, or chicken.
  • human antibodies includes, e.g., without limitation, antibodies having an amino acid sequence of a human immunoglobulin including, e.g., without limitation, an antibody isolated from a human immunoglobulin library or from an animal transgenic for one or more human immunoglobulins and that do not express endogenous immunoglobulins, as described herein or, as taught, e.g., in U.S. Patent No. 5,939,598.
  • An antibody of the present invention may be monospecific, bispecific, trispecific or of greater multispecificity.
  • Multispecific antibodies may be specific for different epitopes of an LP polypeptide (or fragment thereof) or may be specific for both a polypeptide of the present invention as well as for a heterologous epitope, such as a heterologous polypeptide or solid support material (see, e.g., WO 2093/17715; WO
  • An antibody of the present invention may be described or specified in terms of an epitope(s) or portion(s) of an LP polypeptide (or fragment thereof) that it recognizes or selectively binds.
  • An epitope(s) or polypeptide portion(s) may be specified as described herein, e.g., by N-terminal and C-terminal positions, by size in contiguous amino acid residues, or as listed in an accompanying Table and/or Figure. Additionally, an antibody that specifically binds an epitope, polypeptide, protein, or fragment of a polypeptide or protein of the present invention, may also be specifically excluded from this invention. For instance, Applicants reserve the right to proviso out any antibody that specifically binds an epitope, polypeptide, protein, or fragment of a polypeptide or protein of the present invention.
  • the present invention can encompass a first (or other) antibody that specifically binds a polypeptide or protein, or fragment thereof, of the present invention, and, at the same time, it can exclude a second (or other) antibody that may also selectively bind the same protein or polypeptide, or fragment thereof, e.g., by binding a different epitope.
  • Antibodies of the present invention may also be described or specified in terms of their cross-reactivity. Antibodies that do not bind any other analog, ortholog, paralog, or homolog of an LP polypeptide (or fragment thereof) are included.
  • Antibodies that bind polypeptides with at least 95%, at least 90%, at least 85%, at least 80%, at least 75%, at least 70%, at least 65%, at least 60%, at least 55%, and at least 50% identity (as calculated using known methods or as described herein) to an LP polypeptide (or fragment thereof) are also included.
  • Specific embodiments include, e.g., antibodies of the present invention cross-react with murine, rat and/or rabbit homologs of human proteins, and the corresponding epitopes thereof.
  • Antibodies that do not bind polypeptides with less than 95%, less than 90%, less than 85%, less than 80%, less than 75%, less than 70%, less than 65%, less than 60%, less than 55%, and less than 50% identity (as calculated using known methods or as described herein) to an LP polypeptide (or fragment thereof) are also included.
  • Specific embodiments include, e.g., the above-described cross- reactivity with respect to any single specific antigenic or immunogenic polypeptide, or combination(s) of 2, 3, 4, 5, or more of the specific antigenic and or immunogenic polypeptides disclosed herein.
  • an antibody that selectively binds a polypeptide, which is encoded by a polynucleotide that stably hybridizes, under stringent hybridization conditions (as described herein), to an LP polynucleotide sequence.
  • An antibody of the present invention may also be characterized or specified in terms of its binding affinity to a protein or polypeptide (fragment thereof), or epitope of the invention.
  • a preferred binding affinity of a binding composition includes, e.g., a binding affinity that demonstrates a dissociation constant or Kd of less than about: 5 X 10 "2 M, 10 "2 M, 5 X 10 "3 M, 10 "3 M, 5 X lO ⁇ M, 10 "4 M, 5 X 10 "5 M, 10 '5 M, 5 X 10 _6 M, 10 "6 M, 5 X 10 "7 M, 10 "7 M, 5 X 10 “8 M, 10 “8 M, 5 X 10 "9 M, 10 "9 M, 5 X 10- 10 M, 10- 10 M, 5 X 10 _11 M, 10 " ⁇ M, 5 X 10 "12 M, 10 “12 M, 5 X 10 " 13 M, 10 _13 M, 5 X 10 "14 M, 10 '14 M, 5 X 10 "15 M, or 10 "15 M.
  • the invention also encompasses antibodies that competitively inhibit binding of a binding composition to an epitope of the invention as determined by any known art method for determining competitive binding, e.g., the immunoassays described herein.
  • the antibody competitively inhibits binding to the epitope by at least 95%, at least 90%, at least 85%, at least 80%, at least 75%, at least 70%, at least
  • Antibodies of the present invention may act as agonists or antagonists of an LP polypeptide (or fragment thereof).
  • an antibody or binding composition of present invention can disrupt, e.g., an interaction, either partially or completely, of a polypeptide of the invention with its cognate receptor/ligand.
  • antibodies of the present invention bind an antigenic epitope disclosed herein, or a portion thereof.
  • the invention encompasses both receptor-specific antibodies and ligand-specific antibodies.
  • Receptor activation e.g., signaling
  • receptor activation can be determined by techniques described herein or otherwise known in the art. For example, receptor activation can be determined by detecting phosphorylation
  • antibodies 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
  • the invention also features receptor-specific antibodies that 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.
  • receptor-specific antibodies that 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.
  • neutralizing antibodies that bind a ligand and prevent it binding to a receptor.
  • ligand-binding antibodies that inhibit receptor activation without inhibiting receptor binding are also included.
  • Antibodies of the invention may act as receptor agonists, e.g., by potentiating or activating either all or a subset of the biological activities of the ligand-mediated receptor activation, e.g., by inducing dimerization of a receptor.
  • the antibodies may be specified as agonists, antagonists, or inverse agonists for biological activities comprising the specific biological activities of a peptide of the invention disclosed herein.
  • An antibody agonist can be made using known methods art (see, e.g., WO 96/40281; U.S. Patent No.
  • Antibodies of the present invention may be used, e.g., without limitation, to purify, detect, or target a polypeptide (or fragment thereof) of the present invention for, e.g., in vitro and/or in vivo diagnostic and therapeutic methods.
  • the antibodies have use in immunoassays for qualitatively and/or quantitatively measuring levels of a polypeptide (or fragment thereof) of the present invention in a biological sample (see, e.g., Harlow, et al., Antibodies: A Laboratory Manual, Cold Spring Harbor Laboratory Press, cur. ed.; incorporated by reference).
  • an antibody of the present invention may be used either alone or in combination with other compositions.
  • an antibody may be recombinantly fused to a heterologous polypeptide at the N- or C-terminus, or chemically conjugated (including covalently and non-covalently conjugations) to a polypeptide or other compositions.
  • antibodies of the present invention may be recombinantly fused or conjugated to molecules useful as labels in detection assays and effector molecules such as heterologous polypeptides, drugs, radionuclides, or toxins (see, e.g., WO 92/08495; WO 91/14438; WO 89/12624; U.S. Patent No. 5,314,995; and EP 396,387).
  • An antibody of the invention includes, e.g., derivatives that are modified, e.g., by the covalent attachment of any type of molecule to the antibody such that the covalent attachment does not prevent the antibody from generating an anti-idiotypic response.
  • an antibody derivative includes, e.g., antibodies that have been modified, e.g., by glycosylation, acetylation, pegylation, phosphylation, amidation, derivatization by known protecting/blocking groups, proteolytic cleavage, linkage to a cellular ligand or other protein, etc.
  • a derivative may contain one or more non-classical amino acids.
  • An antibody of the present invention may be generated by any suitable known art method.
  • Polyclonal antibodies to an antigen-of-interest can be produced by various procedures known in the art.
  • a polypeptide of the invention can be administered to various host animals including, e.g., without limitation, rabbits, mice, rats, etc., to induce the production of sera containing polyclonal antibodies specific for the antigen.
  • adjuvants may be used to increase an immunological response depending on the host species, these include, e.g., without limitation, Freund's (complete and incomplete), mineral gels such as e.g., aluminum hydroxide, surface active substances such as e.g., lysolecithin, plutonic polyols, polyanions, peptides, oil emulsions, keyhole limpet hemocyanins, dinifrophenol, and potentially useful human adjuvants such as, e.g., BCG (bacille Calmette-Guerin) and corynebacterium parvum.
  • BCG Bacille Calmette-Guerin
  • corynebacterium parvum Such adjuvants are known in the art.
  • Monoclonal antibodies can be prepared using a variety of art known techniques including, e.g., the use of hybridoma, recombinant, and phage display technologies, or combinations thereof.
  • monoclonal antibodies can be produced using hybridoma techniques including those known in the art and taught, e.g., in Harlow, et al., Antibodies: A Laboratory Manual, (Cold Spring Harbor Laboratory Press, current edition); Hammerling, et al., in: Monoclonal Antibodies and T-Cell Hybridomas 563-68 1 (Elsevier, N.Y., 1981) (each of which are incorporated by reference for theses teachings).
  • the mouse spleen is harvested, and splenocytes isolated.
  • the splenocytes are then fused by known techniques to any suitable myeloma cells; e.g., SP20 cells (available from the ATCC).
  • Hybridomas are then selected and cloned by limited dilution.
  • the hybridoma clones are then assayed by art known methods to discover cells that secrete antibodies that bind an LP polypeptide (or fragment thereof).
  • Ascites fluid which generally contains high levels of antibodies, can be generated by immunizing mice with positive hybridoma clones.
  • the present invention provides methods of generating monoclonal antibodies as well as antibodies produced by the method comprising culturing a hybridoma cell secreting an antibody of the invention 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 an LP polypeptide.
  • Antibody fragments that recognize specific epitopes may be generated by known techniques.
  • Fab and F(ab')2 fragments of the invention may be produced by proteolytic cleavage of immunoglobulin molecules, using enzymes such as papain (to produce Fab fragments) or pepsin (to produce F(ab')2 fragments).
  • F(ab')2 fragments contain the variable region, the light chain constant region and the CHI domain of the heavy chain.
  • an antibody of the present invention can also be generated using various phage display methods known in the art in which functional antibody domains are displayed on the surface of phage particles, which carry a polynucleotide sequence encoding them.
  • a phage display method is used to display antigen- binding domains expressed from a repertoire or combinatorial antibody library (e.g., human or murine).
  • Phage that express an antigen binding domain that binds an 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 filamentous phage including, e.g., fd and M 13 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.
  • phage display methods contemplated for use include, e.g., those of Brinkman, et al., J. Immunol. Methods 182:41-50 (1995); Ames, et al., J. Immunol. Methodsl84:177-186 (1995); Kettleborough, et al, Eur. J. Immunol. 24:952-958 (1994); Persicet, et al., Gene 1879-18 (1997); Burton, et al, Advances in rmmunology 57:191-280(1994); PCT application No.
  • antibody coding regions from a phage are 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 herein and in the literature.
  • Fab, Fab' and F(ab')2 fragments can also be employed using art known methods such as, e.g., WO 92122324; 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) (which are incorporated by reference for these teachings).
  • WO 92122324 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) (which are incorporated by reference for these teachings).
  • Examples of producing single-chain Fvs and antibodies include, e.g., U.S.
  • Patents 4,946,778 and 5,258,498 Huston, et al., Methods m Enzymology 203:46-88 (1991); Shu, et al., Proc. Natl. Acad. Sci. USA 90:7995-7999(1993); and Skerra, et al., Science 240: 1038-1040 (1988).
  • chimera 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.
  • Humanized antibodies are antibody molecules from non-human species that bind a desired antigen having one or more complementarity determining regions (CDRs) from a non-human species and a framework region from a human immunoglobulin molecule.
  • CDRs complementarity determining regions
  • Human antibodies can be made using a variety of known methods including, e.g., phage display methods described herein using antibody libraries derived from human immunoglobulin sequences (see e.g., U.S. Patent Nos. 4,444,887 and4,716,ll l; and WO 98/46645, WO 98150433, WO 00/58513104 WO 98124893, WO 98116654, WO 96134096, WO 96133735, and WO 91/10741; each of which is incorporated herein by reference for its teachings on human antibodies). Human antibodies can also be produced using transgenic mice that are incapable of expressing functional endogenous immunoglobulins, but which can express human immunoglobulin genes.
  • human heavy and light chain immunoglobulin gene complexes may be introduced randomly or by homologous recombination into mouse embryonic stem cells.
  • human variable regions, constant regions, and diversity regions may be introduced into mouse embryonic stem cells in addition to the human heavy and light chain genes.
  • the mouse heavy and light chain immunoglobulin genes may be rendered non-functional (separately or simultaneously) with the introduction of human immunoglobulin loci by homologous recombination.
  • homozygous deletion of the JH region prevents endogenous antibody production.
  • the modified embryonic stem cells are expanded and microinjected into blastocysts to produce chimeric mice. The chimeric mice are then bred to produce homozygous offspring that express human antibodies.
  • the transgenic mice are immunized in the normal fashion with a selected antigen, e.g., all or a portion of a (or fragment thereof) polypeptide of the invention.
  • Monoclonal antibodies directed against the antigen can be obtained from an immunized, transgenic mouse 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, it is possible to produce therapeutically useful IgG, IgA, IgM, and IgE antibodies.
  • antibodies of the invention can, in turn, be used to generate anti-idiotype antibodies that "mimic" a polypeptide (or fragment thereof) of the invention using known techniques (see, e.g., Greenspan & Bona, FASEB J. 7(5):437-444; (1989) and Nissinoff, J. (1991) Immunol. 147(8):2429-2438).
  • antibodies that bind and competitively inhibit polypeptide multimerization and/or competitively inhibit binding of a polypeptide of the invention to a ligand can be used to generate anti-idiotypes that "mimic" the polypeptide multimerization and or binding domain and, as a consequence, bind to and neutralize a polypeptide and/or its ligand.
  • Such neutralizing anti-idiotypes, or Fab fragments of such anti-idiotypes can be used in therapeutic regimens to neutralize a polypeptide ligand.
  • anti-idiotypic antibodies can be used to bind a polypeptide of the invention (or fragment thereof) and/or to bind its ligand/receptor, and thereby block its biological activity.
  • Polynucleotides Encoding Antibodies The invention further provides a nucleic acid molecule comprising a polynucleotide sequence encoding an antibody of the invention and/or a fragment thereof.
  • the invention also encompasses polynucleotides that hybridize under stringent or lower stringency hybridization conditions, e.g., as defined herein, to a polynucleotide that encodes an antibody, preferably, that specifically and/or selectively binds a mature polypeptide or protein of the invention, preferably, an antibody that binds to a mature polypeptide of SEQ ID NO:Y.
  • the polynucleotides may be obtained, and the nucleotide sequence of the polynucleotides determined, by any known art method.
  • a polynucleotide encoding the antibody may be assembled from chemically synthesized oligonucleotides (e.g., as described in Kutmeier, et al., (1994) BioTechniques 17:242), which, briefly described, involves synthesizing overlapping oligonucleotides containing portions of the sequence encoding the antibody, annealing, and ligating those oligonucleotides, then, amplifying the ligated oligonucleotides using a polymerase chain reaction.
  • a polynucleotide encoding an antibody can be generated from nucleic acid of any suitable source.
  • a nucleic acid encoding the immunoglobulin may be chemically synthesized or obtained from a suitable source.
  • the source may be an antibody cDNA library, or a cDNA library generated from poly A+ RNA, isolated from any tissue or cell expressing the antibody of interest, such as, e.g., a hybridoma cells selected to express an antibody of the invention by PCR amplification using synthetic primers hybridizable to the 3' and 5' ends of a polynucleotide sequence of interest 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, a nucleic acid molecule for the antibody can be generated.
  • a hybridoma cells selected to express an antibody of the invention by PCR amplification using synthetic primers hybridizable to the 3' and 5' ends of a polynucleotide sequence of interest or by cloning using an oligonucleotide probe specific for the particular gene sequence to identify, e.g., a
  • Amplified nucleic acids may be cloned into replicable cloning vectors using any known art method.
  • the nucleotide sequence of the antibody may be manipulated using any known art method, e.g., recombinant DNA techniques, site directed mutagenesis, PCR, etc. to generate antibodies having a different amino acid sequence to create amino acid substitutions, deletions, and/or insertions (see, e.g., Sambrook, et al., and Ausubel, et al., eds., cur.
  • the amino acid sequence of the heavy and/or light chain variable domains may be inspected to identify the sequences of complementarity determining regions (CDRs) by known methods, e.g., by comparing known amino acid sequences of other heavy and chain light variable regions to determine regions of sequence hypervariability.
  • CDRs complementarity determining regions
  • one or more CDRs may be inserted within framework regions, e.g., into human framework regions to humanize a non-human antibody.
  • the framework regions may be naturally occurring or consensus framework regions, and are preferably human framework regions (for a listing of human framework regions see, e.g., Chothia, et al. (1998) J. Mol. Biol. 278: 457-479).
  • the polynucleotide generated by the combination of the framework regions and CDRs encodes an antibody that specifically binds a polypeptide of the invention (or fragment thereof).
  • one or more amino acid substitutions may be made within the framework regions to improve binding of the antibody to its antigen.
  • Such methods may be used to make amino acid substitutions or deletions of one or more variable-region, cysteine residues participating in an intrachain disulfide bond to generate antibody molecules lacking one or more intrachain disulfide bonds.
  • Other alterations to the polynucleotide are encompassed by the present invention and within the skill of an ordinary artisan, e.g., such as a molecular biologist.
  • "chimeric antibody” techniques can be used by, e.g., splicing genes from a mouse antibody molecule of appropriate antigen specificity together with genes from a human antibody molecule of appropriate biological activity (see, e.g., Morrison, et al., Proc. Natl. Acad. Sci.
  • a chimeric antibody e.g., humanized antibodies
  • a chimeric antibody is a molecule in which different portions are derived from different animal species, such as those having a variable region, derived from a murine mAb and constant region from a human immunoglobulin, e.g., humamzed antibodies.
  • techniques can be adapted to produce single chain antibodies (see, e.g., U.S. Patent No. 4,946,778; Bird, Science 242:423-42 (1988); Huston, et al., Proc. Natl.
  • Single chain antibodies are formed by linking the heavy and light chain fragments of the Fv region via an ammo 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. (1988) Science 242: 1038- 1041).
  • Methods of Producing Antibodies An antibody or binding composition of the invention can be produced by any known art method, in particular, by chemical synthesis or preferably, by recombinant expression techniques.
  • Recombinant expression of an antibody of the invention, or fragment, derivative or analog thereof, requires construction of an expression vector containing a polynucleotide sequence that encodes the antibody.
  • an expression vector containing a polynucleotide sequence that encodes the antibody Once a polynucleotide sequence 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 known recombinant DNA technology techniques. Methods known in the art can be used to construct expression vectors containing antibody coding sequences and appropriate transcriptional and translational control signals.
  • the invention provides replicable vectors comprising a nucleotide sequence encoding an antibody molecule of the invention, or a heavy or light chain thereof, or a heavy or light chain variable domain, operably-linked to a promoter.
  • Such vectors may include, e.g., the nucleotide sequence encoding the constant region of the antibody molecule (see, e.g., WO 86/05807; WO 89/01036; or U.S. Patent No.
  • the variable domain of the antibody may be cloned into such a vector for expression of the entire heavy or light chain.
  • an expression vector is transferred to a host cell by conventional techniques and the transfected cells are then cultured to produce an antibody.
  • the invention includes, e.g., host cells containing a polynucleotide encoding an antibody of the invention, or a heavy or light chain thereof, or a single chain antibody of the invention, operably linked to a heterologous promoter.
  • vectors encoding both the heavy and light chains may be co-expressed in a host cell for expression of the entire immunoglobulin molecule, as detailed herein or known the art.
  • host-expression vector systems may be utilized to express antibody molecules of the invention.
  • Such host-expression systems represent vehicles by which any coding sequence of interest may be produced and subsequently purified.
  • host- expression system cells may also represent an antibody molecule of the invention in situ. These cells include, e.g., without limitation, microorganisms such as bacteria (e.g., E. coli, B.
  • 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
  • plant cell systems infected with recombinant virus expression vectors e.g., cauliflower mosaic virus, CaMV; tobacco mosaic virus, TMV
  • recombinant plasmid expression vectors e.g., Ti plasmid
  • mammalian cell systems e.g., COS, CHO, BHK, 293, 3T3 cells harboring recombinant expression constructs containing promoters derived from the genome of mammalian cells (e.g., metallothionein promoter)
  • bacterial cells such as Escherichia coli, and more preferably, eukaryotic cells, especially for the expression of whole recombinant antibody molecule, are used for the expression of a recombinant antibody molecule.
  • mammalian cells such as Chinese hamster ovary cells (CHO)
  • CHO Chinese hamster ovary cells
  • a vector such as the major intermediate early gene promoter element from human cytomegalovirus is an effective expression system for antibodies (Foecking, et al. (1986) Gene 45:101; Cockett, et al. (1990) Bio/Technology 8:2).
  • a number of expression vectors may be advantageously selected depending upon the intended use of the expressed antibody molecule.
  • vectors that direct the expression of high levels of fusion protein products, which are readily purified may be desirable.
  • Such vectors include, e.g., without limitation, the E. coli expression vector pUR278 (Ruther, et al., ⁇ MBO J. 2: 1791 (1983)), in which the antibody coding sequence may be ligated individually into the vector in frame with the lac Z coding region so that a fusion protein is produced; pIN vectors (Inouye and Inouye, Nucleic Acids Res. 13:3 101- 3 109 (1985); Van Heeke and Schuster, J.
  • pG ⁇ X vectors may also be used to express foreign polypeptides as fusion proteins with glutathione S-transferase (GST).
  • GST glutathione S-transferase
  • fusion proteins are soluble and can easily be purified from lysed cells by adsorption and binding to matrix glutathione-agarose beads followed by elution in the presence of free glutathione.
  • the pG ⁇ X vectors are designed to include, e.g., thrombin, or factor Xa protease cleavage sites so that the cloned target gene product can be released from a GST moiety.
  • AcNPV Autographa californica nuclear polyhedrosis virus
  • the AcNPV virus grows in Spodopteru frugiperda cells.
  • the antibody coding sequence may be cloned individually into non-essential regions (e.g., the polyhedrin gene) of the virus and placed under control of an AcNPV promoter (e.g., the polyhedrin promoter).
  • an AcNPV promoter e.g., the polyhedrin promoter
  • a number of viral-based expression systems may be utilized.
  • the antibody coding sequence of interest may be ligated to an adenovirus transcription/translation control complex, e.g., the late promoter and tripartite leader sequence.
  • This chimeric gene may then be inserted in the adenovirus genome using in vitro or in vivo recombination. Insertion in a non-essential region of the viral genome (e.g., region ⁇ l or ⁇ 3) results in a recombinant virus that is viable and capable of expressing the antibody molecule in infected hosts (see, e.g., Logan and Shenk, (1984) Proc. Natl. Acad. Sci. USA 8 1 :355-359).
  • Specific initiation signals may be required for efficient translation of inserted antibody coding sequences. These signals include, e.g., 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 proper 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, e.g., Bittner, et al., Methods in Enzymol. 153:5 1-544 (1987)).
  • a host cell strain may be chosen that modulates the expression of the inserted sequence, or modifies and processes the gene product in the specific fashion desired. Such modifications (e.g., glycosylation) and processing (e.g., cleavage) of protein products may be important for the function of a protein.
  • Different host cells have characteristic and specific mechanisms for the post- translational processing and modification of proteins and gene products. Appropriate cell lines or host systems can be chosen to ensure the correct modification and processing of a foreign protein that is expressed. To this end, eukaryotic host cells that possess the cellular machinery for proper: processing of the primary transcript, glycosylation, and phosphorylation may be used.
  • Such mammalian host cells include, e.g., without limitation, CHO, VERY, BHK, Hela, COS, MDCK, 293, 3T3, W138, and, in particular, breast cancer cell lines such as, e.g., BT483, Hs578T, HTB2, BT20 and T47D, and normal mammary gland cell line such as, e.g., CRL7030 and Hs578Bst.
  • breast cancer cell lines such as, e.g., BT483, Hs578T, HTB2, BT20 and T47D
  • normal mammary gland cell line such as, e.g., CRL7030 and Hs578Bst.
  • stable expression is preferred.
  • cell lines that stably express an antibody molecule may be engineered.
  • host cells can be transformed with controlled by appropriate expression control elements (e.g., promoter, a polynucleotide sequence enhancer sequences, transcription terminators, polyadenylation sites, etc.), and a selectable marker.
  • expression control elements e.g., promoter, a polynucleotide sequence enhancer sequences, transcription terminators, polyadenylation sites, etc.
  • selectable marker in a recombinant plasmid confers resistance to selection and allows cells to stably integrate the plasmid into their chromosomes and to form foci that can be subsequently cloned and expanded into cell lines. This method can be used to engineer cell lines that express an antibody molecule.
  • Such engineered cell lines are particularly useful in screening and evaluating compounds that interact either directly or indirectly with an antibody molecule of the invention.
  • a number of selection systems can be used, including, e.g., without limitation, herpes simplex virus thymidine kinase (Wigler, et al., Cell 11 :223 (1977)) in the cells, hypoxanthine-guanine phosphoribosyltransferase in hgprt- cells (Szybalska and Szybalski, Proc. Natl. Acad. Sci.
  • anti- metabolite resistance can be used as the basis of selection for the following genes: dhfr — which confers resistance to methotrexate (Wigler, et al., Natl. Acad. Sci. USA 77:357 (1980); O'Hare, et al, Proc. Natl. Acad. Sci. USA 78:1527 (1981)); gpt— which confers resistance to mycophenolic acid (Mulligan and Berg, Proc. Natl. Acad. Sci.
  • neo which confers resistance to the aminoglycoside G-20418 (Clinical Pharmacy 12:488-505; Wu and Wu, Biotherapy 3:87-95 (1991)); Tolstoshev, Ann. Rev. Pharmacol. Toxicol. 32:573-596 (1993); Mulligan, Science 260:926-932 (1993); and Morgan and Anderson, Ann. Rev. Biochem. 62: 19 1-2 17 (1993); May, 1993, TJB TECH 1 1(5): 155-215); and hygro — which confers resistance to hygromycin (Santerre, et al., Gene 30: 147 (1984)).
  • Known art methods can be routinely applied to select a desired recombinant clone
  • 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 that enable equal expression of heavy and light chain polypeptides.
  • a single vector may be used that encodes, and is capable of expressing, both heavy and light chain polypeptides.
  • the light chain should be placed before the heavy chain to avoid an excess of toxic free heavy chain (Proudfoot, Nature 322:52 (1986); Kohler, Proc. Natl. Acad. Sci. USA 77:2197 (1980)).
  • Coding sequences for heavy and light chains may comprise cDNA or genomic DNA.
  • an antibody molecule of the invention may be purified by any known method, e.g., by chromatography (e.g., ion exchange, affinity, particularly by affinity for the specific antigen after Protein A, and sizing column chromatography), centrifugation, differential solubility, or any other technique for protein purification.
  • chromatography e.g., ion exchange, affinity, particularly by affinity for the specific antigen after Protein A, and sizing column chromatography
  • centrifugation e.g., differential solubility, or any other technique for protein purification.
  • an antibody of the present invention or fragments thereof can be fused to heterologous polypeptide sequences to facilitate purification using any art known method or one described herein.
  • antibodies may be used to target an LP polypeptide (or fragment thereof) to particular cell types, either in vitro or in vivo, by fusing or conjugating a polypeptide (or fragment thereof) of the present invention to an antibody specific for a particular cell surface receptor.
  • Antibodies fused or conjugated to a polypeptide of the invention may also be used in in viti'o irnmunoassays and in purification methods using known art methods (see e.g., Harbor, et al, supra, and WO 9312 1232; EP 439,095; Naramura et al. (1994) Immunol. Lett. 39:9 1-99; U.S. Patent No.
  • the present invention further includes compositions comprising a polypeptide of the invention (or fragment thereof) fused or conjugated to an antibody domain other than a variable region.
  • a polypeptide of the invention (or fragment thereof) may be fused or conjugated to an antibody Fc region, or portion thereof.
  • a polypeptide, polypeptide fragment, or a variant of SEQ ID NO:Y may be fused or conjugated to an antibody portion described herein or known in the art to increase the in vivo half-life. Further, a polypeptide, polypeptide fragment, or a variant of SEQ ID NO: Y (or part thereof) may be fused or conjugated to an antibody portion to facilitate purification.
  • chimeric proteins comprising the first two domains of the human CD4-polypeptide and various domains of the constant regions of the heavy or lighl chains of mammalian immunoglobulins.
  • S A polypeptide, polypeptide fragment, or a variant of SEQ ID NO: Y (or part thereof) fused or conjugated to an antibody having disulfide-linked dimeric structures (due to the IgG) may also be more efficient in binding and neutralizing other molecules, than the monomeric secreted protein or protein fragment alone (see, e.g., Fountoulakis, et al.
  • the Fc part of a fusion protein is beneficial in therapy and diagnosis, and thus can result in, e.g., improved pharmacokinetic properties (see, e.g., EP A232, 262).
  • deleting the Fc part after the fusion protein has been expressed, detected, and purified, can be favored.
  • the Fc portion may hinder therapy and diagnosis if the fusion protein is used as an antigen for immunizations.
  • human proteins such as hIL-5
  • Fc portions for the purpose of high-throughput screening assays to identify antagonists of hIL-5
  • an antibody of the present invention can be fused to marker sequences, such as a peptide to facilitate purification.
  • the marker amino acid sequence is a hexa-histidine peptide, such as the tag provided in a pQE vector (QIAGEN, Inc., Chatsworth, CA), among others, many of which are commercially available.
  • Hexa-histidine provides for convenient purification of a fusion protein (Gentz, et al. (1989) Proc. Natl. Acad. Sci. USA 86:821-824).
  • Other peptide tags useful for purification include, e.g., the "HA” tag, which corresponds to an epitope derived from the influenza hemagglutinin protein (Wilson, et al. (1984) Cell 37:767) and the "flag" tag.
  • DNAs which encode a LP protein or fragments thereof can be obtained by chemical synthesis, screening cDNA libraries, or by screening genomic libraries prepared from a wide variety of cell lines or tissue samples. Methods for doing so, or making expression vectors are either art known or are described herein. These DNAs can be expressed in a wide variety of host cells for the synthesis of a full-length protein or fragments which can in turn, e.g., be used to generate polyclonal or monoclonal antibodies; for binding studies; for construction and expression of modified molecules; and for structure/function studies. Each LP protein or its fragments can be expressed in host cells that are transformed or transfected with appropriate expression vectors.
  • LP polypeptides can be substantially purified to be free of protein or cellular contaminants, other than those derived from the recombinant host, and therefore are particularly useful in pharmaceutical compositions when combined with a pharmaceutically acceptable carrier and/or diluent.
  • the antigen e.g., LP protein, or portions thereof, may be expressed as fusions with other proteins or possessing an epitope tag.
  • expression vectors also contain an origin of replication that allows the vector to replicate independently of the host cell.
  • An expression vector will preferably include, e.g., at least one selectable marker.
  • markers include, e.g., without limit, dihydrofolate reductase, G418, or neomycin resistance for eukaryotic cell culture and tetracycline, kanamycin or ampicillin resistance genes for culturing in E. coli and other bacteria.
  • the vectors of this invention contain DNAs which encode an LP protein, or a fragment thereof, typically encoding, e.g., a biologically active polypeptide, or protein.
  • the DNA can be under the control of a viral promoter and can encode a selection marker.
  • This invention further contemplates use of expression vectors capable of expressing eukaryotic cDNA coding for a LP (or fragment) in a prokaryotic or eukaryotic host, where the vector is compatible with the host and where the eukaryotic cDNA coding for the protein is inserted into the vector such that growth of the host containing the vector expresses the cDNA in question.
  • expression vectors are designed for stable replication in their host cells or for amplification to greatly increase the total number of copies of the desirable gene per cell. It is not always necessary to require that an expression vector replicate in a host cell, e.g., it is possible to effect transient expression of the protein or its fragments in various hosts using vectors that do not contain a replication origin that is recognized by the host cell. It is also possible to use vectors that cause integration of an LP protein gene or its f agments into the host DNA by recombination, or to integrate a promoter that controls expression of an endogenous gene.
  • Vectors encompass plasmids, viruses, bacteriophage, integratable DNA fragments, and other vehicles that enable the integration of DNA fragments into the genome of the host.
  • Expression vectors are specialized vectors that contain genetic control elements that effect expression of operably linked genes. Plasmids are the most commonly used form of vector, but many other forms of vectors that perform an equivalent function are also suitable for use (see, e.g., Pouwels, et al. (1985 and Supplements) Cloning Vectors: A Laboratory Manual Elsevier, N.Y.; and Rodriquez, et al. (eds.) (1988) Vectors: A Survey of Molecular Cloning Vectors and Their Uses Buttersworth, Boston, MA).
  • a plasmid vector is introduced in a precipitate, such as, e.g., a calcium phosphate precipitate, or in a complex with a charged lipid.
  • a virus If the vector is a virus, it may be packaged in vitro using an appropriate packaging cell line and then transduced into host cells.
  • the polynucleotide insert should be operatively linked to an appropriate promoter, such as, e.g., without limit, the phage lambda PL promoter, the E. coli lac, trp, phoA, and tat promoters, the SV40 early or late promoters, and promoters of retroviral LTRs.
  • an appropriate promoter such as, e.g., without limit, the phage lambda PL promoter, the E. coli lac, trp, phoA, and tat promoters, the SV40 early or late promoters, and promoters of retroviral LTRs.
  • Suitable host cells include prokaryotes, lower eukaryotes, and higher eukaryotes.
  • Prokaryotes include both gram negative and gram positive organisms, e.g., E. coli and B. subtilis.
  • Lower eukaryotes include yeasts, e.g., S. cerevisiae and Pichia, and species of the genus Dictyostelium.
  • Higher eukaryotes include established tissue culture cell lines from animal cells, both of non-mammalian origin, e.g., insect cells, and birds, and of mammalian origin, e.g., human, primates, and rodents.
  • Prokaryotic host- vector systems include a variety of vectors for many different species.
  • E. coli and its vectors will be used generically to include equivalent vectors used in other prokaryotes.
  • a representative vector for amplifying DNA is pBR322 or its derivatives.
  • Vectors that can be used to express these proteins or protein fragments include, but are not limited to, such vectors as those containing the lac promoter (pUC-series); trp promoter (pBR322-trp); Ipp promoter (the pIN-series); lambda-pP or pR promoters (pOTS); or hybrid promoters such as ptac (pDR540). See Brosius, et al.
  • bacterial vectors include, e.g., without limit, pQE70, pQE60, and pQE-9, (available from QIAGEN, Inc.); pBluescript vectors, Phagescript vectors, pNH8A, pNH16a, pNH18A, pNH46A,, (available from Stratagene Cloning Systems, Inc.); and ptrc99a, pKK223-3, pKK233-3, pDR540, pRIT5 (available from Pharmacia Biotech, Inc). Lower eukaryotes, e.g., yeasts and Dictyostelium, may be transformed with LP protein sequence containing vectors.
  • yeasts and Dictyostelium may be transformed with LP protein sequence containing vectors.
  • yeast vectors typically consist of a replication origin (unless of the integrating type), a selection gene, a promoter, DNA encoding the desired protein or its fragments, and sequences for translation termination, polyadenylation, and transcription termination.
  • Suitable expression vectors for yeast include such constitutive promoters as 3-phosphoglycerate kinase and various other glycolytic enzyme gene promoters or such inducible promoters as the alcohol dehydrogenase 2 promoter or metallothionine promoter.
  • Suitable vectors include derivatives of the following types: self-replicating low copy number (such as the YRp-series), self-replicating high copy number (such as the YEp-series); integrating types (such as the Ylp-series), or mini-chromosomes (such as the YCp-series).
  • Additional representative yeast expression vectors include, e.g., without limit, pYES2, pYDl, pTEFl/Zeo, pYES2/GS, pPIGZ, pGAPZ, pGAPZalph, pPIC9, pPIC3.5, PHIL-D2, PHIL-SI, pPIC3.5K, pPIC9K, and PA0815 (available from Invifrogen, Carlsbad, CA).
  • Other suitable vectors will be readily apparent to the skilled artisan.
  • Higher eukaryotic tissue culture cells are typically the preferred host cells for expression of the functionally active LP protein.
  • tissue culture cell lines may be used, e.g., insect baculovirus expression systems, whether from an invertebrate or vertebrate source.
  • mammalian cells are preferred to achieve proper processing, both co-translationally and post-translationally. Transformation or transfection and propagation of such cells are routine in the art.
  • Useful cell lines include HeLa cells, Chinese hamster ovary (CHO) cell lines, baby rat kidney (BRK) cell lines, insect cell lines, bird cell lines, and monkey (COS) cell lines.
  • Expression vectors for such cell lines usually include an origin of replication, a promoter, a translation initiation site, RNA splice sites (e.g., if genomic DNA is used), a polyadenylation site, and a transcription termination site. These vectors also may contain a selection gene or amplification gene. Suitable expression vectors may be plasmids, viruses, or retroviruses carrying promoters derived, e.g., from such sources as from adenovirus, SV40, parvoviruses, vaccinia virus, or cytomegalovirus. Representative examples of suitable expression vectors include pCDNAl; pCD (Okayama, et al. (1985) Mol. Cell Biol.
  • pMClneo Poly-A (Thomas, et al. (1987) CeU 51:503- 512); and a baculovirus vector such as pAC 373 or pAC 610.
  • Additional representative eukaryotic vectors include, e.g., without limit, pWLNEO, pSV2CAT, pOG44, pXTl and pSG (available from Stratagene); and pSVK3, pBPV, pMSG and pSVL (available from Pharmacia Biotech, Inc.).
  • Introduction of the construct into a host cell can be effected by, e.g., without limit, by calcium phosphate transfection, DEAE-dextran mediated transfection, cationic lipid- mediated transfection, electroporation, transduction, infection, or other methods, (see, e.g., Davis, et al. (1986) Basic Methods in Molecular Biology). It is specifically contemplated that a polypeptide (or fragment thereof) of the present invention may in fact be expressed by a host cell lacking a recombinant vector.
  • the polypeptide can be recovered and purified from recombinant cell cultures by known methods including, e.g., without limit, ammonium sulfate or ethanol precipitation, acid extraction, anion or cation exchange chromatography, phosphocellulose chromatography, hydrophobic interaction chromatography, affinity chromatography, hydroxylapatite chromatography and pectin chromatography. Most preferably, high performance liquid chromatography (“HPLC”) is employed for purification of the polypeptide.
  • HPLC high performance liquid chromatography
  • a polypeptide (or fragment thereof) of the present invention can also be recovered from natural sources, including, e.g., without limit, bodily fluids, tissues, and cells, (whether directly isolated or cultured); products of chemical synthetic procedures; and products produced by recombinant techniques from a prokaryotic or eukaryotic host (including, e.g., bacterial, yeast, higher plant, insect, and mammalian cells). It is likely that LP proteins need not be glycosylated to elicit biological responses.
  • an LP protein or LP polypeptide in a system that provides a specific or defined glycosylation pattern.
  • the usual pattern will be that provided naturally by the expression system.
  • the pattern will be modifiable by exposing the polypeptide, e.g., in unglycosylated form, to appropriate glycosylating proteins introduced into a heterologous expression system.
  • the LP protein gene may be co-transformed with one or more genes encoding mammalian or other glycosylating enzymes. It is further understood that over glycosylation may be detrimental to LP protein biological activity, and that one of skill may perform routine testing to optimize the degree of glycosylation which confers optimal biological activity.
  • an LP polypeptide may also include, e.g., an initial modified methionine residue (in some cases because of host-mediated processes).
  • an initial modified methionine residue in some cases because of host-mediated processes.
  • the N-terminal methionine encoded by the translation initiation codon removed with high efficiency from any protein after translation in all eukaryotic cells. While the N-terminal methionine on most proteins is also efficiently removed in most prokaryotes, for some proteins depending on the nature of the amino acid to which the N-terminal methionine is covalently linked, the removal process is inefficient.
  • the yeast Pichia pastoris is used to express a polypeptide of the present invention(or fragment thereof) in an eukaryotic system.
  • Pichia pastoris is a methylotrophic yeast, which can metabolize methanol as its sole carbon source.
  • a main step in the methanol metabolization pathway is the oxidation of methanol to formaldehyde catalyzed by the alcohol oxidase.
  • Pichiu pustoris To metabolize methanol as its carbon source, Pichiu pustoris must generate high levels of alcohol oxidase due, in part, to the relatively low affinity of alcohol oxidase for O 2 . Consequently, in a growth medium using methanol as a primary carbon source, the promoter region of one of the two alcohol oxidase genes (AOX1) is highly active.
  • alcohol oxidase produced from the AOX1 gene comprises up to approximately 30% of the total soluble protein in Pichiu pastoris (see, e.g., Ellis, et al., Mol. Cell. Biol. 5:1111-21 (1985); Koutz, et al., Yeast 5: 167-77 (1989); Tschopp, et al., Nucl. Acids Res. 15:3859-76 (1987)).
  • a heterologous coding sequence such as, e.g., an LP polynucleotide sequence, (or fragment thereof) under the transcriptional regulation of all or part of the AOXl regulatory sequence is expressed at exceptionally high levels in Pichia yeast grown in the presence of methanol.
  • the plasmid vector pPIC9K is used to express polynucleotide sequence encoding a polypeptide of the invention, (or fragment thereof) as set forth herein, in a Pichea yeast system essentially as described in "Pichia Protocols: Methods in Molecular Biology," D.R. Higgins and J. Cregg, eds. The Humana Press, Totowa, NJ, 1998.
  • This expression vector allows expression and secretion of a protein of the invention by virtue of the strong AOXl promoter linked to the Pichia pastoris alkaline phosphatase (PHO) secretory signal peptide located upstream of a multiple cloning site.
  • PHO Pichia pastoris alkaline phosphatase
  • yeast vectors could be used in place of pPIC9K, such as, e.g., pYES2, pYDl, pTEFl/Zeo, pYES2/GS, pPICZ, pGAPZ, pGAPZalpha, pPIC9, pPIC3.5, PHIL-D2, PHIL- Sl, pPIC3.5K, and, PA08, as a skilled in the artisan would appreciate, as long as the proposed expression construct provides appropriately located and operably linked signals for transcription, translation, secretion (if desired), and the like, (including an in-frame stop codon as required).
  • high-level expression of a heterologous coding sequence such as, e.g., a polynucleotide sequence of the present invention
  • a heterologous coding sequence such as, e.g., a polynucleotide sequence of the present invention
  • an expression vector such as, e.g., pGAPZ or pGAPZ alpha
  • the invention also encompasses primary, secondary, and immortalized host cells of vertebrate origin, particularly mammalian origin, and more particularly, human origin, which have been engineered to delete or replace endogenous genetic material (e.g., coding sequence), and/or to include, e.g., genetic material (e.g., heterologous polynucleotide sequences) in operable linkage with a polynucleotide (or fragment thereof) of the invention, and which activate, alter, and/or amplify an endogenous polynucleotide(s).
  • endogenous genetic material e.g., coding sequence
  • genetic material e.g., heterologous polynucleotide sequences
  • known art techniques may be used to operably associate heterologous control regions (e.g., promotes and/or enhances) and an endogenous polynucleotide sequence(s) via, e.g., homologous recombination, resulting in the formation of a new transcription unit (see, e.g., U.S. Patent No. 5641,670; U.S. Patent No. 5,733,761; WO 96/29411; WO 94/12650; Roller, et al. (1989) Proc. Natl. Acad. Sci. USA 86:8932-8935; and Zijlstra, et al.
  • heterologous control regions e.g., promotes and/or enhances
  • an endogenous polynucleotide sequence(s) via, e.g., homologous recombination
  • heterologously expressed proteins or polypeptides can also be expressed in plant cells.
  • viral expression vectors e.g., cauliflower mosaic virus and tobacco mosaic virus
  • plasmid expression vectors e.g., TI plasmid
  • Such cells are available from a wide range of sources (e.g., the American Tissue Type Culture Collection, Rockland, MD; also, see for example, Ausubel, et al. (cur. ed. and Supplements; expression vehicles may be chosen from those provided e.g., in Pouwels, et al. (Cur.
  • a LP protein, or a fragment thereof may be engineered to be phosphatidyl inositol (PI) linked to a cell membrane, but can be removed from membranes by treatment with a phosphatidyl inositol cleaving enzyme, e.g., phosphatidyl inositol phospholipase-C.
  • PI phosphatidyl inositol
  • a phosphatidyl inositol cleaving enzyme e.g., phosphatidyl inositol phospholipase-C.
  • LP proteins have been characterized, fragments or derivatives thereof can be prepared by conventional processes for synthesizing peptides. These include processes such as are described in Stewart and Young (1984) Solid Phase Peptide Synthesis Pierce Chemical Co., Rockford, IL; Bodanszky and Bodanszky (1984) The Practice of Peptide Synthesis Springer-Verlag, New York, NY; and Bodanszky (1984) The Principles of Peptide Synthesis Springer-Verlag, New York, NY.
  • an azide process for example, an acid chloride process, an acid anhydride process, a mixed anhydride process, an active ester process (for example, p-nitrophenyl ester, N-hydroxysuccinimide ester, or cyanomethyl ester), a carbodiimidazole process, an oxidative-reductive process, or a dicyclohexylcarbodiimide (DCCD)/additive process
  • an azide process for example, p-nitrophenyl ester, N-hydroxysuccinimide ester, or cyanomethyl ester
  • a carbodiimidazole process for example, p-nitrophenyl ester, N-hydroxysuccinimide ester, or cyanomethyl ester
  • DCCD dicyclohexylcarbodiimide
  • Solid phase and solution phase syntheses are both applicable to the foregoing processes.
  • the prepared protein and fragments thereof can be isolated and purified from the reaction mixture by means of peptide separation, for
  • An LP protein of this invention can be obtained in varying degrees of purity depending upon its desired use. Purification can be accomplished by use of known protein purification techniques or by the use of the antibodies or binding partners herein described (e.g., in immunoabsorbant affinity chromatography). Immunoabsorbant affinity chromatography is carried out by first linking the antibodies to a solid support and then contacting the linked antibodies with solubilized lysates of appropriate source cells, lysates of other cells expressing the protein, or lysates or supernatants of cells producing the LP proteins as a result of known recombinant DNA techniques. Multiple cell lines may be screened for one cell line that expresses an LP protein (or fragment thereof) at a high level when compared to other cells.
  • Natural LP proteins can be isolated from natural sources, or by expression from a transformed cell using an appropriate expression vector. Purification of the expressed protein is achieved by standard procedures, or may be combined with engineered means for effective purification at high efficiency from cell lysates or supernatants. Epitope or other tags, e.g., FLAG or His6 segments, can be used for such purification features. VII. Physical Variants The invention also encompasses proteins or peptides having substantial amino acid sequence similarity with an amino acid sequence of an LP protein described herein. Natural variants include individual, polymorphic, allelic, strain, or species variants.
  • Amino acid sequence similarity, or sequence identity is determined by optimizing residue matches, if necessary, by introducing gaps as required. This changes when considering conservative substitutions as matches.
  • Conservative substitutions typically include substitutions within the following groups: gly cine, alanine; valine, isoleucine, leucine; aspartic acid, glutamic acid; asparagine, glutamine; serine, threonine; lysine, arginine; and phenylalanine, tyrosine.
  • Homologous amino acid sequences include natural polymorphic, allelic, and interspecies variations in each respective protein sequence.
  • Stretches of amino acids will be at least about 10 amino acids, usually about 20 amino acids, usually 50 amino acids, preferably 75 amino acids, and in particularly preferred embodiments at least about 100 amino acids. Identity can also be measures over amino acid stretches of about 98, 99, 110, 120, 130, etc. Nucleic acids encoding mammalian LP proteins will typically hybridize to the nucleic acid sequence of SEQ ID NO: X under stringent conditions.
  • nucleic acids encoding human LP proteins will normally hybridize to a nucleic acid of SEQ ID NO: X under stringent hybridization conditions (as described herein).
  • stringent conditions are selected to be about 10° C lower than the thermal melting point (Tm) for the probe sequence at a defined ionic strength and pH.
  • Tm is the temperature (under defined ionic strength and pH) at which 50% of the target sequence hybridizes to a perfectly matched probe.
  • stringent conditions will be those in which the salt concentration is about 0.2 molar at pH 7 and the temperature is at least about 50° C.
  • nucleic acids that bind to a disclosed sequence in 50% formamide and 200 mM NaCl at 42° C are useful as cloning probes, primers (e.g., a PCR primer), or diagnostic probes.
  • Hybridizing nucleic acids can be splice variants encoded by one of the LP genes described herein. Thus, the hybridizing nucleic acids may encode a polypeptide that is shorter or longer than the various forms of LP described herein.
  • Hybridizing nucleic acids may also encode proteins that are related to LP (e.g., polypeptides encoded by genes that include a portion having a relatively high degree of identity to a LP gene described herein).
  • An isolated LP protein encoding DNA can be readily modified by nucleotide substitutions, nucleotide deletions, nucleotide insertions, and short inversions of nucleotide stretches. Such modifications result in novel DNA sequences, which encode LP protein antigens, their derivatives, or proteins having highly similar physiological, immunogenic, or antigenic activity. Modified sequences can be used to produce mutant antigens or to enhance expression. Enhanced expression may involve gene amplification, increased transcription, increased translation, and other mechanisms.
  • mutant LP protein derivatives include predetermined or site-specific mutations of the respective protein or its fragments.
  • “Mutant LP protein” encompasses a polypeptide otherwise falling within the homology definition of a human LP protein as set forth herein (or in a deposited clone), but having an amino acid sequence which differs from that of a LP protein as found in nature, whether by way of deletion, substitution, or insertion.
  • site specific mutant LP protein generally includes proteins having significant similarity with a protein having a sequence of SEQ ID NO: Y, e.g., natural embodiments, and as sharing various biological activities, e.g., antigenic or immunogenic, with those sequences, and in preferred embodiments contain most or all of the disclosed sequence.
  • LP proteins proteins that are functionally related to an LP encoded by the specific sequence identifiers of the present application.
  • Functionally related proteins and polypeptides include any protein or polypeptide sharing a functional characteristic with LP of the present invention (e.g., the ability to stimulate a Janus family tyrosine kinase).
  • Such functionally related LP polypeptides include, without limitation, additions or substitutions of amino acid residues within the amino acid sequence encoded by the LP sequences described herein; particularly, those that result in a silent change, thus producing a functionally equivalent LP polypeptide.
  • Amino acid substitutions may be made based on similarity in polarity, charge, solubility, hydrophobicity, hydrophilicity, and/or the amphiphatic nature of the residues involved.
  • nonpolar (hydrophobic) amino acids include alanine, leucine, isoleucine, valine, proline, phenylalanine, tryptophan, and methionine; polar neutral amino acids include glycine, serine, threonine, cysteine, tyrosine, asparagine, and glutamine; positively charged (basic) amino acids include arginine, lysine, and histidine; and negatively charged (acidic) amino acids include aspartic acid and glutamic acid.
  • non-classical amino acids or chemical amino acid analogs may be substituted or added into an LP polypeptide sequence.
  • Non-classical amino acids include, e.g., without limitation, D-isomers of the common amino acids; 2,4-diaminobutyric acid; a-amino isobutyric acid, 4-aminobutyric acid, Abu, 2-amino butyric acid, g-Abu, e-Ahx, 6-amino hexanoic acid, Aid, 2-amino isobutyric acid, 3 -amino propionic acid, omithine, norleucine, norvaline, hydroxyproline, sarcosine, citrulline, homocitrulline, cysteic acid, t-butylglycine, t-butylalanine, phenylglycine, cyclohexylalanine, b-alanine, fluoro-amino acids, designer amino acids such as b-methyl amino acids, Ca-methyl amino acids, Na- methyl amino acids, and amino acid analogs in general.
  • the amino acid can be either dextrorotary(D ) or levorotary (L).
  • random mutations can be made to an LP nucleic acid molecule (using well known random mutagenesis techniques) and the resulting LP polypeptides can be tested for activity
  • site-directed mutations of LP coding sequences can be engineered (using well known site-directed mutagenesis techniques) to generate mutant LP with increased function (e.g. greater inhibition (or stimulation) of a kinase activity, greater resistance to degradation, increased or decreased binding affinity).
  • LP protein mutagenesis can be conducted by making amino acid insertions or deletions. Substitutions, deletions, insertions, or any combinations may be generated to arrive at a final construct.
  • Insertions include amino- or carboxyl- terminal fusions, e.g. epitope tags. Random mutagenesis can be conducted at a target codon and the expressed mutants can then be screened for the desired activity. Methods for making substitution mutations at predetermined sites in DNA having a known sequence are well known in the art (e.g., by Ml 3 primer mutagenesis or polymerase chain reaction (PCR) techniques; see also, Sambrook, et al. (cur. ed.) and Ausubel, et al. (cur. ed., and Supplements). The mutations in the DNA normally should not place coding sequences out of reading frames and preferably do not create complementary regions that could hybridize to produce secondary mRNA structure such as loops or hairpins.
  • PCR polymerase chain reaction
  • An LP polypeptide, or fragment thereof can be used to generate a fusion protein.
  • an LP polypeptide, or fragment thereof when fused to a second polypeptide, can be used as an antigenic tag or an immunogen.
  • Antibodies raised against an LP polypeptide (or fragment thereof) can be used to indirectly detect a second protein by binding thereto.
  • an LP protein has amino acid sequence portion that targets a cellular location (e.g., based on trafficking signals)
  • that portion of the polypeptide can be used by fusing it to another protein (or fragment) to target a protein.
  • domains that can be fused to an LP polypeptide include, e.g., not only heterologous signal sequences, but also other heterologous functional regions.
  • a fusion does not necessarily need to be direct, but may occur, e.g., through linker sequences.
  • fusion proteins may also be engineered to improve characteristics of an LP polypeptide. For instance, a region of additional amino acids, particularly charged amino acids, may be added to the N-terminus of the polypeptide to improve stability and persistence during purification from a host cell or during subsequent handling and storage.
  • peptide moieties can be added to the polypeptide to facilitate purification. Such regions may be removed before final preparation of the polypeptide.
  • an LP polypeptide (including any fragment thereof, and specifically an epitope) can be combined with parts of the constant domain of an immunoglobulin e.g., (IgA, IgE, IgG, IgM) portions thereof (CH 1, CH2, CH3), and any combination thereof including both entire domains and portions thereof), resulting in a chimeric polypeptide.
  • immunoglobulin e.g., (IgA, IgE, IgG, IgM) portions thereof (CH 1, CH2, CH3), and any combination thereof including both entire domains and portions thereof
  • antigens e.g., insulin
  • FcRn binding partner such as IgG or Fc fragments
  • IgG fusion proteins that have a disulfide-linked dimeric structure due to the IgG portion disulfide bonds have also been found more efficient in binding and neutralizing other molecules than monomeric polypeptides or fragments thereof alone (Fountoulakis, et al. (1995) J. Biochem. 270:3958-3964).
  • a fusion protein can comprise various portions of the constant region of an immunoglobulin molecule together with a human protein (or part thereof) EP-A-O 464 533 (Canadian counterpart 2045869).
  • the Fc part in a fusion protein is beneficial in therapy and diagnosis, and thus, can result in, e.g., improved pharmacokinetic properties (EP-A 0232262.).
  • deleting the Fc part after the fusion protein has been expressed, detected, and purified, may be desired.
  • the Fc portion may hinder therapy and/or diagnosis if the fusion protein is used as an immunogen for immunizations.
  • hIL-5 human proteins, such as hIL-5, have been fused with Fc portions for the purpose of high-throughput screening assays to identify hIL-5 antagonists (Bennett, et al. (1995) I. Molecular Recognition 8:52-58; and Johanson, et al. (1995) J. Biol. Chem. 270:9459-9471).
  • new constructs may be made by combining similar functional domains from other proteins.
  • protein-binding or other segments may be "swapped" between different new fusion polypeptides or fragments (see, e.g., Cunningham, et al. (1989) Science 243:1330-1336; and O'Dowd, et al.
  • HA hemagglutinin protein
  • Nucleic acid molecules containing LP polynucleotide sequences encoding an LP epitope can also be recombined with a gene of interest as an epitope tag (e.g., the "HA” or flag tag) to aid in detection and purification of the expressed polypeptide.
  • an epitope tag e.g., the "HA” or flag tag
  • one system purifies non-denatured fusion proteins expressed in human cell lines (Janknecht, et al. (1991) Proc. Natl. Acad. Sci.
  • a gene of interest is subcloned into a vaccinia recombination plasmid such that the open reading frame of the sequence of interest is translationally fused to an ammo-terminal tag consisting of six histidine residues.
  • the tag serves as a matrix-binding domain for the fusion protein. Extracts from cells infected with the recombinant vaccinia virus are loaded onto Ni2+ nitriloacetic acid-agarose column and histidine-tagged proteins can be selectively eluted with imidazole-containing buffers.
  • LP fusion constructions may be generated through the techniques of gene-shuffling, motif-shuffling, exon shuffling, and/or codon shuffling (collectively referred to as "DNA shuffling").
  • DNA shuffling may be employed to modulate an activity of an LP polypeptide.
  • Such methods can be used to generate LP polypeptides (or fragments thereof) with altered activity, as well as agonists and antagonists of an LP polypeptide (see, e.g., U.S. Patent Nos. 5,605,793; 5,811,238; 5,830,721; 5,834,252; and 5,837,458, and Patten, et al. (1997) Cur. Opinion Biotechnol.
  • an LP polynucleotide, or its encoded LP polypeptide or fragment thereof may be altered using random mutagenesis by error-prone PCR, random nucleotide insertion, or other methods before recombination. Similar concepts apply to heterologous nucleic acid sequences.
  • any of type fusion described herein can be easily engineered using an LP polynucleotide sequence (or fragment thereof) or an LP polypeptide (or fragment thereof) of the present invention.
  • the blocking of physiological response to an LP protein may result from the inhibition of binding of the protein to its binding partner (e.g., through competitive inhibition).
  • in vitro assays of the present invention will often use isolated protein, membranes from cells expressing a recombinant membrane associated LP protein, soluble fragments comprising binding segments of these proteins, or fragments attached to solid phase substrates. These assays also allow for the diagnostic determination of the effects either of binding segment mutations and modifications, or of protein mutations and modifications (e.g., protein analogs).
  • This invention also contemplates the use of competitive drug screening assays, e.g., where neutralizing antibodies to antigen or binding partner fragments compete with a test compound for binding to the protein.
  • the antibodies can be used to detect the presence of a polypeptide which shares one or more antigenic binding sites of the protein and can also be used to occupy binding sites on the protein that might otherwise interact with a binding partner.
  • "Derivatives" of LP protein antigens include amino acid sequence mutants, glycosylation variants, and covalent or aggregate conjugates with other chemical moieties. Covalent derivatives can be prepared by linkage of functionalities to groups which are found in LP protein amino acid side chains or at the N- or C- termini, by any art known means.
  • These derivatives can include, without limitation, aliphatic esters or amides of the carboxyl terminus, or of residues containing carboxyl side chains, O-acyl derivatives of hydroxyl group-containing residues, and N-acyl derivatives of the amino terminal amino acid or amino-group containing residues, e.g., lysine or arginine.
  • Acyl groups are selected from the group of alkyl-moieties including C3 to C18 normal alkyl, thereby forming alkanoyl aroyl species. Covalent attachment to carrier proteins may be important when immunogenic moieties are haptens.
  • Additional post- translational modifications encompassed by the invention include, e.g., without limit, N- linked, or O-linked carbohydrate chains, processing of N-terminal or C-terminal ends, attachment of chemical moieties to the amino acid backbone, chemical modifications of N-linked or O-linked carbohydrate chains, and addition or deletion of an N-terminal methionine residue, e.g., because of prokaryotic host cell expression.
  • the polypeptides or fragments thereof may also be modified with a detectable label, such as an enzymatic, fluorescent, isotopic, or affinity label to permit detection and/or isolation.
  • a chemically modified derivative of a polypeptide of the invention may provide additional advantages such as increased solubility, increased stability increased circulating time, or decreased immunogenicity or antigenicity (see U.S. Patent no: 4,179,337).
  • a chemical moieties for derivatization may be selected from water soluble polymers such as, e.g., polyethyleneglycol, ethylene glycol, propylene glycol, copolymers, carboxymethylcellulose, dextran, polyvinyl alcohol, etc.
  • a polypeptide of the invention, (or fragment thereof) may be modified at random or at predetermined positions within the molecule and may include, e.g., one, two, three, or more attached chemical moieties.
  • the polymer may be of any molecular weight, and may be branched or unbranched.
  • a preferred molecular weight is between about 1 kDa and about 100 kDa (the term "about” means that in polyethylene glycol preparations, some molecules will weigh more and some will weigh less, than the stated molecular weight).
  • Other sizes may be used, depending on the desired effect (e.g., the [period of sustained release, the effects, if any, on biological activity, ease in handling, the degree or lack of antigenicity, and other known effects of polyethylene glycol on a protein, polypeptide or an analog).
  • Polyethylene glycol molecules should be attached with consideration of the effect on functional, immunogenic, and/or antigenic domains of a polypeptide (or fragment thereof). Attachment methods include; e.g., without limit, (coupling PEG to G-CSF); EP 0 401 384, pegylating GM-CSF using tresyl chloride (Malik, et al. (1992) Exp. Hematol. 20:1028-1035).
  • polyethylene glycol may be covalently bound through amino acid residues via a reactive group, such as, e.g., a free amino or carboxyl group. Reactive groups are those to which an activated polyethylene glycol molecule may be bound.
  • Amino acid residues having a free amino group may include, e.g., lysine residues, and N-terminal amino acid residue.
  • Amino acid residues having a free carboxyl group may include, e.g., aspartic acid residues, glutamic acid residues, and C-terminal amino acid residues.
  • Sulfhydryl groups may also be used to attach to a polyethylene glycol molecule.
  • a preferred attachment is at an amino group, such as, e.g., an attachment at the N-terminus or a lysine group.
  • One may specifically desire a protein, or a polypeptide (or fragment thereof) that is chemically modified at the N-terminus.
  • 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 a protein (polypeptide) molecule in the reaction mix, the type of pegylation reaction to be performed, and the method of obtaining the selected N-terminally pegylated, e.g., polypeptide.
  • the method of obtaining an N-terminally pegylated preparation may be by purification of the N-terminally pegylated material from a population of pegylated protein molecules.
  • Selective protein chemical modification at the N-terminus 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 appropriate reaction conditions, substantially selective derivatization of a protein or polypeptide (or fragment thereof) at the N-terminus with a carbonyl-group-containing-polymer is achieved.
  • a maj or group of derivatives are covalent conjugates of an LP protein (or fragments thereof) with other proteins or polypeptides. These derivatives can be synthesized in recombinant culture such as N- or C-terminal fusions or by the use of agents known in the art for their usefulness in cross-linking proteins through reactive side groups.
  • Preferred protein derivatization sites with cross-linking agents are at free amino groups, carbohydrate moieties, and cysteine residues.
  • Fusion polypeptides between LP protein and other homologous or heterologous proteins are also provided.
  • Heterologous polypeptides may be fusions between different surface markers, resulting in, for example, a hybrid protein exhibiting binding partner specificity.
  • heterologous fusions may be constructed which would exhibit a combination of properties or activities of the derivative proteins.
  • Typical examples are fusions of a reporter polypeptide, e.g., luciferase, with a segment or domain of a protein, e.g., a segment involved in binding partner interaction, so that the presence or location of the fused protein may be easily determined (see, e.g., Dull, et al., U.S. Patent No. 4,859,609).
  • Other gene fusion partners include bacterial D-galactosidase, trpE, Protein A, ⁇ -lactamase, alpha amylase, alcohol dehydrogenase, and yeast alpha mating factor (see, e.g., Godowski, et al. (1988) Science 241 :812-816).
  • these derivatives fall into the three classes: (1) salts, (2) side chain and terminal residue covalent modifications, and (3) adsorption complexes (e.g., with cell membranes).
  • covalent or aggregative derivatives are useful as immunogens, as reagents in immunoassays, or in purification methods such as for affinity purification of proteins or other binding proteins.
  • a LP protein antigen can be immobilized by covalent bonding to a solid support such as cyanogen bromide-activated SEPHAROSE, by methods which are well known in the art, or adsorbed onto polyolefin surfaces, with or without glutaraldehyde cross-linking, for use in an assay or purification of anti-LP protein antibodies or its respective binding partner.
  • An LP protein can also be labeled for use in diagnostic assays with a detectable group (such as, e.g., radioiodinated by the chloramine T procedure; covalently bound to rare earth chelates; or conjugated to another fluorescent moiety).
  • a detectable group such as, e.g., radioiodinated by the chloramine T procedure; covalently bound to rare earth chelates; or conjugated to another fluorescent moiety.
  • Purification of an LP protein may be effected by immobilized antibodies or a binding partner.
  • Isolated LP protein genes will allow transformation of cells lacking expression of corresponding LP protein (e.g., either species types or cells that lack corresponding proteins and exhibit negative background activity). Expression of transformed genes will allow isolation of antigenically pure cell lines, with defined or single specie variants. This approach allows for detection that is more sensitive and discrimination of the physiological effects of LP binding proteins.
  • heteromeric refers to a multimer containing one or more heterologous polypeptides.
  • a multimer of the invention is a heterodimer, a heterofrimer, or a heterotetramer.
  • the heteromeric multimer of the invention is at least a heterodimer, at least a heterofrimer, or at least a heterotetramer.
  • Multimers of the invention may be the result of hydrophobic, hydrophilic, ionic and/or covalent associations and/or may be indirectly linked, by e.g., liposome formation.
  • a multimer of the invention such as, e.g., homodimers or homotrimers, are formed when polypeptides of the invention (or fragments thereof) contact one another in solution.
  • a heteromultimer of the invention such as, e.g., a heterofrimer or a heterotetramer, is formed when, e.g., a polypeptide of the invention contacts an antibody (generated against a polypeptide; or fragment thereof of the invention (including antibodies to the heterologous polypeptide sequence in a fusion protein of the invention)) in solution.
  • a multimer of the invention is formed by covalent association with and/or between a polypeptide and a binding partner such as mentioned herein (or fragment thereof).
  • covalent associations may involve one or more amino acid residues contained in a polypeptide sequence (e.g., as recited in a sequence listing herein, or contained in a polypeptide encoded by a deposited clone specified herein).
  • a covalent association is a cross-link, e.g., between cysteine residues.
  • the covalent associations are the consequence of chemical or recombinant manipulation.
  • covalent associations may involve one or more amino acid residues contained in a heterologous polypeptide sequence such as, e.g., a fusion protein of the invention.
  • covalent associations form with a heterologous sequence contained in a fusion protein of the invention (see, e.g., US Patent No. 5,478,925).
  • a covalent association is between a heterologous sequence contained in an Fc fusion protein of the invention (as described herein).
  • a covalent association of a fusion protein of the invention is with a heterologous polypeptide sequence such as, e.g., oseteoprotegerin (see, e.g., WO 98149305, incorporated by reference for these teachings).
  • a heterologous polypeptide sequence such as, e.g., oseteoprotegerin (see, e.g., WO 98149305, incorporated by reference for these teachings).
  • two or more polypeptides of the invention (or fragment thereof) are joined through peptide linkers. Examples include, e.g., peptide linkers described in U.S. Pat. No. 5,073,627 (incorporated by reference for these teachings).
  • a protein comprising multiple polypeptides of the invention that are separated by peptide linkers may be produced using conventional recombinant DNA technology.
  • Another method for preparing multimer polypeptides of the invention involves fusing a polypeptide of the invention (or fragment thereof) to a leucine zipper or an isoleucine zipper polypeptide sequence.
  • Leucine zipper and isoleucine zipper domains promote multimerization of polypeptides in which they are found.
  • Leucine zippers were originally identified in several DNA-binding proteins (Landschulz, et al., Science 240: 1759, (1988)), and have been found since in a variety of different proteins.
  • leucine zippers are naturally occurring peptides and derivatives thereof that dimerize or trimerize.
  • leucine zipper domains suitable for producing soluble, multimeric polypeptides of the invention are those described in, e.g., WO 94/10308, (incorporated by reference for these teachings).
  • Recombinant fusion proteins comprising a polypeptide of the invention (or fragment thereof) fused to a polypeptide sequence that dimerizes or trimerizes in solution can be expressed in a suitable host cell.
  • the resulting soluble multimeric fusion protein can be recovered from a supernatant using any art known technique or method described herein.
  • Trimeric polypeptides of the invention may offer an advantage of enhanced biological activity (as defined herein).
  • Preferred leucine zipper moieties and isoleucine moieties are those that preferentially form trimers.
  • polypeptides or proteins of the invention are associated by interactions with a Flag polypeptide sequence (e.g., contained in a fusion protein of the invention having a Flag sequence).
  • a protein or a polypeptide of the invention is associated by an interaction with a heterologous polypeptide sequence (contained in a Flag fusion protein of the invention) and an anti-Flag antibody.
  • a multimer of the invention may be generated using chemical art known techniques. For example, polypeptides (or fragments thereof) desired to be contained in a multimer of the invention may be chemically cross-linked using a linker molecule e.g., linker molecules and linker molecule length optimization techniques are known in the art; see, e.g., US Patent No. 5,478,925, which is incorporated by reference for such teachings.
  • a multimer of the invention may be generated using techniques known in the art to form one or more inter-molecule cross-links between the cysteine residues (see, e.g., US Patent No. 5,478,925, incorporated by reference for these teachings).
  • a polypeptide of the invention modified by the addition of cysteine or biotin to the C or N- terminus of a polypeptide can be generated by art known methods (see, e.g., US Patent No. 5,478,925, incorporated by reference for these teachings).
  • a multimer of the invention can be generated by art known methods (see, e.g., US Patent No. 5,478,925, incorporated by reference for these teachings).
  • a multimer of the invention can be generated using other commonly known genetic engineering techniques.
  • a polypeptide contained in a multimer of the invention is produced recombinantly with fusion protein technology described herein or otherwise known in the art (see, e.g., US Patent No. 5,478,925, incorporated by reference for these teachings).
  • a polynucleotide encoding a homodimer of the invention can be generated by ligating a polynucleotide sequence encoding a polypeptide (or fragment thereof) of the invention to another sequence encoding a linker polypeptide and then subsequently, further to a synthetic polynucleotide encoding the translated product of the polypeptide in the reverse orientation from the original C-terminus to the N-terminus (lacking the leader sequence) (see, e.g., US Patent No. 5,478,925, incorporated by reference for these teachings).
  • recombinant techniques described herein or otherwise known in the art can be applied to generate a recombinant polypeptide of the invention (or fragment thereof) that contains a transmembrane domain (or hyrophobic or signal peptide) and that can be incorporated by membrane reconstitution techniques into a liposome (see, e.g., US Patent No. 5,478,925, incorporated by reference for these teachings).
  • an LP polynucleotide sequence (or fragment thereof) can be used in numerous ways, e.g., such as a reagent.
  • the following descriptions are non-limiting examples of ways to use an LP polynucleotide sequence (or fragment thereof).
  • an LP polynucleotide sequence (or fragment thereof) is useful for chromosome identification. There exists an ongoing need to identify new chromosome markers, since few chromosome-marking reagents, based on actual sequence data (repeat polymorphisms), are presently available. Each polynucleotide of the present invention can therefore, be used as a chromosome marker.
  • somatic hybrids provide a rapid method of PCR mapping a polynucleotide sequence of the invention (or fragment thereof) to a particular chromosome without undue experimentation.
  • sub-localization of an LP polynucleotide sequence (or fragment thereof) can be achieved using panels of specific chromosome fragments.
  • Other gene mapping strategies that can be used include, e.g., in situ hybridization, prescreening with labeled flow-sorted chromosomes, and pre-selection by hybridization to construct chromosome specific-cDNA libraries.
  • Precise chromosomal location of a polynucleotide can also be achieved using fluorescence in situ hybridization (FISH) of a metaphase chromosomal spread.
  • FISH fluorescence in situ hybridization
  • the FISH technique can use a polynucleotide sequence of about 500-600 bases in length; however, a polynucleotide length of 2,000-4,000 bp is preferred (see, e.g., Verma, et al. (1988) "Human Chromosomes: a Manual of Basic Techniques," Pergamon Press, New York).
  • an LP polynucleotide sequence (or fragment) can be used individually (to mark a single chromosome or a single site on that chromosome) or in panels (for marking multiple sites and/or multiple chromosomes).
  • an LP polynucleotide sequence (or fragment thereof) corresponds to a non-coding region since coding sequences are more likely to be conserved within gene families, thus increasing the chance of non-specific cross hybridization during chromosome mapping.
  • Linkage analysis can be used to establish correlation between a chromosomal location and disease, syndrome, disorder or presentation of a particular condition (e.g., diseases associated with chromosomal mapping can be found, e.g., in V. McKusick, Mendelian Inheritance in Man (available on line through Johns Hopkins University
  • a cDNA precisely localized to a chromosomal region associated with a disease, syndrome, disorder or condition could be one of approximately 50-500 potential causative genes.
  • differences in a polynucleotide sequence and a corresponding gene can be examined between an affected individual and an individual unaffected by a particular disease, syndrome, disorder, or condition.
  • visible structural alterations in a chromosome such as, e.g., deletions or translocations, are examined in chromosome spreads or by using PCR. If no structural alterations are found, then the presence or absence of point mutations is determined.
  • a mutation in a sequence of interest that correlates with some or all individuals affected with a particular disease, syndrome, disorder or condition but that is not found in individuals without the disease, syndrome disorder or condition suggests that the mutation in the sequence may be the cause of the disease, syndrome, disorder or condition.
  • expression of an LP polynucleotide sequence (or fragment thereof) in an individuals as compared to another individual can be accomplished by the present invention to screen for individuals that have a particular condition, disorder, syndrome or disease state.
  • a typical alteration e.g., altered expression, chromosomal rearrangement, or mutation
  • the invention provides a method useful during diagnosis of a disease, syndrome, disorder or condition involving measuring the level of a polynucleotide mRNA, fragment, or degradation product of the present invention (or fragment thereof) in, e.g., a cell, tissue, sample, or fluid from an individual and comparing, e.g., a polynucleotide mRNA, fragment, or degradation product level with a corresponding standard level, whereby an increase or decrease in a level compared to a standard indicates or prognosticates a disease, syndromes, disorder or condition, or tendency to develop such a disease, syndromes, disorder or condition.
  • the invention encompasses a kit, e.g., for analyzing a sample for the presence of a polynucleotide associated with a proliferative disease, syndrome, disorder, or condition.
  • the kit includes, e.g., at least an LP polynucleotide sequence (or fragment thereof) probe containing a polynucleotide sequence that hybridizes with an LP polynucleotide sequence(or fragment thereof) and directions, e.g., such as for disposal.
  • a kit in another specific embodiment, includes, e.g., two polynucleotide probes defining an internal region of an LP polynucleotide sequence, where each probe has one strand containing a 31 mer-end internal to a region the polynucleotide.
  • a probe may be useful as a primer for amplification using a polymerase chain reaction (PCR).
  • the present invention is useful as a prognostic indicator, for a subject exhibiting an enhanced or diminished expression of an LP polynucleotide sequence (or fragment thereof) by comparison to a subject expressing the polynucleotide of the present invention (or fragment thereof) at a level nearer a standard level.
  • measuring level of a composition of the present invention is intended to mean herein measuring or estimating (either qualitatively and/or quantitatively) a level of, e.g., a polypeptide (or fragment thereof), or a polynucleotide (or fragment thereof) including, e.g., mRNA, DNA, or cDNA, in a first sample (e.g., preferably a biological sample) either directly (e.g., by determining or estimating an absolute protein or mRNA level) or relatively (e.g., by comparing to a polypeptide or mRNA level in a second sample).
  • a first sample e.g., preferably a biological sample
  • the level in the first sample is measured or estimated from an individual having, or suspected of having, a disease, syndrome, disorder or condition and comparing that level to a second level, wherein the second level is obtained from an individual not having and/or not being suspected of having a disease, syndrome, disorder or condition.
  • the second level is determined by averaging levels from a population of individuals not having or suspected of having a disease, syndrome, disorder, or condition.
  • a “biological sample” is intended to mean herein any sample comprising biological material obtained from, using, or employing, e.g., an organism, body fluid, exudate, lavage product, waste product, cell (or part thereof), cell line, organ, biopsy, tissue culture, or other source originating from, or associated with, a living cell, tissue, organ, or organism, which contains, e.g., a polypeptide (or fragment thereof), a protein (or fragment thereof), a mRNA (or fragment thereof), or polynucleotide sequence (or fragment thereof) of the present invention, including, e.g., without limitation, a sample such as from, e.g., hair, skin, blood, saliva, semen, vomit, synovial fluid, amniotic fluid, breast milk, lymph, pulmonary sputum, urine, fecal matter, a lavage product, etc.
  • a biological sample can include, e.g., without limitation, body fluids
  • tissue biopsies e.g., such as semen, lymph, sera, plasma, urine, synovial fluid and spinal fluid
  • a polypeptide (or fragment thereof), mRNA (or fragment thereof), a protein (or fragment thereof), or polynucleotide (or fragment thereof) of the present invention by product, or, waste product; and/or other tissue source found to express a polypeptide (or fragment thereof), mRNA (or fragment thereof), or nucleic acid (or fragment thereof), by product, or, waste product; of the present invention.
  • Methods for obtaining biological samples e.g., tissue biopsies, body fluids, cells, or waste products from mammals are known in the art.
  • a tissue biopsy is a preferred source.
  • the method(s) provided herein may preferably be applied in a diagnostic method and/or a kit in which a polynucleotide and/or an LP polypeptide (or fragment thereof) are attached to a solid support.
  • a support may be a "gene chip” or a "biological chip” as described in, e.g., US Patents 5,837,832; 5,874,219; 5,856,174; 5,700,637 and European Patent 0-373-203 (each of which is incorporated by reference herein for these teachings).
  • such a gene chip comprising an LP polynucleotide sequence(or fragment thereof) may be used, e.g., to identify polymorphisms between a polynucleotide sequence from one source, and a polynucleotide from a second, third, or multiple sources.
  • the present invention further encompasses an LP polynucleotide sequence (or fragment thereof) that is chemically synthesized, or reproduced as a peptide nucleic acid (PNA) using art known methods.
  • PNA peptide nucleic acid
  • a PNA peptide nucleic acid
  • a polynucleotide or a fragment thereof
  • a peptide nucleic acid is a polyamide type of polynucleotide analog in which, generally, e.g., the monomeric units for adenine, guanine, thymine and cytosine are available commercially (see, e.g., Perceptive Biosystems).
  • Certain components of a polynucleotide, such as DNA, like phosphorus, phosphorus oxides, or deoxyribose derivatives, are not present in a PNA.
  • PNAs bind specifically and tightly to complementary DNA strands and are not degraded by nucleases (Nielsen, et al. (1993) Nature 365: 666). In fact, a PNA binds more strongly to DNA than DNA binds to itself, probably, as there is no electrostatic repulsion between PNA/DNA; furthermore, the PNA polyamide backbone is more flexible than DNA. Because of this, PNA/DNA duplexes can bind under a wider range of stringency conditions than DNA/DNA duplexes thus, making it easier to perform multiplex hybridizations. Moreover, smaller probes can be used with PNA than with DNA due to the strong binding.
  • the present invention is also useful for detecting a cell proliferative condition, e.g., such as cancer, in a mammal.
  • the invention is useful during diagnosis of pathological cell proliferative neoplasias like, e.g., without limit: acute myelogenous leukemias including acute monocytic leukemia, acute myeloblastic leukemia, acute promyelocytic leukemia, acute myelomonocytic leukemia, acute erythroleukemia, acute megakaryocyticleukemia, and acute undifferentiated leukemia, etc.; and chronic myelogenous leukemias including chronic myelomonocytic leukemia, chronic granulocyticleukemia, etc.
  • acute myelogenous leukemias including acute monocytic leukemia, acute myeloblastic leukemia, acute promyelocytic leukemia, acute myelomonocytic leukemia, acute erythroleukemia, acute megakaryocyticleukemia, and acute undifferentiated leukemia, etc.
  • a preferred mammal includes, e.g., a primate, a monkey, a cat, a dog, a cow, a pig, a sheep, a goat, a horse, and a rabbit. Particularly preferred are human primates.
  • Pathological cell proliferative diseases, disorders, syndromes, and/or conditions are often associated with inappropriate activation of proto-oncogenes.
  • Neoplasias can result from, e.g., a qualitative alteration of a normal cellular gene product, or from a quantitative modification of nucleic acid expression by insertion of a viral sequence, by chromosomal translocation of a polynucleotide sequence to a more actively transcribed region, or by some other mechanism.
  • c-myc expression is highly amplified in the non-lymphocytic leukemia cell line HL-60.
  • HL-60 cells When HL-60 cells are chemically induced to stop proliferation, the level of c-myc is found to be down-regulated (WO91/15580).
  • exposure of HL-60 cells to a DNA construct that is complementary to the 5' end of c-myc or c-myb blocks translation of the corresponding mRNAs which down regulates expression of the c-myc or c-myb proteins and causes arrest of cell proliferation and differentiation of the treated cells (see, e.g., WO 91115580; Wickstrom, et al (1988) Proc. Natl. Acad. Sci.
  • an LP polynucleotide sequence (or fragment thereof) can be used to control polynucleotide expression through triple helix formation, or antisense DNA, or antisense RNA, in which binding of a polynucleotide sequence is to a complementary stretch of DNA or RNA.
  • the polynucleotide sequence used to contour expression is an oligonucleotide about 20-40 bases in length that is typically complementary to a target region of a polynucleotide sequence involved in transcription.
  • triple helix formation blocks RNA transcription from DNA, while antisense RNA hybridization blocks mRNA translation.
  • Either technique can be used to design antisense or triple helix polynucleotides to treat, prevent, or ameliorate a disease or condition associated with cell proliferation when coupled with the sequence information disclosed herein, (see, e.g., J. Okano, (1991) Neurochem. 56:560;"Oligodeoxynucleotides as Antisense Inhibitors of Gene Expression CRC Press, Boca Raton, FL (1988); Lee, et al. (1979) Nucleic Acids Research 6: 3073; Cooney, et al. (1988) Science 241 : 456; and Dervan, et al. (1991) Science 251:1360).
  • An LP polynucleotide sequence (or fragment thereof) is also useful in polynucleotide delivery.
  • One goal of polynucleotide delivery is to insert a polynucleotide sequence into an organism so that it is stably expressed.
  • Polynucleotides of the invention (or fragments thereof) offer a means, e.g., of targeting a genetic defect in a highly accurate manner.
  • Another goal is to insert a polynucleotide sequence that is not normally present in a host genome.
  • An LP polynucleotide sequence is also useful to identify an individual from a sample such as, e.g., a biological sample.
  • RFLP restriction fragment length polymorphism
  • a polynucleotide sequence (or fragment thereof) of the present invention can also be used as an alternative to RFLP, by determining the actual base-by-base DNA sequence of selected portions of an individual's genome. Such polynucleotide sequences can be used to prepare PCR primers for amplifying and isolating selected portions of DNA, which can then be sequenced. Using a RFLP technique, an individual can be identified because each individual has a unique set of DNA sequences. Once an ID database is established for a particular individual, then a positive identification of a biological sample from that individual (living or dead) can be made. Forensic techniques can also benefit from using a DNA-based identification technique as disclosed herein.
  • a polymerase chain reactor e.g., blood, saliva, semen, synovial fluid, amniotic fluid, breast milk, lymph, pulmonary sputum or surfactant, urine
  • fecal matter e.g., fecal matter, etc.
  • polymorphic loci such as a DQa class II HLA gene
  • An appropriate reagent can comprise, e.g., a DNA probe, or primer that is specific to a particular tissue, which is prepared from a polynucleotide sequence of the present invention. Panels of such reagents can then be used to identify tissue by, e.g., species and/or by tissue or organ type. In a similar fashion, such reagents can be used to screen tissue cultures for contamination by, e.g., a non-specific tissue. Furthermore, an LP polynucleotide sequence can be used to create a unique polynucleotide sequence identifier, which can be placed in a material that needs future verification or authentication, e.g., in clothing, explosives, food stuffs, seed lots, etc.
  • a reliable, duplication-proof means of authenticating goods is needed as counterfeit goods in the United States amount to approximately $200 billion a year.
  • a unique sequence can be amplified, e.g., using PCR techniques, to supply sufficient quantities of the unique sequence identifier so that it can be embedded in a material for future identification, validation, and/or authentication.
  • an ink or similar marker can be laced with a unique DNA sequence(s) to insure authenticity and to identify counterfeiting in areas such as, e.g., pharmaceuticals or cosmetics, fine arts, sports collectibles, or to secure documents and financial instruments, including, e.g., passports, currency, and ID cards (see, e.g., DNA Technologies of Los Angeles, USA).
  • an LP polynucleotide sequence (or fragment thereof) can be used as a molecular weight marker e.g., on a Southern gel, as a diagnostic probe for the presence of a specific mRNA in a particular cell type, as a probe to "subtract-out" known sequences in the process of discovering novel polynucleotides, for selecting and making oligomers for attachment to a "gene chip” or other support, as an immunogen, e.g., to raise anti-DNA antibodies using DNA immunization techniques, or as an antigen to elicit an immune response.
  • an immunogen e.g., to raise anti-DNA antibodies using DNA immunization techniques, or as an antigen to elicit an immune response.
  • LP polypeptide can be used in numerous ways. The following descriptions are non-limiting, exemplars that use art known techniques.
  • a polypeptide (or fragment thereof) can be used to assay a protein level, e.g., of a secreted protein, in a sample, e.g., such as a bodily fluid by using antibody-based techniques.
  • protein expression in a tissue can be studied by an immunohistological method (see, e.g., Jalkanen, et al. (1985) J. Cell Biol. 101:976-985; Jalkanen, et al. (1987) J. Cell Biol. 105:3087-303096).
  • Another useful antibody-based method for detecting protein or polypeptide expression includes, e.g., an immunoassay like an enzyme linked immunosorbent assay or a radioimmunoassay (RIA).
  • Suitable labels for an antibody assay are known in the art and include without limit, e.g., enzyme labels, such as e.g., glucose oxidase, and radioisotopes, such as, e.g., iodine ( 125 1, 13 1), carbon ( 14 C), sulfur ( 35 S), tritium ( 3 H), indium ( 112 In), and technetium ( 99 Tc); and fluorescent labels, such as, e.g., fluorescein, rhodamine, or biotin.
  • enzyme labels such as e.g., glucose oxidase
  • radioisotopes such as, e.g., iodine ( 125 1, 13 1), carbon ( 14 C), sulfur ( 35 S), tritium ( 3 H), indium (
  • a protein in addition to assaying, e.g., the level of a secreted protein in a sample, a protein can also be detected by in vivo imaging.
  • Antibody labels or markers for in vivo imaging of a protein (or polypeptide) include, e.g., those detectable by X-radiography, NMR or ESR.
  • a suitable label for X-radiography includes, e.g., a radioisotope such as barium or cesium, which emits detectable radiation but is not detrimental to a subject.
  • Suitable markers for NMR and ESR include, e.g., those with a detectable characteristic spin, such as, e.g., deuterium, which may be incorporated into an antibody by labeling, e.g., the nutrients of a particular hybridoma.
  • a protein-specific antibody or antibody fragment that has been labeled with an appropriate detectable imaging moiety such as a radioisotope (e.g., 131 1, 112 In, 99 Tc), a radio-opaque substance, or a material detectable by nuclear magnetic resonance, can be introduced into a subject (e.g., parenterally, subcutaneously, or intraperitoneally).
  • the subject's size and the imaging system used will both effect the amount of an imaging moiety that is needed to produce a diagnostic image.
  • the quantity of the imaging moiety ranges from about 5 to 20 millicuries of label, e.g., 99 Tc.
  • a labeled antibody or antibody fragment preferentially accumulates at the location of cells that contain the targeted protein or polypeptide (see, e.g., Burchiel, et al. (1982) "l munopharmacokinetics of Radiolabeled Antibodies and Their Fragments.” (Chapter 13 in Tumor Imaging: The Radiochemical Detection of Cancer, S.W. Burchiel and B. A.
  • the invention provides a means for detecting, marking, locating or diagnosing a disease, syndrome, syndrome, disorder, and/or condition comprising assaying the expression of a polynucleotide (or fragment thereof), or a polypeptide (or fragment thereof), of the present invention that is in a sample, e.g., cells or body fluid of an individual by comparing one level of expression with another level of expression, e.g., a standard level of expression to indicate, e.g., a disease, syndrome, disorder, and/or condition, (or predilection to the same), or to make a prognosis or prediction.
  • a polynucleotide or fragment thereof
  • a polypeptide (or fragment thereof) of the present invention that is in a sample, e.g., cells or body fluid of an individual by comparing one level of expression with another level of expression, e.g., a standard level of expression to indicate, e.g., a disease, syndrome, disorder, and/or condition,
  • a cell proliferation condition e.g., such as cancer
  • the presence of a high level of expression in a sample relative to another lower level or lower standard level may indicate a predisposition for development of a disease, syndrome, or it may provide a means for condition, or state detecting a pre-clinical disease, condition, syndrome, state, or disorder before the appearance of clinical symptoms by other means.
  • Such a use may be beneficial by allowing early intervention thereby preventing and/or ameliorating the development or further progression of the condition.
  • an LP polypeptide (or fragment thereof) can be used to treat, prevent, modulate, ameliorate, and/or diagnose a disease, syndrome, condition, and/or a disorder.
  • a subject can be administered a polypeptide (or fragment thereof) of the invention to replace absent or decreased levels of a polynucleotide or polypeptide (e.g., insulin); to supplement absent or decreased levels of a different polynucleotide or polypeptide (e.g., hemoglobin S for hemoglobin B; SOD to catalyze DNA repair proteins); to inhibit the activity of a polynucleotide or polypeptide (e.g., an oncogene or tumor suppressor); to activate a polynucleotide or polypeptide (e.g., by binding to a receptor), to reduce activity of a membrane bound receptor by competing with the receptor for free ligand (e.g., soluble TNF receptors can be used to reduce inflammation), or to bring about a desired response (e.g., blood vessel growth inhibition, enhancement of an immune response to proliferating cells or to an infectious agent).
  • a polynucleotide or polypeptide e.g., insulin
  • an antibody directed to a polypeptide (or fragment thereof) of the present invention can also be used to treat, prevent, modulate, ameliorate, and/or diagnose a condition, syndrome, state, disease or disorder.
  • administration of an antibody directed to an LP polypeptide (or fragment thereof) can bind and reduce the level of the targeted polypeptide.
  • administration of an antibody can activate an LP polypeptide (or fragment thereof), such as by binding to the polypeptide that is bound to a membrane (e.g., a receptor).
  • Polypeptides of the present invention can also be used as molecular weight markers on SDS-PAGE gels or on molecular sieve gel filtration columns using methods known to those of skill in the art.
  • kits and assay methods which are capable of screening compounds for binding activity to the proteins.
  • Several methods of automating assays have been developed in recent years to permit screening of tens of thousands of -Ill- compounds in a short period. See, e.g., Fodor, et al. (1991) Science 251:161-113, and other descriptions of chemical diversity libraries, which describe means for testing of binding affinity by a plurality of compounds.
  • the development of suitable assays can be greatly facilitated by the availability of large amounts of purified, soluble LP protein as provided by this invention. For example, antagonists can normally be found once the protein has been structurally defined.
  • the advantages of using a recombinant protein in screening for specific binding partners include: (a) improved renewable source of the LP protein from a specific source; (b) potentially greater number of binding partners per cell giving better signal to noise ratio in assays; and (c) species variant specificity (theoretically giving greater biological and disease specificity).
  • One method of drug screening uses eukaryotic or prokaryotic host cells, which are stably transformed with recombinant DNA molecules expressing a LP protein-binding counterpart. Cells may be isolated which express a binding counterpart in isolation from any others. Such cells, either in viable or fixed form, can be used for standard protein binding assays. See also, Parce, et al.
  • any one of numerous techniques can be used to separate bound from free protein to assess the degree of protein binding.
  • This separation step could typically involve a procedure such as adhesion to filters followed by washing, adhesion to plastic followed by washing, or centrifugation of the cell membranes.
  • Viable cells could also be used to screen for the effects of drugs on LP protein mediated functions, e.g., second messenger levels, i.e., cell proliferation; inositol phosphate pool changes, transcription using a luciferase-type assay; and others.
  • Some detection methods allow for elimination of a separation step, e.g., a proximity-sensitive, detection system.
  • Another method uses membranes from transformed eukaryotic or prokaryotic host cells as the source of a LP protein.
  • LP protein e.g., an engineered membrane bound form.
  • the membranes would be prepared from the cells and used in a protein- binding assay such as the competitive assay set forth above.
  • solubilized, unpurified or solubilized, purified LP protein from transformed eukaryotic or prokaryotic host cells. This allows for a "molecular" binding assay with the advantages of increased specificity, the ability to automate, and high drug test throughput.
  • Another technique for drug screening involves an approach which provides high throughput screening for compounds having suitable binding affinity to a LP protein antibody and is described in detail in Geysen, European Patent Application 84/03564, published on September 13, 1984.
  • a solid substrate e.g., plastic pins or some other appropriate surface, see Fodor, et al., supra.
  • all the pins are reacted with solubilized- unpurified or solubilized-purif ⁇ ed LP protein antibody, and washed.
  • the next step involves detecting bound LP protein antibody.
  • Rational drug design may also be based upon structural studies of the molecular shapes of the LP protein and other effectors or analogs.
  • Effectors may be other proteins that mediate other functions in response to protein binding, or other proteins that normally interact with the binding partner.
  • One means for determining which sites interact with specific other proteins is a physical structure determination, e.g., x-ray crystallography or 2 dimensional NMR techniques. These will provide guidance as to which amino acid residues form molecular contact regions.
  • a purified LP protein can be coated directly onto plates for use in the aforementioned drug screening techniques.
  • non-neutralizing antibodies to these proteins can be used as capture antibodies to immobilize the respective protein on the solid phase.
  • At least one and up to a plurality of test compounds such as, e.g., antibodies, oligonucleotides, proteins (e.g., receptors), or small molecules
  • LP polypeptide or fragment thereof
  • the identified compound is closely related to the natural ligand of LP, e.g., a ligand or fragment thereof, a natural substrate, a structural or functional mimetic, or a natural binding partner.
  • the compound can be closely related to, e.g., a natural receptor to which LP (or fragment thereof) binds, or to at least a fragment of the receptor, e.g.,. the ligand binding site. In either case, the compound can be rationally designed using known techniques.
  • An assay may test binding of a test compound to the polypeptide, wherein binding is detected by, e.g., a fluorophore, radioisotope, enzyme conjugate, or other detectable label.
  • the assay may comprise combining at least one test compound with an LP polypeptide (or fragment thereof), either in solution or affixed to a solid support, and detecting binding of LP to the compound.
  • the assay may detect or measure binding of a test compound in the presence of a labeled competitor.
  • the assay may be carried out using cell-free preparations, chemical libraries, or natural product mixtures, and the test compound(s) may be free in solution or affixed to a solid support.
  • An LP polypeptide (or fragment thereof) may also be used to screen for compounds that modulate the activity of an LP polypeptide (or fragment thereof). Such compounds may include agonists, antagonists, or partial or inverse agonists.
  • an assay is performed under conditions permissive for an LP polypeptide (or fragment thereof) activity, wherein an LP polypeptide (or fragment thereof) is combined with at least one test compound, and the activity of an LP polypeptide (or fragment thereof) in the presence of a test compound is compared with the activity of LP in the absence of the test compound. A change in the activity of LP in the presence of the test compound is indicative of a compound that modulates the activity of LP.
  • a test compound is combined with an in vitro or cell-free system comprising an LP polypeptide (or fragment thereof) under conditions suitable for LP activity, and the assay is performed.
  • a test compound which modulates an activity of an LP polypeptide (or fragment thereof) may do so indirectly and need not come in direct contact with the test compound. At least one and up to a plurality of test compounds may be screened. In addition, one could identify a molecule that binds an LP polypeptide (or fragment thereof) by using a beta-pleated sheet region(s) contained in the LP sequence. Accordingly, a specific embodiment of the invention is directed to an LP polynucleotide sequence encoding the corresponding polypeptide comprising, or alternatively consisting of, an amino acid sequence of a beta pleated sheet region in a disclosed polypeptide sequence of the invention.
  • This invention also provides reagents with significant therapeutic value.
  • An LP protein or polypeptide (naturally occurring or recombinant), fragments thereof, and antibodies thereto, along with compounds identified as having binding affinity to an LP, are useful in the treatment of conditions associated with abnormal physiology or development, including abnormal proliferation, e.g., cancerous conditions, or degenerative conditions. Abnormal proliferation, regeneration, degeneration, and atrophy may be modulated by appropriate therapeutic treatment using a composition(s) provided herein.
  • a disease or disorder associated with abnormal expression or abnormal signaling by a LP protein is a target for an agonist or antagonist of the protein.
  • Another therapeutic approach included within the invention involves direct administration of reagents, formulations, or compositions by any conventional administration techniques (such as, e.g., without limit, local injection, inhalation, or systemic administration) to a subject.
  • the reagents, formulations, or compositions included within the bounds and metes of the invention may also be targeted to a cell by any of the methods described herein (e.g., polynucleotide delivery techniques).
  • a dosage is typically 0.1 mg/kg to 100 mg/kg of a recipients body weight.
  • a dosage is between 0.1 mg kg and 20 mg/kg of a recipients body weight, more preferably 1 mg/kg to 10 mg/kg of a recipients body weight.
  • homo-specific antibodies have a longer half-life than hetero-specific antibodies, (e.g., human antibodies last longer within a human host than antibodies from another species, e.g., such as a mouse, probably, due to the immune response of the host to the foreign composition).
  • hetero-specific antibodies e.g., human antibodies last longer within a human host than antibodies from another species, e.g., such as a mouse, probably, due to the immune response of the host to the foreign composition.
  • 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) by using modifications such as, e.g., lipidation.
  • the invention also provides a pharmaceutical pack or kit comprising one or more containers filled with one or more of the ingredients of the compositions of the invention and instructions such as, e.g., for disposal (typically, in a form prescribed by a governmental agency regulating the manufacture, use or sale of pharmaceuticals or biological products).
  • a pharmaceutical pack or kit comprising one or more containers filled with one or more of the ingredients of the compositions of the invention and instructions such as, e.g., for disposal (typically, in a form prescribed by a governmental agency regulating the manufacture, use or sale of pharmaceuticals or biological products).
  • the quantities of reagents necessary for effective treatment will depend upon many different factors, including means of administration, target site, physiological state of the patient, and other medicaments administered. Thus, treatment dosages should be titrated to optimize safety and efficacy. Typically, dosages used in vitro may provide useful guidance in the amounts useful for in situ administration of these reagents. Animal testing of effective doses for treatment of particular disorders will provide further predictive indication of human dosage.
  • Formulations typically comprise at least one active ingredient, as defined above, together with one or more acceptable carriers thereof. Each carrier should be both pharmaceutically and physiologically acceptable in the sense of being compatible with the other ingredients and not injurious to the patient.
  • Formulations include those suitable for oral, rectal, nasal, or parenteral (including subcutaneous, intramuscular, intravenous and intradermal) administration.
  • the formulations may conveniently be presented in unit dosage form and may be prepared by any methods well known in the art of pharmacy. See, e.g., Gilman, et al.
  • the present invention also provides a pharmaceutical composition.
  • a composition comprises, e.g., a therapeutically effective amount of a composition of the invention in a pharmaceutically acceptable carrier.
  • pharmaceutically acceptable carrier means a carrier approved by a federal regulatory agency of the United States of America, or a regulatory/administrative agency of a state government of the United States or a carrier that is listed in the U.S. Pharmacopeia or other pharmacopeia; which is generally recognized by those in the art for use in an animal, e.g., a mammal, and, more particularly, in a primate, e.g., a human primate.
  • carrier refers to a diluent, adjuvant, excipient, or vehicle that is administered with a composition of the invention.
  • a pharmaceutical carrier typically can be a sterile liquid, such as water or oils, (including those of petroleum, animal, vegetable, or synthetic origin, e.g., such as peanut oil, soybean oil, mineral oil, sesame oil and the like).
  • sterile water is a preferred carrier when a 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, e.g., without limit, starch, glucose, lactose, sucrose, gelatin, malt, rice, flour, chalk, silica gel, sodium stearate, glycerol monostearate, talc, sodium chloride, dried skim milk, glycerol, propylene, glycol, water, ethanol and the like.
  • a composition of the invention can also contain minor amounts of wetting or emulsifying agents, or pH buffering agents.
  • a composition of the invention can be in a solution, suspension, emulsion, tablet, pill, capsule, powder, sustained-release formulation, etc., or it can be formulated as a suppository (with traditional binders, and/or carriers, e.g., such as triglycerides).
  • Oral formulations encompassed include, e.g., without limit, standard carriers such as pharmaceutical grades of mannitol, lactose, starch, magnesium stearate, sodium saccharine, cellulose, magnesium carbonate, etc. Additional examples of suitable pharmaceutical carriers are described in the current edition of "Remington's Pharmaceutical Sciences" by E.W. Martin.
  • compositions for intravenous administration will contain a therapeutically effective amount of a composition of the invention, preferably in purified form, together with a suitable amount of carrier to provide for proper administration to a subject.
  • a formulation will suit the mode of administration.
  • a composition is formulated in accordance with routine procedures as a pharmaceutical composition adapted for intravenous administration to, e.g., a human.
  • compositions for intravenous administration are solutions in sterile isotonic aqueous buffer.
  • the composition may also include, e.g., a solubilizing agent and a local anesthetic such as lidocaine to promote comfort at the injection site.
  • ingredients are supplied either separately or mixed together in unit dosage form, e.g., as a dry lyophilized powder or water free concentrate in a hermetically sealed container (such as an ampoule or sachet indicating the quantity of active agent).
  • a composition is to be administered by infusion, it can be dispensed using an infusion bottle containing sterile pharmaceutical grade water or saline.
  • an ampoule of sterile water for injection or saline can be provided so that the ingredients may be mixed before administration.
  • Compositions of the invention can be formulated as neutral or salt forms.
  • Pharmaceutically acceptable salts include, e.g., without limit, anionic salts (such as those derived from hydrochloric, phosphoric, acetic, oxalic, tartaric acids, etc.,) and cationic salts, (e.g., such as those derived from sodium, potassium, ammonium, calcium, ferric hydroxides, isopropylamine, triethylamine, 2-ethylamino ethanol, histidine, procaine, etc).
  • anionic salts such as those derived from hydrochloric, phosphoric, acetic, oxalic, tartaric acids, etc.
  • cationic salts e.g., such as those derived from sodium, potassium, ammonium, calcium, ferric hydroxides, isopropylamine, triethylamine, 2-ethylamino ethanol, histidine, procaine, etc).
  • compositions of the invention that will be effective in the modulation treatment, inhibition, amelioration, or prevention of a disease, syndrome, condition, or disorder associated with aberrant expression and/or activity of a polypeptide (or fragment thereof), or a polynucleotide (or fragment thereof) of the invention can be determined without undue experimentation by the ordinary artisan using standard clinical techniques.
  • in vitro assays may optionally be employed to help identify optimal dosage ranges. Dosage requirements in a circumstance typically will depend on, e.g., the route of administration, the seriousness of the disease, condition, syndrome, or disorder; and the judgment of the practitioner or clinician.
  • compositions of the invention by any conventional administration technique (such as, e.g., without limit, local injection, inhalation, or systemic a ⁇ jninistration), to a subject with e.g., an infectious, a microbial, a bacterial, a viral or a fungal condition.
  • a composition or formulation may also be targeted to a specific cell or a receptor by any method described herein or known in the art.
  • LP FVIIa truncated FVIIa
  • FVIIa-TF pathway has been shown to induce several genes that mediate angiogenesis.
  • FVIIa induces IL-8 expression in macrophage and fibroblasts ( Ner, E., Gjemes, E., Wiiger, M., Pringel S. and Prydz, H. (2000) J. Biol. Chem. 275, 6580-6585.).
  • IL-8 is a chemoattractant for endothelial cells and monocyte, and promotes angiogenesis in vivo (Radcliffe, R., and Nemerson, Y. (1976) J. Biol. Chem. 251, 4797-4802; Szekanecz, Z., Shah, M. R., Harlow, L. A., Pearce, W. H., Koch, A. E.
  • FVIIa also induces the expression of the CCN gene family members CYR61 and connective tissue growth factor (CTGF) (NCr, E., Gjernes, E., Wiiger, M., Pringel S. and Prydz, H. (2000) J. Biol. Chem. 275, 6580-6585).
  • CGF connective tissue growth factor
  • the cystein rich protein CYR61 acts as a ligand for the integrin alpha v beta 3 (Leu, Shr-Jeng., Lam, S. C, Lai, L. F. (2002) J. Boil. Chem. 277, 46248-46255).
  • CYR61 stimulates endothelial cell growth and migration, and in vivo angiogenesis (Kireeva, M, L., Mo, F. E., Yang, G. P., Lau, L. F. 9 1996). Mol Cell Biol 16, 1326-1334; Babic, A. M., Kireeva, K. L., Kolesnikova, T. V., and Lau, L. F. (1998) Proc. Natl. Acad. Sci. U.S.A. 95, 6355-6360). CYR 61 gene knock out mice show gross vascular defects (Mo, F.-E., Mutean, A. G., Chen, C.-C, Stolz, D. B., Watkins, S C, and Lau, L. F.
  • FVIIa induced angiogenic genes include VEGF and FGF-5 (Ollivier, V., Chabbat. J., Herbert. J. M., Hakim, J., and de Prost. D. (2000). Thromb. Vase. Biol. 20, 1374-1381; skyr, E., Gjemes, E., Wiiger, M., Pringel S. and Prydz, H. (2000) J. Biol. Chem. 275, 6580-6585). Although FVIIa may induce angiognesis, TF-VIIa activation in vivo is usually associated with the induction of coagulation and platelet activation.
  • an LP can be used to modulate hemostatic or thrombolytic activity.
  • increasing hemostatic or thrombolytic activity can treat or prevent a blood coagulation condition such as e.g., afibrinogenemia, a factor deficiency, a blood platelet disease (e.g. thrombocytopenia), or a wound resulting from e.g., trauma, surgery, early wound repair as well as hemostasis, profuse bleeding, and patients with impaired liver function, etc.
  • compositions of the invention can be used to decrease hemostatic or thrombolytic activity or to inhibit or dissolve a clotting condition.
  • Such compositions can be important in a treatment or prevention of a heart condition, e.g., an attack infarction, stroke, or mycardial scarring.
  • An LP may also be useful in ameliorating, treating, preventing, modulating and/or diagnosing an autoimmune disease, disorder, syndrome, and/or condition such as results, e.g., from the inappropriate recognition by a cell of the immune system of the self as a foreign material.
  • an inappropriate recognition results in an immune response leading to detrimental effect destruction on the host, e.g., on a host cell, tissue, protein, or moiety, e.g., a carbohydrate side chain. Therefore, administration of an LP which inhibits a detrimental immune response, particularly, e.g., a proliferation, differentiation, or chemotaxis of a T-cell, may be effective in detecting, diagnosing, ameliorating, or preventing such an autoimmune disease, disorder, syndrome, and/or condition.
  • a detrimental immune response particularly, e.g., a proliferation, differentiation, or chemotaxis of a T-cell
  • autoimmune conditions examples include, e.g., without limit Addison's Disease syndrome hemolytic anemia, anti-phospholipid syndrome, rheumatoid arthritis, dermatitis, allergic encephalomyelitis, glomerulonephritis, Goodpasture's Syndrome, Graves' Disease syndrome, Multiple Sclerosis, Myasthenia Gravis, Neuritis, Ophthalmia, BuUous Pemphigoid, Pemphigus, Polyendocrinopathies, Purpura, Reiter's Disease syndrome, Stiff-Man Syndrome, Autoimmune Thyroiditis, Systemic Lupus Erythematosus, Autoimmune Pulmonary Inflammation, Guillain-BarreSyndrome, insulin dependent diabetes mellitis, and autoimmune inflammatory eye disease.
  • Addison's Disease syndrome hemolytic anemia e.g., without limit Addison's Disease syndrome hemolytic anemia, anti-phospholipid syndrome, rheumatoid arthritis, dermatiti
  • allergic reactions and conditions such as asthma (e.g., allergic asthma) or other respiratory problems, may also be ameliorated, treated, modulated or prevented, and/or diagnosed by an LP polynucleotide or polypeptide (or fragment thereof), or an agonist or antagonist thereto.
  • inventive compositions can be used to effect, e.g., anaphylaxis, hypersensitivity to an antigenic molecule, or blood group incompatibility.
  • An LP may also be used to modulate, ameliorate, treat, prevent, and/or diagnose organ rejection or graft- versus-host disease (GVHD).
  • organ rejection occurs by a host's, immune-cell destruction of a transplanted tissue or cell.
  • a similarly destructive immune response is involved in GVHD, however, in this case, transplanted foreign immune cells destroy host tissues and/or cells.
  • Administration of a composition of the invention which ameliorates or modulates such a deleterious immune response (e.g., a deleterious proliferation, differentiation, or chemotaxis of a T cell), can be effective in modulating, ameliorating, diagnosing, and/or preventing organ rejection or GVHD.
  • an LP may also be used to detect, treat, modulate, ameliorate, prevent, and/or diagnose an inflammation, e.g., by inhibiting the proliferation and/or differentiation of a cell involved in an inflammatory response, or an inflammatory condition (either chronic or acute), including, e.g., without limitation, chronic prostatitis, granulomatous prostatitis and malacoplakia, an inflammation associated with an infection (such as, e.g., septic shock, sepsis, or systemic inflammatory response syndrome (SIRS)), ischemia-reperfusion injury, endotoxin lethality, arthritis, complement-mediated hyperacute rejection, nephritis, cytokine or chemokine induced lung injury, inflammatory bowel disease syndrome, Crohn's disease syndrome, or a condition resulting from an over production of a cytokine(s) (e.g., TNF or IL-1 .)
  • a cytokine(s) e.g.,
  • angiogenesis is stringently regulated, and delimited spatially and temporally.
  • pathological angiogenesis such as, e.g., during solid tumor formation, these regulatory controls fail and unregulated angiogenesis can become pathologic by sustaining progression of many neoplastic and non-neoplastic diseases.
  • a number of serious diseases are dominated by abnormal neovascularization (including, e.g., solid tumor growth and metastases, arthritis, some types of eye conditions, and psoriasis; see, e.g., reviews by Moses, et al., Biotech. 9630-634 (1991); Folkman, et al., N. Engl. J. Med., 333: 1757-1763 (1995); Auerbach, et al., J. Microvasc. Res. 29:401-4 11 (1985); Folkman, "Advances in Cancer Research", eds. Klein and Weinhouse, Academic Press, New York, pp. 175-203 (1985); Patz, Am. J. Opthalmol.
  • angiogenesis contributes to a disease- state, e.g., for example, significant data have accumulated suggesting that solid tumor formation is dependent on angiogenesis (see, e.g., Folkman and Klagsbrun, Science 235:442-447 (1987)).
  • administration of an LP provides for the treatment, amelioration, modulation, diagnosis, and/or inhibition of a disease, disorder, syndrome, and/or condition associated with neovascularization.
  • Malignant and metastatic conditions that can be effected in a desired fashion using an LP include, e.g., without limitation, a malignancy, solid tumor, and a cancer as described herein or as otherwise known in the art (for a review of such disorders, syndromes, etc. see, e.g., Fishman, et al., Medicine, 2d Ed., J. B. Lippincott Co., Philadelphia (1985)).
  • the present invention provides a method of ameliorating, modulating, treating, preventing, and/or diagnosing an angiogenesis-related disease and/or disorder, comprising administering to a subject in need thereof a beneficially effective amount of an LP.
  • cancers that may be so affected using a composition of the invention includes, e.g., without limit a solid tumor, including e.g., 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 metastases; melanomas; glioblastoma; Kaposi's sarcoma; leiomyosarcoma; non-small cell lung cancer; colorectal cancer; advanced malignancies; and blood bom tumors such as e.g., leukemia.
  • a solid tumor including e.g., prostate, lung, breast, ovarian, stomach, pancreas, larynx, esophagus, testes, liver, parotid, biliary tract, colon, rectum, cer
  • kits This invention also contemplates use of LP proteins, fragments thereof, peptides, and their fusion products in a variety of diagnostic kits and methods for detecting the presence of LP protein or a binding partner.
  • the kit will have a compartment containing either a defined LP protein peptide or gene segment or a reagent, which recognizes one or the other, e.g., binding partner fragments or antibodies.
  • a kit for determining the binding affinity of a test compound to a LP protein would typically comprise a test compound; a labeled compound, e.g., a binding agent or antibody having known binding affinity for the LP protein; a source of LP protein (naturally occurring or recombinant); and a means for separating bound from free labeled compound, such as a solid phase for immobilizing the LP protein.
  • a labeled compound e.g., a binding agent or antibody having known binding affinity for the LP protein
  • a source of LP protein naturally occurring or recombinant
  • a means for separating bound from free labeled compound such as a solid phase for immobilizing the LP protein.
  • a preferred kit for determining the concentration of, e.g., a LP protein in a sample would typically comprise a labeled compound, e.g., binding partner or antibody, having known binding affinity for the LP protein, a source of LP protein (naturally occurring or recombinant), and a means for separating the bound from free labeled compound, for example, a solid phase for immobilizing the LP protein. Compartments containing reagents, and instructions, will normally be provided. Antibodies, including antigen binding fragments, specific for an LP protein or fragments thereof are useful in diagnostic applications to detect the presence of elevated levels of LP protein and or its fragments.
  • diagnostic assays can employ lysates, live cells, fixed cells, immunofluorescence, cell cultures, body fluids, and further can involve the detection of antigens related to the protein in serum, or the like. Diagnostic assays may be homogeneous (without a separation step between free reagent and antigen-LP or - WDS protein complex) or heterogeneous (with a separation step).
  • Various commercial assays exist, such as radioimmunoassay (RIA), enzyme-linked immunosorbent assay (ELISA), enzyme immunoassay (EIA), enzyme-multiplied immunoassay technique
  • EMIT substrate-labeled fluorescent immunoassay
  • SFIA substrate-labeled fluorescent immunoassay
  • unlabeled antibodies are employed by using a second antibody which is labeled and which recognizes an antibody to a LP protein or to a particular fragment thereof.
  • Similar assays are also extensively discussed in the literature (see, e.g., Harlow and Lane (1988) Antibodies: A Laboratory Manual. CSH Press, NY; Chan (ed.) (1987) Immunoassay: A Practical Guide Academic Press, Orlando, FL; Price and Newman (eds.) (1991) Principles and Practice of Immunoassay Stockton Press, NY; and Ngo (ed.) (1988) Nonisotopic Immunoassay Plenum Press, NY).
  • Anti-idiotypic antibodies may have similar use to diagnose the presence of antibodies against an LP protein or polypeptide, as such may be diagnostic of various abnormal states, conditions, disorders, or syndromes.
  • overproduction of LP protein may result in production of various immunological or other medical reactions which may be diagnostic of abnormal physiological states, e.g., in cell growth, activation, or differentiation.
  • the reagents for diagnostic assays are supplied in kits, to optimize the sensitivity of the assay.
  • the protocol, and the label either labeled or unlabeled antibody or binding partner, or labeled LP protein is provided.
  • the kit contains instructions for proper use and disposal of the contents after use.
  • the kit has compartments for each useful reagent.
  • the reagents are provided as a dry lyophilized powder, where the reagents may be reconstituted in an aqueous medium providing appropriate concenfrations of reagents for performing the assay.
  • Many of the aforementioned constituents of the drug screening and the diagnostic assays may be used without modification, or may be modified in a variety of ways.
  • labeling may be achieved by covalently or non-covalently joining a moiety that directly or indirectly provides a detectable signal.
  • the protein, test compound, LP protein or polypeptide (or antibodies thereto) are labeled either directly or indirectly.
  • Possibilities for direct labeling include label groups such as, e.g., without limitation, radiolabels (e.g., ⁇ 1); enzymes (U.S. Pat. No. 3,645,090) such as peroxidase and alkaline phosphatase; and fluorescent labels (U.S. Pat. No. 3,940,475) that are capable of monitoring the change in fluorescence intensity, wavelength shift, or fluorescence polarization.
  • Possibilities for indirect labeling include biotinylation of one constituent followed by binding to avidin coupled to an art known label such as one of the above. There are also numerous methods of separating the bound from the free protein, or alternatively bound from free test compound.
  • An LP protein is immobilized on various matrices followed by washing. Suitable matrices include plastic such as an ELISA plate, filters, and beads. Methods of immobilizing the LP protein to a matrix include, without limitation, direct adhesion to plastic, use of a capture antibody, chemical coupling, and biotin-avidin.
  • the last step in this approach involves the precipitation of protein binding partner or antigen/antibody complex by any of several methods including those utilizing, e.g., an organic solvent such as polyethylene glycol or a salt such as ammonium sulfate.
  • suitable separation techniques include, without limitation, a fluorescein antibody magnetizable particle method described in Rattle, et al. (1984) Clin. Chem. 30:1457- 1461, and the double antibody magnetic particle separation as described in U.S. Pat. No. 4,659,678.
  • oligonucleotide or polynucleotide sequences taken from the sequence of a LP protein. These sequences are used as probes for detecting levels of the LP protein message in samples from natural sources, or patients suspected of having an abnormal condition, e.g., cancer or developmental problem.
  • RNA and DNA nucleotide sequences The preparation of both RNA and DNA nucleotide sequences, the labeling of the sequences, and the preferred size of the sequences has received ample description and discussion in the literature.
  • an oligonucleotide probe should have at least about 14 nucleotides, usually at least about 18 nucleotides, and the polynucleotide probes may be up to several kilobases.
  • Various labels may be employed, most commonly radionuclides, particularly 32p. However, other techniques may also be employed, such as using biotin-modif ⁇ ed nucleotides for introduction into a polynucleotide.
  • the biotin then serves as the site for binding to avidin or antibodies, which may be labeled with a variety of labels, such as radionuclides, fluorophores, enzymes, or the like.
  • antibodies may be employed which can recognize specific duplexes, including DNA duplexes, RNA duplexes, DNA-RNA hybrid duplexes, or DNA-protein duplexes.
  • the antibodies in turn may be labeled and the assay carried out where the duplex is bound to a surface, so that upon the formation of duplex on the surface, the presence of antibody bound to the duplex is detected.
  • probes to the novel anti-sense RNA may be carried out using many conventional techniques such as nucleic acid hybridization, plus and minus screening, recombinational probing, hybrid released translation (HRT), and hybrid arrested translation (HART). This also includes amplification techniques such as polymerase chain reaction (PCR). Diagnostic kits, which also test for the qualitative or quantitative presence of other markers, are also contemplated. Diagnosis or prognosis may depend on the combination of multiple indications used as markers. Thus, kits may test for combinations of markers. See, e.g., Viallet, et al. (1989) Progress in Growth Factor Res. 1:89-97.
  • a kit may include, e.g., a recombinantly produced or chemically synthesized polypeptide antigen.
  • the polypeptide antigen of the kit may also be attached to a solid support.
  • the detecting means of the above-described kit includes, e.g., a solid support to which said polypeptide antigen is attached.
  • Such a kit may also include, e.g., a non-attached reporter-labeled anti-human antibody. In this embodiment, binding of the antibody to the polypeptide antigen is detected by binding of the reporter-labeled antibody.
  • the invention includes, e.g., a diagnostic kit for use in screening a biological sample, e.g., such as serum, containing an antigen of a polypeptide (or fragment thereof) of the invention.
  • the diagnostic kit can include, e.g., a substantially isolated antibody specifically and/or selectively immunoreactive with a polypeptide or polynucleotide antigen, and, a means for detecting the binding of the polynucleotide or polypeptide antigen to the antibody.
  • the antibody is attached to a solid support.
  • the antibody may be a monoclonal antibody.
  • the detecting means of the kit may include, e.g., a second, labeled monoclonal antibody.
  • the detecting means may include, e.g., a labeled, competing antigen.
  • test serum is reacted with a solid phase reagent having a surface-bound antigen obtained by an art known method or as described herein. After binding with specific antigen antibody to the reagent and removing unbound serum components, e.g., by washing; the reagent is reacted with reporter-labeled anti-human antibody to bind reporter to the reagent in proportion to the amount of bound anti-antigen antibody on the solid support. The reagent is again washed to remove unbound, labeled antibody, and the amount of reporter associated with the reagent is determined.
  • the reporter is an enzyme that is detected by incubating the solid phase in the presence of a suitable fluorometric, luminescent or calorimetric substrate (Sigma, St. Louis, MO).
  • a suitable fluorometric, luminescent or calorimetric substrate (Sigma, St. Louis, MO).
  • the solid surface reagent in the above assay is prepared by art known techniques for attaching proteinaceous material to a solid support, such as, e.g., polymeric beads, dip sticks, 96-well plate, or filter material.
  • Methods for attachment generally include, e.g., non-specific adsorption of a protein or polypeptide (or fragment thereof) to a solid support or covalent attachment of a polypeptide, protein (or fragment thereof), typically, e.g., through a free amine group, to a chemically reactive group, such as, e.g., an activated carboxyl, hydroxyl, or aldehyde group on the solid support.
  • streptavidin coated plates are used in conjunction with biotinylated antigen(s).
  • Preferred Embodiments include an isolated or recombinant nucleic acid molecule comprising a polynucleotide sequence that is at least 95% identical to a polynucleotide sequence of at least about: 10, 12, 14, 16, 18, 20, 22, 24, 26, 28, 30, 32, 34, 36, 38, 40, 42, 44, 46, 48, 50, 52, 54, 56, 58, 60, 62, 64, 66, 68, 70, 72, 74, 76, 78, 80, 82, 84, 86, 88, 90, 92, 94, 96, 98, 100, 110, 120, 130, 140, or 150 contiguous nucleotides of a sequence of Table 1.
  • inventions include an isolated or recombinant nucleic acid molecule comprising a polynucleotide sequence that is at least 95% identical to a polynucleotide sequence of at least about: 10, 12, 14, 16, 18, 20, 22, 24, 26, 28, 30, 32, 34, 36, 38, 40, 42, 44, 46, 48, 50, 52, 54, 56, 58, 60, 62, 64, 66, 68, 70, 72, 74, 76, 78, 80, 82, 84, 86, 88, 90, 92, 94, 96, 98, 100, 110, 120, 130, 140, or 150 contiguous nucleotides of a mature coding portion of a sequence of Table 1.
  • nucleic acid molecule wherein said sequence of contiguous nucleotides is include, e.g. in the nucleotide sequence of SEQ ID NO:X in the range of positions beginning with the nucleotide at about the position of the 5' nucleotide and ending with the nucleotide at about the position of the 3' nucleotide of a sequence of Table 1.
  • nucleic acid molecule wherein said sequence of contiguous nucleotides is included, e.g., in the nucleotide sequence of SEQ ID NO:X in the range of positions beginning with the nucleotide at about the position of the 5' nucleotide of the Start Codon and ending with the nucleotide at about the position of the 3' nucleotide of a sequence of Table 1.
  • nucleic acid molecule comprising polynucleotide sequence of SEQ ID NO:X in the range of positions beginning with the nucleotide at about the position of the 5' nucleotide of a correspondingly encoded First Amino Acid of a Signal Peptide and ending with the nucleotide at about the position of the 3' nucleotide of a sequence of Table 1.
  • an isolated or recombinant nucleic acid molecule comprising a polynucleotide sequence that is at least 95% identical to a polynucleotide sequence of at least about: 10, 12, 14, 16, 18, 20, 22, 24, 26, 28, 30, 32, 34, 36, 38, 40, 42, 44, 46, 48, 50, 52, 54, 56, 58, 60, 62, 64, 66, 68, 70, 72, 74, 76, 78, 80, 82, 84, 86, 88, 90, 92, 94, 96, 98, 100, 110, 120, 130, 140, or 150 contiguous nucleotides in at least one polynucleotide sequence fragment of SEQ ID NO:X.
  • nucleic acid molecule comprising a polynucleotide sequence that is at least 95% identical to a polynucleotide sequence of at least about: 200, 250, 300, 350, 400, 450, or 500 contiguous nucleotides of the mature coding portion of a sequence of Table 1.
  • nucleic acid molecule comprising a polynucleotide sequence that is at least 95% identical to a sequence of at least about: 10, 12, 14, 16, 18, 20, 22, 24, 26, 28, 30, 32, 34, 36, 38, 40, 42, 44, 46, 48, 50, 52, 54, 56, 58, 60, 62, 64, 66, 68, 70, 72, 74, 76, 78, 80, 82, 84, 86, 88, 90, 92, 94, 96, 98, 100, 110, 120, 130, 140, or 150 contiguous nucleotides in at least one nucleotide sequence fragment of SEQ ID NO:X, wherein the length of at least one such fragment is about 200, 250, 300, 350, 400, 450, or 500 contiguous nucleotides of a sequence of Table 1.
  • Another preferred embodiment is an isolated or recombinant nucleic acid molecule comprising a polynucleotide sequence that is at least 95% identical to a sequence of Table 1 beginning with the nucleotide at about the position of the 5' Nucleotide of the First Amino Acid of the Signal Peptide and ending with the nucleotide at about the position of the 3' Nucleotide of a sequence of Table 1.
  • a further preferred embodiment is an isolated or recombinant nucleic acid molecule comprising a polynucleotide sequence, which is at least 95% identical to the complete mature coding portion of a sequence of Table 1.
  • an isolated or recombinant nucleic acid molecule comprising polynucleotide sequence that hybridizes under stringent hybridization conditions to a mature coding portion of a polynucleotide of the invention (or fragment thereof), wherein the nucleic acid molecule that hybridizes does not hybridize under stringent hybridization conditions to a nucleic acid molecule having a nucleotide sequence consisting of only A residues or of only T residues.
  • Cardiovascular Conditions A composition of the invention may be used to, modulate, ameliorate, effect, treat, prevent, and/or diagnose a cardiovascular disease, disorder, syndrome, and/or condition.
  • cardiovascular abnormalities such as arterio-arterial fistula, arteriovenous fistula, cerebral arteriovenous malformations, congenital heart defects, pulmonary atresia, and Scimitar Syndrome peripheral artery disease, syndrome, such as limb ischemia.
  • Additional cardiovascular disorders encompass, e.g., congenital heart defects which include, e.g., aortic coarctation, car triafriatum, 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 e.g., aortopulmonary septal defect, endocardial cushion defects, Lutembacher's Syndrome, trilogy of Fallot, and ventricular heart septal defects.
  • congenital heart defects which include, e.g., aortic coarctation, car triafriatum, coronary vessel anomalies, crisscross heart, dextrocardia, patent ductus arteriosus
  • cardiovascular conditions include, e.g., heart disease syndrome, such as, e.g., arrhythmias, carcinoid heart disease syndrome, high cardiac output, low cardiac output, cardiac tamponade, endocarditis (including bacterial endocarditis), heart aneurysm, cardiac arrest, congestive heart failure, congestive cardiomyopathy, paroxysmal dyspnea, cardiac edema, heart hypertrophy, congestive cardiomyopathy, left ventricular hypertrophy, right ventricular hypertrophy, post-infarction heart rupture, ventricular septal rupture, heart valve disease, myocardial disease, myocardial ischemia, pericardial effusion, pericarditis (including constrictive and tuberculous pericarditis), pneumopericardium, post-pericardiotomy syndrome, pulmonary heart disease syndrome, rheumatic heart disease syndrome, ventricular dysfunction, hyperemia, cardiovascular pregnancy complications, Scimitar Syndrome, cardiovascular syphilis, and cardiovascular tuberculos
  • cardiovascular disorders include, e.g., arrhythmias including, e.g., sinus arrhythmia, atrial fibrillation, afrial flutter, bradycardia, extra systole, Adams-Stokes Syndrome, bundle-branch block, sinoafrial block, long QT syndrome, parasystole, Lown- Ganong-Levine Syndrome, Mahaim-type pre-excitation syndrome, Wolff-Parkinson- White syndrome, sick sinus syndrome, and ventricular fibrillation tachycardias.
  • arrhythmias including, e.g., sinus arrhythmia, atrial fibrillation, afrial flutter, bradycardia, extra systole, Adams-Stokes Syndrome, bundle-branch block, sinoafrial block, long QT syndrome, parasystole, Lown- Ganong-Levine Syndrome, Mahaim-type pre-excitation syndrome, Wolff-Parkinson- White syndrome, sick sinus syndrome, and
  • Tachycardias encompassed with the cardiovascular condition described herein include, e.g., paroxysmal tachycardia, supravenfricular tachycardia, accelerated idioventricular rhythm, atrioventricular nodal re-entry tachycardia, ectopic atrial tachycardia, ectopic junctional tachycardia, sinoafrial nodal re-entry tachycardia, sinus tachycardia, Torsades de Pointes Syndrome, and ventricular tachycardia.
  • Additional cardiovascular disorders include, e.g., heart valve disease such as, e.g., aortic valve insufficiency, aortic valve stenosis, heart 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.
  • heart valve disease such as, e.g., aortic valve insufficiency, aortic valve stenosis, heart murmurs, aortic valve prolapse, mitral valve prolapse, tricuspid valve prolapse, mitral valve insufficiency, mitral valve stenosis, pulmonary atresia, pulmonary valve insufficiency, pulmonary valve stenos
  • Myocardial conditions associated with cardiovascular disease include, e.g., myocardial diseases such as, e.g., alcoholic cardiomyopathy, congestive cardiomyopathy, hypertrophic cardiomyopathy, aortic subvalvular stenosis, pulmonary subvalvular stenosis, restrictive cardiomyopathy, Chagas cardiomyopathy, endocardial fibroelastosis, endomyocardial fibrosis, Kearns Syndrome, myocardial reperfusion injury, and myocarditis.
  • myocardial diseases such as, e.g., alcoholic cardiomyopathy, congestive cardiomyopathy, hypertrophic cardiomyopathy, aortic subvalvular stenosis, pulmonary subvalvular stenosis, restrictive cardiomyopathy, Chagas cardiomyopathy, endocardial fibroelastosis, endomyocardial fibrosis, Kearns Syndrome, myocardial reperfusion injury, and myocarditis.
  • Cardiovascular conditions include, e.g., myocardial ischemias such as, e.g., coronary disease syndrome, such as e.g., angina pectoris, coronary aneurysm, coronary arteriosclerosis, coronary thrombosis, coronary vasispasm, myocardial infarction, and myocardial stunning.
  • myocardial ischemias such as, e.g., coronary disease syndrome, such as e.g., angina pectoris, coronary aneurysm, coronary arteriosclerosis, coronary thrombosis, coronary vasispasm, myocardial infarction, and myocardial stunning.
  • coronary disease syndrome such as e.g., angina pectoris, coronary aneurysm, coronary arteriosclerosis, coronary thrombosis, coronary vasispasm, myocardial infarction, and myocardial stunning.
  • Cardiovascular diseases also encompassed herein include, e.g., vascular diseases such as e.g., aneurysms, angiodysplasia, angiomatosis, bacillary angiomatosis, Hippel- Lindau Disease syndrome, Klippel-Trenaunay- Weber Syndrome, Sturge- Weber Syndrome, angioneurotic edema, aortic disease, Takayasu's Arteritis, aortitis, Leriche's Syndrome, arterial occlusive disease, arteritis, enarteritis, polyarteritis nodosa, cerebrovascular disease, diabetic angiopathies, diabetic retinopathy, embolism, thrombosis, erytbromeialgia, hemorrhoids, hepatic veno-occlusive disease syndrome, hypertension, hypotension, ischemia, peripheral vascular diseases, phlebitis, pulmonary veno-occ
  • Cardiovascular conditions further include, e.g., aneurysms such as, e.g., dissecting aneurysms, false aneurysms, infected aneurysms, ruptured aneurysms, aortic aneurysms, cerebral aneurysms, coronary aneurysms, heart aneurysms, and iliac aneurysms.
  • aneurysms such as, e.g., dissecting aneurysms, false aneurysms, infected aneurysms, ruptured aneurysms, aortic aneurysms, cerebral aneurysms, coronary aneurysms, heart aneurysms, and iliac aneurysms.
  • Arterial occlusive cardiovascular conditions include, e.g., arteriosclerosis, intermittent claudication, carotid stenosis, fibromuscular dysplasias, mesenteric vascular occlusion, Moyamoya disease syndrome, renal artery obstruction, retinal artery occlusion, and thromboangiitis obliterans.
  • Cerebrovascular cardiovascular conditions include, e.g., carotid artery disease, cerebral amyloid angiopathy, cerebral aneurysm, cerebral anoxia, cerebral arteriosclerosis, cerebral arteriovenous malformation, cerebral artery disease, cerebral embolism and thrombosis, carotid artery thrombosis, sinus thrombosis, Wallenberg's syndrome, cerebral hemorrhage, epidural hematoma, subdural hematoma, subarachnoid hemorrhage, cerebral infarction, cerebral ischemia (including transient cerebral ischemia), subclavian steal syndrome, perivenfricular leukomalacia, vascular headache, cluster headache, migraine, and vertebrobasilar insufficiency.
  • Embolic cardiovascular conditions include, e.g., air embolisms, amniotic fluid embolisms, cholesterol embolisms, blue toe syndrome, fat embolisms, pulmonary embolisms, and thromboembolisms
  • Thrombotic cardiovascular conditions include, e.g., coronary thrombosis, hepatic vein thrombosis, retinal vein occlusion, carotid artery thrombosis, sinus thrombosis, Wallenberg's syndrome, and thrombophlebitis.
  • Ischemic conditions include, e.g., cerebral ischemia, ischemic colitis, compartment syndromes, anterior compartment syndrome, myocardial ischemia, reperfusion injuries, and peripheral limb ischemia.
  • Vasculitic conditions include, e.g., 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.
  • a binding composition can be beneficial in ameliorating critical limb ischemia and coronary disease.
  • a binding composition may be administered using any art known method, described herein
  • a binding composition may administered as part of a therapeutic composition or formulation, as described in detail herein. Methods of delivering a binding composition are also described herein.
  • angiogenesis is stringently regulated, and delimited spatially and temporally.
  • pathological angiogenesis such as, e.g., during solid tumor formation, these regulatory controls fail and unregulated angiogenesis can become pathologic by sustaining progression of many neoplastic and non-neoplastic diseases.
  • a number of serious diseases are dominated by abnormal neovascularization (including, e.g., solid tumor growth and metastases, arthritis, some types of eye conditions, and psoriasis; see, e.g., reviews by Moses, et al., Biotech. 9630-634 (1991); Folkman, et al., N. Engl. J. Med., 333: 1757-1763 (1995); Auerbach, et al., J. Microvasc. Res. 29:401-4 11 (1985); Folkman, "Advances in Cancer Research," eds. Klein and Weinhouse, Academic Press, New York, pp. 175-203 (1985); Patz, Am. J. Opthalmol.
  • administration of a composition of the invention provides for the treatment, amelioration, modulation, diagnosis, and/or inhibition of a disease, disorder, syndrome, and/or condition associated with neovascularization.
  • Malignant and metastatic conditions that can be effected in a desired fashion using a composition of the invention includes, e.g., without limitation, a malignancy, solid tumor, and a cancer as described herein or as otherwise known in the art (for a review of such disorders, syndromes, etc. see, e.g., Fishman, et al., Medicine, 2d Ed., J. B. Lippincott Co., Philadelphia (1985)).
  • the present invention provides a method of ameliorating, modulating, treating, preventing, and/or diagnosing an angiogenesis-related disease and/or disorder, comprising administering to a subject in need thereof a beneficially effective amount of a composition of the invention.
  • cancers that may be so affected using a composition of the invention includes, e.g., without limit a solid tumor, including e.g., 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 metastases; melanomas; glioblastoma; Kaposi's sarcoma; leiomyosarcoma; non-small cell lung cancer; colorectal cancer; advanced malignancies; and blood born tumors such as e.g., leukemia.
  • a composition may be delivered topically, to treat or prevent cancers such as, e.g., skin cancer, head and neck tumors, breast tumors, and Kaposi's sarcoma.
  • a composition of the invention may be utilized to treat superficial forms of bladder cancer by, e.g., intravesical administration into the tumor, or near the tumor site; via injection or a catheter.
  • intravesical administration into the tumor, or near the tumor site; via injection or a catheter.
  • the appropriate mode of administration will vary according to the cancer to be treated. Other modes of delivery are discussed herein.
  • a composition of the invention may also be useful in modulating, ameliorating, treating, preventing, and/or diagnosing another disease, disorder, syndrome, and/or condition, besides a cell proliferative condition (e.g., a cancer) that is assisted by abnormal angiogenic activity.
  • a cell proliferative condition e.g., a cancer
  • Such close group conditions include, e.g., without limitation, benign tumors, e.g., such as hemangiomas, acoustic neuromas, neurofibromas, trachomas, and pyogenic granulomas; atherosclerotic plaques; ocular angiogenic diseases, e.g., diabetic retinopathy, retinopathy of prematurity, macular degeneration, cornea graft rejection, neovascular glaucoma, retrolental fibroplasia, rubeosis, retinoblastoma, uvietis and Pterygia (abnormal blood vessel growth) of the eye; rheumatoid arthritis; psoriasis; delayed wound healing; endomefriosis; vasculogenesis; granulations; hypertrophic scars (keloids); nonunion fractures; scleroderma; trachoma; vascular adhesions; myocardial angiogenesis; coronary
  • the present invention also provides methods for ameliorating, treating, preventing, and/or diagnosing neovascular diseases of the eye, including e.g., corneal graft neovascularization, neovascular glaucoma, proliferative diabetic retinopathy, retrolental fibroplasia and macular degeneration.
  • neovascular diseases of the eye including e.g., corneal graft neovascularization, neovascular glaucoma, proliferative diabetic retinopathy, retrolental fibroplasia and macular degeneration.
  • ocular diseases, disorders, syndromes, and/or conditions associated with neovascularization that can be modulated ameliorated, treated, prevented, and/or diagnosed with a composition of the invention include, e.g., without limit; neovascular glaucoma, diabetic retinopathy, retinoblastoma, retrolental fibroplasia, uveitis, retinopathy of premature macular degeneration, corneal graft neovascularization, as well as other inflammatory eye diseases, ocular tumors, and diseases associated with choroidal or iris neovascularization (see, e.g., reviews by Waltman, et al., (1978) Am. J. Ophthal.
  • comeal neovascularization including e.g., comeal infections (e.g., trachoma, herpes simplex keratitis, leishmaniasis and onchocerciasis), immunological processes (e.g., graft rejection and Stevens- Johnson's syndrome), alkali bums, trauma, inflammation (of any cause), toxic and nutritional deficiency states, and as a complication of using contact lenses.
  • comeal infections e.g., trachoma, herpes simplex keratitis, leishmaniasis and onchocerciasis
  • immunological processes e.g., graft rejection and Stevens- Johnson's syndrome
  • alkali bums e.g., trauma, inflammation (of any cause), toxic and nutritional deficiency states, and as a complication of using contact lenses.
  • methods for treating or preventing proliferative diabetic retinopathy, comprising administering to a patient a therapeutically effective amount of a composition of the invention to the eyes, such that the formation of blood vessels is inhibited.
  • Additional, diseases, disorders, syndromes, and/or conditions that can be modulated, ameliorated, treated, prevented, and/or diagnosed with a composition of the invention include, e.g., without limitation, hemangioma, arthritis, psoriasis, angiofibroma, atherosclerotic plaques, delayed wound healing, granulations, hemophilic joints, hyperfrophic scars, nonunion fractures, Osier- Weber syndrome, pyogenic granuloma, scleroderma, trachoma, and vascular adhesions.
  • diseases, disorders, states, syndromes, and/or conditions that can be modulated, ameliorated, treated, prevented, and/or diagnosed with a composition of the invention include, e.g., without limitation, solid tumors, blood bom tumors such as leukemias, tumor metastasis, Kaposi's sarcoma, benign tumors (e.g., hemangiomas), acoustic neuromas, neurofibromas, trachomas, and pyogenic granulomas, rheumatoid arthritis, psoriasis, ocular angiogenic diseases, e.g., diabetic retinopathy, retinopathy of prematurity, macular degeneration, comeal graft rejection, neovascular glaucoma, retrolental fibroplasia, rubeosis, retinoblastoma, and uvietis, delayed wound healing, endometriosis, vasculogenesis, granulations, hyperfrophi
  • cancers such as, e.g., follicular lymphomas, carcinomas with p53 mutations, and hormone-dependent tumors, including, e.g., but without limit, colon cancer, cardiac tumors, pancreatic cancer, melanoma, retinoblastoma, glioblastoma, lung cancer, intestinal cancer, testicular cancer, stomach cancer, neuroblastoma, myxoma, myoma, lymphoma, endothelioma, osteoblastoma, osteoclastoma, osteosarcoma, chondrosarcoma, adenoma, breast cancer, prostate cancer, Kaposi's sarcoma and ovarian cancer); autoimmune conditions (such as, e.g., multiple s
  • a composition of the invention is used to inhibit growth, progression, and/or metastases of cancers such as, in particular, those listed herein.
  • Additional diseases, states, syndromes, or conditions associated with increased cell survival that could be modulated, ameliorated, freated, prevented, or diagnosed by a composition of the invention include, e.g., without limitation, progression, and/or metastases of malignancies and related disorders such as leukemia including acute leukemias (such as, e.g., acute lymphocytic leukemia, acute myelocytic leukemia, including myeloblastic, promyelocytic, myelomonocytic, monocytic, and erythroleukemia) and chronic leukemias (e.g., chronic myelocytic, chronic granulocytic, leukemia, and chronic lymphocytic leukemia)), polycythemia Vera, lymphomas (e
  • acute leukemias such as, e.g., acute lympho
  • Diseases associated with increased apoptosis that could be modulated, ameliorated, treated, prevented, and/or diagnosed by a composition of the invention include, e.g., AIDS, conditions (such as, e.g., Alzheimer's disease syndrome, Parkinson's disease syndrome, Amyotrophic lateral sclerosis, Retinitis pigmentosa, Cerebellar degeneration and brain tumor, or prion associated disease); autoimmune conditions (such as, e.g., multiple sclerosis, Sjogren's syndrome, Hashimoto's thyroiditis, biliary cirrhosis, Behcet's disease syndrome, Crohn's disease syndrome, polymyositis, systemic lupus erythematosus, immune-related glomerulonephritis, and rheumatoid arthritis); myelodysplastic syndromes (such as aplastic anemia), graft v.
  • AIDS conditions
  • conditions such as, e.g., Alzheimer's disease syndrome
  • ischemic injury such as that caused by myocardial infarction, stroke and reperfusion injury
  • liver injury such as, e.g., hepatitis related liver injury, ischemia reperfusion injury, cholestosis (bile duct injury), and liver cancer
  • toxin-induced liver disease such as, e.g., that caused by alcohol
  • septic shock such as, e.g., cachexia, and anorexia.
  • a composition of the invention could be used to prevent and heal damage to the lungs due to various pathological states, such as, e.g., stimulating proliferation and differentiation to promote repair of alveoli and bronchiolar epithelium.
  • emphysema inhalation injuries, that (e.g., from smoke inhalation) and burns, which cause necrosis of the bronchiolar epithelium and alveoli could be effectively ameliorated, treated, prevented, and/or diagnosed using a polynucleotide or polypeptide of the invention (or fragment thereof), or an agonist or antagonist thereto.
  • a composition of the invention could be used to stimulate the proliferation of and differentiation of type II pneumocytes, to help treat or prevent hyaline membrane diseases, such as e.g., infant respiratory distress syndrome and bronchopulmonary displasia, (in premature infants).
  • Kits This invention also contemplates use of binding compositions in a variety of diagnostic kits and methods for detecting the presence of an LP composition of the invention.
  • ti e kit will have a compartment containing either a defined an LP composition of the invention or a binding composition, which recognizes one or the other, e.g., binding partner fragments or antibodies.
  • a preferred kit for determining the concentration of an LP composition of the invention in a sample would typically comprise a labeled compound, e.g., binding composition or antibody, having known binding affinity for the an LP composition of the invention protein, a source of an LP composition of the invention(naturally occurring or recombinant), and a means for separating the bound from free labeled compound, for example, a solid phase for immobilizing the an LP composition of the invention protein. Compartments containing reagents, and instructions, will normally be provided. Antibodies, including antigen binding fragments, specific for a an LP composition of the invention or fragments thereof are useful in diagnostic applications to detect the presence of elevated levels of an LP composition of the invention and/or its fragments.
  • diagnostic assays can employ lysates, live cells, fixed cells, immunofluorescence, cell cultures, body fluids, and further can involve the detection of antigens related to the protein in serum, or the like. Diagnostic assays may be homogeneous (without a separation step between free reagent and antigen-an LP composition of the invention or - WDS protein complex) or heterogeneous (with a separation step).
  • Various commercial assays exist, such as radioimmunoassay (RIA), enzyme-linked immunosorbent assay (ELISA), enzyme immunoassay (EIA), enzyme-multiplied immunoassay technique (EMIT), substrate-labeled fluorescent immunoassay (SLFIA), and the like.
  • unlabeled antibodies are employed by using a second antibody which is labeled and which recognizes an antibody to an LP composition of the invention or to a particular fragment thereof.
  • Similar assays are also extensively discussed in the literature (see, e.g., Harlow and Lane (1988) Antibodies: A Laboratory Manual, CSH Press, NY; Chan (ed.) (1987) Immunoassay: A Practical Guide Academic Press, Orlando, FL; Price and Newman (eds.) (1991) Principles and Practice of Immunoassay Stockton Press, NY; and Ngo (ed.) (1988) Nonisotopic immunoassay Plenum Press, NY).
  • Anti-idiotypic antibodies may have similar use to diagnose the presence of antibodies against an an LP composition of the invention or polypeptide, as such may be diagnostic of various abnormal states, conditions, disorders, or syndromes.
  • overproduction of an LP composition of the invention may result in production of various immunological or other physiological reactions which may be diagnostic of abnormal physiological states, e.g., in cell growth, activation, or differentiation.
  • the reagents for diagnostic assays are supplied in kits, to optimize the sensitivity of the assay.
  • the protocol, and the label either labeled or unlabeled antibody or binding composition, or labeled an LP composition of the invention is provided.
  • the kit contains instructions for proper use and disposal of the contents after use.
  • the kit has compartments for each useful reagent.
  • the reagents are provided as a dry lyophilized powder, where the reagents may be reconstituted in an aqueous medium providing appropriate concentrations of reagents for performing the assay.
  • Many of the aforementioned constituents of the drug screening and the diagnostic assays may be used without modification, or may be modified in a variety of ways.
  • labeling may be achieved by covalently or non-covalently joining a moiety that directly or indirectly provides a detectable signal.
  • the protein, test compound, an LP composition of the invention or polypeptide (or antibodies thereto) are labeled either directly or indirectly.
  • Possibilities for direct labeling include label groups such as, e.g., without limitation, radiolabels (e.g., 125j).
  • enzymes U.S. Pat. No. 3,645,090
  • fluorescent labels U.S. Pat. No. 3,940,475) that are capable of monitoring the change in fluorescence intensity, wavelength shift, or fluorescence polarization.
  • Possibilities for indirect labeling include biotinylation of one constituent followed by binding to avidin coupled to an art known label such as one of the above. There are also numerous methods of separating the bound from the free protein, or alternatively bound from free test compound.
  • An LP composition of the invention is immobilized on various matrices followed by washing. Suitable matrices include plastic such as an ELISA plate, filters, and beads. Methods of immobilizing the an LP composition of the invention to a matrix include, without limitation, direct adhesion to plastic, use of a capture antibody, chemical coupling, and biotin-avidin.
  • the last step in this approach involves the precipitation of protein/binding composition or antigen/antibody complex by any of several methods including those utilizing, e.g., an organic solvent such as polyethylene glycol or a salt such as ammonium sulfate.
  • suitable separation techniques include, without limitation, a fluorescein antibody magnetizable particle method described in Rattle, et al. (1984) Clin. Chem. 30:1457- 1461, and the double antibody magnetic particle separation as described in U.S. Pat. No. 4,659,678.
  • a kit may include, e.g., a recombinantly produced or chemically synthesized polypeptide antigen.
  • the polypeptide antigen of the kit may also be attached to a solid support.
  • the detecting means of the above-described kit includes, e.g., a solid support to which said polypeptide antigen is attached.
  • Such a kit may also include, e.g., a non-attached reporter-labeled anti-human antibody. In this embodiment, binding of the antibody to the polypeptide antigen is detected by binding of the reporter-labeled antibody.
  • the invention includes, e.g., a diagnostic kit for use in screening a biological sample, e.g., such as serum, containing an antigen of a polypeptide (or fragment thereof) of the invention.
  • the diagnostic kit can include, e.g., a substantially isolated antibody specifically and/or selectively immunoreactive with a polypeptide or polynucleotide antigen, and, a means for detecting the binding of the polynucleotide or polypeptide antigen to the antibody.
  • the antibody is attached to a solid support.
  • the antibody may be a monoclonal antibody.
  • the detecting means of the kit may include, e.g., a second, labeled monoclonal antibody.
  • the detecting means may include, e.g., a labeled, competing antigen.
  • test serum is reacted with a solid phase reagent having a surface-bound antigen obtained by an art known method or as described herein. After binding with specific antigen antibody to the reagent and removing unbound serum components, e.g., by washing; the reagent is reacted with reporter-labeled anti-human antibody to bind reporter to the reagent in proportion to the amount of bound anti-antigen antibody on the solid support. The reagent is again washed to remove unbound, labeled antibody, and the amount of reporter associated with the reagent is determined.
  • the reporter is an enzyme that is detected by incubating the solid phase in the presence of a suitable fluorometric, luminescent or calorimetric substrate (Sigma, St. Louis, MO).
  • a suitable fluorometric, luminescent or calorimetric substrate (Sigma, St. Louis, MO).
  • the solid surface reagent in the above assay is prepared by art known techniques for attaching proteinaceous material to a solid support, such as, e.g., polymeric beads, dip sticks, 96-well plate, or filter material.
  • Methods for attachment generally include, e.g., non-specific adsorption of a protein or polypeptide (or fragment thereof) to a solid support or covalent attachment of a polypeptide, protein (or fragment thereof), typically, e.g., through a free amine group, to a chemically reactive group, such as, e.g., an activated carboxyl, hydroxyl, or aldehyde group on the solid support.
  • streptavidin coated plates are used in conjunction with biotinylated antigen(s).
  • Methods for protein purification include such methods as ammonium sulfate precipitation, column chromatography, electrophoresis, centrifugation, crystallization, and others. See, e.g., Ausubel, et al. (1987 and periodic supplements); Deutscher (1990) "Guide to Protein Purification," Methods in Enzymology vol. 182, and other volumes in this series; Coligan, et al. (1995 and supplements) Current Protocols in Protein Science John Wiley and Sons, New York, NY; P.
  • THP-1 cells were obtained from ATCC. Cells were cultured in RPMI-1640 medium supplemented with 0.05 mM b-mercaptoethanol and 10% FBS. For all experiments, cells were plated at a density of 500,000 cells/ml in 24 well plates.
  • Cytokine measurements THP-1 cells were treated with 10 nM PMA for 24 hours. Appropriate agents were added to the cultures and then incubated for an additional 24 hours. Culture supernatants were collected for measurement of IL-6 and IL-8 using ELISA kits from R&D systems. To determine the concentration of other cytokines, supernatants were analyzed with the LINCO 10-plex cytokine kit, using luminex technology.
  • Factor Xa assay was performed using soluble tissue factor or PMA treated THP-1 cells as a source of TF. For assays using THP-1 cells, the cells were first freated with 10 nM PMA for 24 hours before the addition of FVIIa inhibitors. The cells were scraped in assay buffer containing 0.03M Tris and 0.15M NaCL, pH 7.4. Equal volume of FX and FVIIa (Enzyme Research Labs) cocktail prepared fresh in assay buffer containing 0.1% HSA, was added to the cell suspension to achieve a final concentration of 60nM of FX and FVIIA.
  • Amino acids sequencing Protein from SDS-gels were extracted in water and subjected to amino acid sequencing using automatic amino acid sequencer.
  • FVII and truncated FVII Genes encoding FVII and truncated FVII were amplified by utilizing the polymerase chain reaction (PCR) from FVII cDNA obtained from Origene (Cat # 006137).
  • PCR polymerase chain reaction
  • oligonucleotide primers 5'- gaggcgcgccgccaccatggccaacgcgttcctggag-3' (HFP-1), 5'- atgcggccgctgggctagggaaatggggc-3' (HFP-2), 5'-gaggctagccgccaccatggtctcccaggccctcagg- 3' (HFP-3), 5'-gaaatccagaacagcttcgtcctctcg-3' (HFP-4) were designed based on the published human cDNA sequences (NCBI accession #s NM_019616.1 and JO 2933.1).
  • the vector pRB12-137A was then converted into vector pRB12-142A containing cDNA sequence that encoded the truncated form of F VII plus 38 amino acid residues of signal peptide by using site-directed loop out mutagenesis with oligonuceloti.de primers, 5'-gcaccggcgccggcgcctgttctggatttc-3' (HFP-5) and 5'-gaaatccagaacaggcgccggcgcggtgcggtgc-3' (HFP-6). After confirming the sequences, both expression vectors were stably transfected into HEK-293 cells by using FuGene 6 transfection reagent (Roche, Cat # 1814443).
  • Transfected cells were selected for hygromycin B resistance (125 ugm mL), individual colonies isolated and screened for high level expression in serum free medium.
  • Western blot analysis FVII, FVIIa purchased from either ERL or American Diagnostic were subjected to non-reducing SDS-PAGE. The proteins were transferred to PVDF membrane and immunoblotted using FVIIa antibody (American Diagnostica). PVDF membranes were developed using ECL detection system (Amersham).
  • Cytokine induction by FVIIa does not require thrombin or factor Xa: To examine if IL-6 production in response to FVIIa involves thrombin, we determined the effect of thrombin inhibitor hirudin on IL-6 production. Addition of hirudin did not affect IL-6 production in either the control or FVIIa treated cells ( Figure 2). Similarly, addition of factor Xa to control or the FVIIa freated cells did not enhance IL-6 expression. In this study, a slight decrease in IL-6 production was observed in FXa treated cultures. These data suggest that neither thrombin nor FXa were required for IL-6 induction by FVIIa.
  • FIG. 1 shows the factor X to factor Xa converting activity of three different preparations of FVIIa.
  • Preparation 1 a recombinant FVIIa produced using mammalian cell expression system showed the expected factor X activating activity.
  • Preparation 2 a freshly purified FVIIa from human plasma was slightly more active than the recombinant preparation.
  • Preparation 3 was also plasma derived FVIIa but had been stored at -20C for several months. The stored material demonstrated low activity for FX to FXa conversion (figure 3A). IL-6 inducing activity of these preparations is shown in figure 3B. Preparations 1 and 2 that exhibited good factor X activation showed little or no induction of IL-6 whereas preparation 3 that exhibited very low FX activation showed the highest induction of IL-6. These results indicated that factor X activation and cytokine induction by FVIIa were dissociable activities.
  • FIG. 4A shows that the recombinant FVIIa mainly contained a 52 kD protein band corresponding to FVIIa.
  • Preparations 2 contained FVIIa as a major protein band but also contained a lower molecular weight band corresponding to 46 kD.
  • the major protein in preparation 3 was the 46 kD band. Both bands cross-reacted with FVIIa antibodies in the western blots. The two major bands corresponding 52 kD and 46 kD were eluted from the gel and subjected to amino acid sequencing.
  • the 52 kD protein band exhibited two N-terminals corresponding to the two chains of FVIIa.
  • the N-terminus corresponded to the N- terminal sequence of the light chain of FVIIa.
  • the second N-terminal (IVGGKVCPK — ) matched with the N-terminal sequence of the protease domain.
  • the 46 kD protein band also contained two peptides.
  • One N-terminal matched with the N-terminus of the protease domain (IVGGKVCPK—) whereas the second N-terminal (LFWISYSDG— ) overlapped with a sequence within FVIIa starting at residue Leu39.
  • Factor X activation activity was co-eluted with the protein corresponding to the 52 kD FVIIa. These data further support that the N-terminal truncated FVIIa lacked FX activation activity but stimulated IL-8 expression.
  • the expressed FVII can be activated to FVIIa by a single cleavage by FXa at amino acid residues Rl 52-1153 (Radcliffe, R., andNemerson, Y. (1976) J. Biol. Chem. 251, 4797- 4802).
  • FXa amino acid residues Rl 52-1153
  • IL-8 inducing activity before after activation with FXa.
  • the recombinant truncated FVII did not produce FX activation.
  • the truncated FVII or the native FVII did not stimulate IL-8 expression unless activated by treatment with FXa.
  • FVII was activated to FVIIa and effectively stimulated IL-8 expression.
  • the truncated FVII also induced IL-8 expression after activation with FXa.
  • FVIIa (LPFVII) that was highly potent inducer of cytokine expression in THP-1 cells.
  • the truncated FVIIa was produced by removal of the first 38 amino acid residues from the protein by proteolytic cleavage.
  • the "Gla" domain resides in the N-terminus of FVIIa, removal of the first 38 amino acid residues rendered the molecule devoid of FX binding ability. This result was consistent with Applicant' s demonstration of a lack of FX to FXa converting activity of LPFVII.
  • a lack of coagulation activity and maintenance of cytokine inducing activity in LP FVIIa was further confirmed by cloning and expression of LP FVIIa in mammalian cells.
  • FVIIa can be generated from FVII by a single proteolytic cleavage by FXa or FXIIa at residues Rl 52-1153 (Radcliffe, R., and Nemerson, Y. (1976) J. Biol. Chem. 251, 4797- 4802).
  • Assessment of the activities of the recombinant FVII and truncated FVII before and after FXa treatment demonstrated that the truncated FVIIa lacks FX activation activity but unexpectedly maintained stimulation of IL-8 expression in THP-1 cells.
  • FVIIa-TF pathway has been shown to induce several genes that mediate angiogenesis.
  • FVIIa induces IL-8 expression in macrophage and fibroblasts (
  • IL-8 is a chemoatfractant for endothelial cells and monocyte, and promotes angiogenesis in vivo (Radcliffe, R., and Nemerson, Y. (1976) J. Biol. Chem. 251, 4797- 4802; Szekanecz, et al., 1994 Pathology 62, 134-139).
  • FVIIa also induces the expression of the CCN gene family members CYR61 and connective tissue growth factor (CTGF) ( Oxfordr, et al., 2000 J. Biol. Chem. 275, 6580-6585).
  • CTGF connective tissue growth factor
  • the cystein rich protein CYR61 acts as a ligand for the integrin alpha v beta 3 (22).
  • CYR61 stimulates endothelial cell growth and migration, and in vivo angiogenesis (Kireeva, et al., 1996 Mol Cell Biol 16, 1326-1334; Babic, et al. 1998 Proc. Natl. Acad. Sci. U.S.A. 95, 6355-6360).
  • CYR 61 gene knock out mice show gross vascular defects (Mo, et al., 2002 Mol. Cell. Biol. 22, 8709-8720).
  • Other FVIIa induced angiogenic genes include VEGF and FGF-5 (OUivier, et al., 2000 Thromb. Vase. Biol. 20, 1374-1381; skyr, et al., 2000 J. Biol. Chem.
  • FVIIa may induce angiognesis
  • TF-VIIa activation in vivo is usually associated with the induction of coagulation and platelet activation. Since the truncated FVIIa is devoid of the coagulation activity but induces proangiogenesis genes, this molecule may be uniquely suited for angiogenic therapy.
  • Tissue factor is expressed at high level in atherosclerotic plaque (Wilcox, et al., 1989 Proc. Natl. Acad. Sci. U.S.A. 86, 2839-2843; Marmur, et al., 1996 Circ. 94, 1226- 1232; and Mallat, et al., 2000 Circ.
  • TF-VIIa pathway has been implicated in the pathogenesis of vascular inflammation and plaque rupture. Generation of truncated FVIIa in the atherosclerotic plaque may provide a potent stimulus for the induction and maintenance of vascular inflammation.
  • SEQ ID NO: 1 is primate LP FVIIa nucleic acid sequence.
  • SEQ ID NO: 2 is primate LP FVIIa amino acid sequence.

Landscapes

  • Health & Medical Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Engineering & Computer Science (AREA)
  • Organic Chemistry (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Zoology (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Genetics & Genomics (AREA)
  • Wood Science & Technology (AREA)
  • Biochemistry (AREA)
  • General Engineering & Computer Science (AREA)
  • General Health & Medical Sciences (AREA)
  • Biomedical Technology (AREA)
  • Molecular Biology (AREA)
  • Biotechnology (AREA)
  • Microbiology (AREA)
  • Medicinal Chemistry (AREA)
  • Peptides Or Proteins (AREA)

Abstract

Isolated and/or recombinant compositions related to Factor VIIa, reagents related thereto are provided. Methods of using said reagents and diagnostic kits are also provided.

Description

LP MAMMALIAN PROTEINS; RELATED REAGENTS
FIELD OF THE INVENTION The present invention generally relates to compositions related to Factor Vila. In particular, it provides purified genes, polynucleotide sequences, proteins, polypeptides, antibodies, binding compositions, and related reagents useful, e.g., in the diagnosis, treatment, and prevention of cell proliferative, autoirnmune/inflarnmatory, cardiovascular, neurological, hematopoetic, and developmental disorders, and in the assessment of the effects of exogenous compounds on the expression of nucleic acid and amino acid sequences of such proteins. BACKGROUND OF THE INVENTION The coagulation factor Vila. (FVIIa) is a key initiator of the proteolytic cascade that leads to the formation of blood clot via the intrinsic pathway. The primary function of FVIIa is the proteolytic activation of factor IX and factor X (FX) after complexation with the cell associated tissue factor (TF). Activated FX (factor Xa) converts prothrombin to thrombin which in turn induces the formation of fibrin from fibrinogen (Martin, et al. 1998 Thromb. Res. 90, 1-25; Craner, et al. 1996 Thromb. Res. 81, 1-41). In addition to its crucial role in coagulation, FVIIa has been shown to act as a ligand for triggering TF mediated signaling pathway. The FVIIa-TF cellular pathway plays an important role in inflammation (Cunningham, et al. 1999 Blood 94, 3413-3420) and angiogenesis (Shoji, et al. 1998 Am. J. Pathol. 152, 399-411; Carmeliet, et al., Nature 383, 73-75). Several lines of evidence suggest that FVIIa-TF pathway is critical for blood vessel development. Gene knock out studies have demonstrated that the mutant mice lacking TF have fragile vessels (Carmeliet, et al., Nature 383, 73-75). FVIIa induces a number of pro-angiogenic genes including VEGF, Egrl, CYR61, CTGF, FGF-5, IL-8, and MMP1, MMP13 (Camerer, et al, 2000 J. Biol. Chem. 275, 6580-6585). In addition, the intracellular signaling induced by FVIIa-TF involves activation of PI3 kinase and Akt (Versteg, et al., 2000 J. Biol. Chem. 275, 28750-28756), a pathway consistent with angiogenesis activation. The role of FVIIa-TF in tumor angiogenesis is corroborated by high level expression of TF in tumors that correlates with metastatic state (Fernandez, and Rickles, 2002 Curr. Opin. Hematol. 9. 401-406). Consequently, there is a need in the fields of hematopoiesis, inflammation, and angiogenesis for novel compositions that can modulate and/or influence conditions or states relating to these fields. The present invention satisfies such a need by disclosing a novel truncated FVIIa, designated LP FVIIa, which is generated by deletion of 38 amino acid residues from an N-terminad portion of native Factor Vila. LP FVIIIa is devoid of Factor X activation activity and is a significantly more potent inducer of proangiogenic cytokines than native Factor Vila.
SUMMARY OF THE INVENTION The present invention is based in part upon the discovery of novel Factor Vila LP compositions (herein designated as LP FVIIa compositions including without limitation, LPs disclosed with sequence identifiers and in a Table herein and variants thereof). The invention provides substantially pure, isolated, and/or recombinant protein or polypeptide exhibiting identity over a length of at least about 12 contiguous amino acids to a corresponding sequence of a Table described herein; a fusion protein comprising a sequence as described herein. In preferred embodiments, an LP portion is at least about: 12, 15, 20, 25, 30, 35, 40, 45, 50, 55, 60, 65, 70, 75, 80, 85, 90, 95, 100, 110, 120, 130, 140, 150, 160, 170, 180, 190, 200, 210, 220, 230, 240, 250, 260, 270, 280, 290, 300, 310, 320, 330, 340, 350, 360, 370, 380, 390, 400, or 405 contiguous amino acid residues in length of a sequence described herein, or of SEQ ID NO: X or Y, or an LP sequence of Table 1. In other embodiments, an LP (e.g., LPFVII, or variants thereof) comprises a mature sequence (or a variant sequence) of Table 1; protein or peptide: is from a warm blooded animal selected from a mammal, including a primate; comprises at least one polypeptide segment of Table 1, exhibits a plurality of portions exhibiting identity to polypeptide segments of Table 1; is a natural allelic variant of the LPFVII; has a length at least about 30, 35, 40, 45, 50, 55, 60, 65, 70, 75, 80, 85, 90, 95, 100, 110, 120, 130, 140, 150, 160, 170, 180, 190, 200, 210, 220, 230, 240, 250, 260, 270, 280, 290, 300, 310, 320, 330, 340, 350, 360, 370, 380, 390, 400, or 405 contiguous amino acid residues exhibiting identity to a polypeptide segment of Table 1; exhibits at least two non-overlapping epitopes which are specific for a mammalian LPFVII; exhibits identity over a length of at least about 30, 35, 40, 45, 50, 55, 60, 65, 70, 75, 80, 85, 90, 95, 100, 110, 120, 130, 140, 150, 160, 170, 180, 190, 200, 210, 220, 230, 240, 250, 260, 270, 280, 290, 300, 310, 320, 330, 340, 350, 360, 370, 380, 390, 400, or 405 contiguous amino acids to LP FVIIa or variants thereof); exhibits at least two non-overlapping epitopes which are specific for a LP FVIIa (or variants thereof); exhibits identity over a length of at least about 30, 35, 40, 45, 50, 55, 60, 65, 70, 75, 80, 85, 90, 95, 100, 110, 120, 130, 140, 150, 160, 170, 180, 190, 200, 210, 220, 230, 240, 250, 260, 270, 280, 290, 300, 310, 320, 330, 340, 350, 360, 370, 380, 390, 400, or 405 contiguous amino acids to a primate LP FVIIa described herein; is glycosylated; is a synthetic polypeptide; is attached to a solid substrate; is conjugated to another chemical moiety; is a 5-fold or less substitution from natural LP FVIIa sequence, or a variant thereof; or is a deletion or insertion variant from a natural LP FVIIa sequence. Various preferred embodiments include a composition comprising: a sterile LP FVIIa protein or peptide and a carrier, wherein the carrier is: an aqueous compound, including water, saline, and/or buffer; and/or formulated for oral, rectal, nasal, topical, or parenteral administration. The invention further provides a fusion protein, comprising: mature protein sequence of Table 1; wherein the mature protein sequence of Table 1 comprises a polypeptide segment of at least about: 30, 35, 40, 45, 50, 55, 60, 65, 70, 75, 80, 85, 90, 95, 100, 110, 120, 130, 140, 150, 160, 170, 180, 190, 200, 210, 220, 230, 240, 250, 260, 270, 280, 290, 300, 310, 320, 330, 340, 350, 360, 370, 380, 390, 400, or 405 contiguous amino acids from an LP sequence of Table 1, a detection or purification tag, including a FLAG, His6, or Ig sequence; or sequence of another LP FVIIa protein or peptide. These reagents also make available a kit comprising such an LP FVIIa protein or polypeptide, and: a compartment comprising the protein or polypeptide; and/or instructions for use or disposal of reagents in the kit. Providing an antigen, the invention further provides a binding compound comprising an antigen binding portion from an antibody, which specifically binds to a natural LP FVIIa protein or polypeptide, wherein: the protein or polypeptide is a primate protein; the binding compound is an Fv, Fab, or Fab2 fragment; the binding compound is conjugated to another chemical moiety; or the antibody: is raised against a peptide sequence of a mature polypeptide comprising sequence of Table 1, is raised against a mature LPFVII; is immunoselected; is a polyclonal antibody; binds to a denatured LPFVII; exhibits a Kd to antigen of at least 30 mM; is attached to a solid substrate, including a bead or plastic membrane; is in a sterile composition; or is detectably labeled, including, for example, a radioactive, enzymatic, structural, or fluorescent label. Preferred kits include those containing the binding compound, and: a compartment comprising the binding compound; and/or instructions for use or disposal of reagents in the kit. Many of the kits will be used for making a qualitative or quantitative analysis. Other preferred compositions will be those comprising: a sterile binding compound, or the binding compound and a carrier, wherein the carrier is: an aqueous compound, including water, saline, and/or buffer; and/or formulated for oral, rectal, nasal, topical, or parenteral administration. The present invention further provides an isolated or recombinant LP nucleic acid encoding a protein or peptide or fusion protein described above, wherein: the LP protein and/or polypeptide is from a mammal, including a primate; or the LP nucleic acid: encodes an antigenic peptide sequence from an LP FVIIa (or variant thereof) of Table 1; encodes a plurality of antigenic peptide sequences from an LP FVIIa of Table 1 , exhibits identity to a natural cDNA encoding the segment; is an expression vector; further comprises an origin of replication; is from a natural source; comprises a detectable label; comprises synthetic nucleotide sequence; is less than 6 kb, preferably less than 3 kb; is from a mammal, including a primate; comprises a natural full length coding sequence; is a hybridization probe for a gene encoding an LP family protein; or is a PCR primer, PCR product, or mutagenesis primer. In certain embodiments, the invention provides a cell or tissue comprising such a recombinant LP nucleic acid. Preferred cells include: a prokaryotic cell; a eukaryotic cell; a bacterial cell; a yeast cell; an insect cell; a mammalian cell; a mouse cell; a primate cell; or a human cell. Other kit embodiments include a kit comprising the described LP nucleic acid, and: a compartment comprising the LP nucleic acid; a compartment further comprising an LP FVIIa ( or a variant thereof) protein or polypeptide; and/or instructions for use or disposal of reagents in the kit. In many versions, the kit is capable of making a qualitative or quantitative analysis. Other LP FVIIa nucleic acid embodiments include those which: hybridize under wash conditions of at least 42°C, 45°C, 47°C, 50°C, 55°C, 60°C, 65°C, or 70°C and less than about 500 mM, 450 mM, 400 mM, 350 mM, 300 mM, 250 mM, 200 mM, 100 mM, to an LP nucleic acid sequence of Table 1) that exhibit identity over a stretch of at least about 30, 32, 34, 36, 38, 39, 40, 42, 44, 46, 48, 49, 50, 52, 54, 56, 58, 59, 75, or at least about 150 contiguous nucleotides to an LPFVII; in further embodiments, an LP nucleic acid embodiment which: hybridizes under wash conditions of at least 42°C, 45°C, 47°C, 50°C, 55°C, 60°C, 65°C, or 70°C and less than about 500 mM, 450 mM, 400 mM, 350 mM, 300 mM, 250 mM, 200 mM, 100 mM, to an LP nucleic acid sequence of Table 1 that exhibits identity over a stretch of at least about 30, 32, 34, 36, 38, 39, 40, 42, 44, 46, 48, 49, 50, 52, 54, 56, 58, 59, 75, or at least about 150 contiguous nucleotides to an LP FVIIa nucleic acid also encodes a protein that binds antibody generated against a mature LPFVII. In other embodiments: the wash conditions are at 55° C and/or 300 mM salt; 60° C and/or 150 mM salt; the identity is over a stretch is at least 35, 40, 45, 50, 55, 60, 65, 70, 75, 80, 85, 90, 95, 100, 110, 120, 130, 140, 150, 160, 170, 180, 190, or 200 nucleotides. In other embodiments, the invention provides a method of modulating physiology or development of a cell or tissue culture cells comprising introducing into such cell an agonist or antagonist of an LPFVII.
FIGURES Figure 1. Factor Vila induces cytokine expression in THP-1 cells:
(A) Human macrophage THP-1 cells were plated in 24 well plates in RPMI-1640 medium containing 10 % serum in the presence of 10 nM PMA to induce TF. After 24 hours, cells were washed and incubated in medium without PMA. Cells were then treated with the indicted concentrations of FVIIa. IL-6 in culture supernatants collected after 24 hour stimulation with FVIIa was determined using an ELISA. (B) For determination of the profile of cytokine induction by FVIIa, THP-1 cells were treated with 10 nM PMA as above and then with 50 nM FVIIa for 24 hours. Levels of cytokines in the supernatant were determined using a multiplex luminex assay. Inset figure IC shows the time course of TF expression in THP-1 cells after the addition of PMA. Cells extracts were prepared immediately (open bars), 24 hours (hatched bares) and 48 hours (solid bars) after treatment with PMA. For determination of IL-6 induction, control or PMA treated cells were incubated without or with 50 nM FVIIa for 0 hour (open bars), 24 hours hatched bars or 48 hours (solid bars). IL-6 was determined after incubation for the indicated interval (C).
Figure 2. Thrombin or FXa are not required for IL-6 induction by FVIIa:
THP-1 cells were treated with 10 nM PMA for 24 hours as in figure 1. Hirudin (lOOnM) or FXa (5nM) was then added. After 10 min, cells were treated with 50 nM FVIIa. IL-6 in culture supernatant collected after 24 hour treatment with FVIIa was determined as before.
Figure 3. Factor X to Xa converting activity and IL-6 inducing activity of FVIIa: (A) Factor X to Xa converting activity of recombinant FVIIa (Prep 1), freshly prepared FVIIa from human plasma (Prep 2) and FVIIa stored at -20C for 3 months ( Prep 3) was determined using an amidolytic assay. Equal concentration (14 pM) of each protein was used in the assay. The data showing the rate of change in optical density indicate FXa activity. (B) THP-1 cells were treated with 50nM of each of the three preparations and IL- 6 induction was determined as in figure legend 1. Figure 4. Identification of a N-terminal truncated FVIIa:
The three FVIIa preparations described in figure legend 3 were subjected to non-reducing SDS-PAGE. FVIIa (lug protein) from each preparation was loaded onto 4-20% SDS-gel. Gels were stained with Coomassie Gelcode staining solution (A) or transferred to PVD membrane for western blotting using a polyclonal FVIIa antibody. Lanes indicate molecular weight standards (MW), 1, 2, 3 are preparation 1, 2 and 3. (B) Shows the N- terminal amino acid sequence of the 52 kD protein band. (C) Shows the N-terminal amino acid sequence of the light chain of the 46 kD protein. The protease domain N-terminal was same as in figure B. (D) Shows the complete amino acid sequence of FVIIa with N- terminal of the light chain starting with H2N-ANAFLEE ) and the N-terminal of the protease domain (153 -H2N-IVGGKVC— ).
Figure 5: Fractionation of FVIIa and truncated FVIIa:
Preparation 2 containing the native and the truncated FVIIa was fractionated using a mono S anion exchange FPLC column. The monoS column (~x cm) was equilibrated with Tris-HCL buffer, pH 7.4. Twenty-five microliters of prep 2 was loaded to the column and then eluted with a linear gradient of 0.1M-0.5M NaCl in the equilibrating buffer. One ml fractions were collected. Column effluent was monitored for protein (absorption at 280nm) using a online detector (diamonds figure 5,). Aliquots from each fraction were subjected to SDS-PAGE and western blotting (A). (B) Each fraction was assayed for FX to Xa converting activity (triangles) and IL-8 induction in THP-1 cells (squares).
Figure 6. IL-8 induction and FX to FXa converting activity of recombinant truncated-FVUa:
FVII and truncated FVII were cloned and expressed in 293 cells. The proteins were secreted in the culture medium. Culture media from cells transfected with empty vector (control), FVII (rFVII) or truncated FVII (rTFVII) were collected and concentrated 10 fold using ultrafilteration membranes. Aliquots of the concentrated supernatants were treated with FXa. IL-6 induction by FXa untreated (open bars) or FXa treated (solid bars) supernatant from 293 cells and a native VII (reference control) was determined. Figure 7. Effect of active site inhibitors on IL-8 induction by FVIIa:
(A) THP-1 cells expressing TF (24-hour treatment with PMA) were treated with the indicated concentrations of the compounds. Factor Xa activity was then determined. (B) THP-1 cells expressing TF were treated with the indicated concentrations of compounds for 10 min before addition of 50 nM FVIIa. IL-8 in the supernatants collected after 24- hour treatment with FVIIa was then determined using an ELISA.
DETAILED DESCRIPTION OF THE PREFERRED EMBODIMENTS
I. General It is to be understood that this invention is not limited to the particular compositions, methods, and techniques described herein, as such compositions, methods, and techniques may, of course, vary. It is also to be understood that the terminology used herein is for the purpose of describing particular embodiments only, and is not intended to limit the scope of the present invention which is only limited by the appended claims. As used herein, including the appended claims, singular forms of words such as "a," "an," and "the" include, e.g., their corresponding plural referents unless the context clearly dictates otherwise. Thus, for example, reference to "an organism" includes, e.g., one or more different organisms, reference to "a cell" includes, e.g., one or more of such cells, and reference to "a method" include, e.g., reference to equivalent steps and methods known to a person of ordinary skill in the art, and so forth. Unless otherwise defined, all technical and scientific terms used herein have the same meaning as commonly understood by a person of ordinary skill in the art to which this invention belongs. Although methods and materials similar or equivalent to those described herein can be used to practice or test the present invention, suitable methods and materials are described below. AU publications, patent applications, patents, and other references discussed herein are provided solely for their disclosure before the filing date of the present application. Nothing herein is to be construed as an admission that the invention is not entitled to antedate any such disclosure by virtue of its prior invention. All publications, patent applications, patents, and other references mentioned herein are incorporated by reference in their entirety for the teachings for which they are cited (as the context clearly dictates), including all figures, drawings, pictures, graphs, hyperlinks, and other form of browser-executable code.
FEATURES OF LP FVIIa LP FVIIa (SEQ ID NO: 2) is a primate polypeptide that is a novel N-terminad truncated factor Vila, which lacks factor X activating activity but acts as a potent stimulator of inflammatory cytokine expression. LP FVIIa is generated by cleavage at amino acid residues Lys38-Leu39 leading to the deletion of the gama-carboxyglutamic acid (Gla) domain (See Table 1 below). LP FVIIa is significantly more potent inducer of cytokine expression than the native factor Vila, suggesting that the N-terminal cleavage may serve as an activation mechanism for generating a potent proinflammatory/proangiogenic form of factor Vila. The coagulation factor Vila (FVIIa) is a key initiator of the proteolytic cascade that leads to the formation of blood clot via the intrinsic pathway. The primary function of FVIIa is the proteolytic activation of factor IX and factor X (FX) after complexation with the cell associated tissue factor (TF). Activated FX (factor Xa) converts prothrombin to thrombin which in turn induces the formation of fibrin from fibrinogen (Martin, et al. 1998 Thromb. Res. 90, 1-25; Craner, et al. 1996 Thromb. Res. 81, 1-41). In addition to its crucial role in coagulation, FVIIa has been shown to act as a ligand for triggering TF mediated signaling pathway. The FVIIa-TF cellular pathway plays an important role in inflammation (Cunningham, et al. 1999 Blood 94, 3413-3420) and angiogenesis (Shoji, et al. 1998 Am. J. Pathol. 152, 399-411; Carmeliet, et al., Nature 383, 73-75). Several lines of evidence suggest that FVIIa-TF pathway is critical for blood vessel development. Gene knock out studies have demonstrated that the mutant mice lacking TF have fragile vessels (Carmeliet, et al., Nature 383, 73-75). FVIIa induces a number of pro-angiogenic genes including VEGF, Egrl, CYR61, CTGF, FGF-5, IL-8, and MMP1, MMP13 (Camerer, et al., 2000 J. Biol. Chem. 275, 6580-6585). In addition, the intracellular signaling induced by FVIIa-TF involves activation of
PI3 kinase and Akt (Versteg, et al., 2000 J. Biol. Chem. 275, 28750-28756), a pathway consistent with angiogenesis activation. The role of FVIIa-TF in tumor angiogenesis is corroborated by high level expression of TF in tumors that correlates with metastatic state (Fernandez, and Rickles, 2002 Curr. Opin. Hematol. 9. 401-406). Tissue factor is also expressed at high levels in atherosclerotic plaques where it may play pathological role in thrombosis after plaque rupture Wilcox, et al., 1989 Proc. Natl. Acad. Sci. U.S.A. 86, 2839-2843; Marmur, et al., 1996 Circ. 94, 1226-1232; and Mallat, et al., 2000 Circ. 101, 841-843). The patterns of cytokine expression in response to FVIIa-TF (IL-6, LIF, IL-8, IL-b, MMPs and CTGF) also indicate its potential role in vascular inflammation (Camerer, et al., 2000 J. Biol. Chem. 275, 6580-6585). The growth factors produced in response to FVIIa may contribute to smooth muscle cell proliferation and intimal thickening (Sato, et al., 1997 Thromb. Haemost. 78, 1138-1141). The latter is supported by experimental data demonstrating a reduction in preclinical restenosis by FVIIa or TF pathway inhibitors (Jang, et al., 1995 Circ. 92, 3041-3050). Although the studies described above have provided convincing evidence for the role of FVIIa-TF in inflammation and angiogenesis, the signal transduction pathways mediating the cellular activities of FVIIa-TF are not clearly understood. The cellular activation by FVIIa could occur indirectly through the generation of thrombin and FXa. Both thrombin and FXa are known to activate cells via the protease-activated receptors (PARs). It has been shown that induction of VEGF requires thrombin and Xa activity (OUivier, et al., 2000 Thromb. Vase. Biol. 20, 1374-1381). Other studies suggest that the cellular signaling (calcium mobilization, activation of MAP kinase and induction of gene expression) occurs in the absence of thrombin and FXa (Camerer, et al., 2000 Proc. Natl Acad. Sci. U.S.A. 97, 5255-5260). Since inactive FVIIa produced by treatment with an active site inhibitor chloromethylketone does not stimulate cell signaling, it is generally believed that proteolytic activity of FVIIa is required for cellular signaling (Camerer, et al., 2000 Proc. Natl Acad. Sci. U.S.A. 97, 5255-5260). It has been proposed that FVIIa may mediate signaling via the PAR-2 receptor (Perersen, et al., 2000 Trends Cardiovasc. Med. 10, 47-52). However, cells in which PAR-2 was desensitized, FVIIa was still active (Camerer, et al., 2000 Proc. Natl Acad. Sci. U.S.A. 97, 5255-5260). Thus, the role of PAR-2 in TF-FVIIa signaling remains unclear. It is possible that the activation of a yet unknown PAR mediates TF-VIIa cellular activities. The role of cytoplasmic domain of TF was demonstrated using cells transfected with TF gene deficient of the cytoplasmic domain. Deletion of cytoplasmic domain abrogated TF- induced metastasis (Pendurthi & Rao 2002 Vitamins and Hormones 64, 323-355; Mueller, B., M., and Ruf, W. 1998 J. Clin. Invest. 101, 1372-1378). The cellular signaling involves recruitment of filamin upon phosphorylation of the cytoplasmic domain of TF (Pendurthi & Rao 2002 Vitamins and Hormones 64, 323-355). Thus, it is still not fully established whether the protease activity of FVIIa is essential or whether TF can act as a direct signaling receptor. Consequently, Applicants demonstrate that FVIIa induces cytokine expression in human macrophage THP-1 cells in a TF dependent manner. The effect of FVIIa is independent of thrombin and FXa. The coagulation activity (generation of factor Xa from factor X) is not required for cytokine induction by FVIIa. Furthermore, the present invention discloses a novel truncated FVIIa, designated LP FVIIa, which is generated by deletion of 38 amino acid residues from an N-terminad portion of native Factor Vila. This novel truncated Factor Vila is a novel composition that is devoid of Factor X activation activity and is a significantly more potent inducer of proangiogenic cytokines than the native Factor Vila.
Table 1 : Primate, e.g., human, LP FVIIa polynucleotide sequence (SEQ ID NO: 1) and corresponding polypeptide (SEQ ID NO: 2) . The ORF for LP FVIIa is 1-1221 bp (with the start (ATG) and stop codons (TGA) identified in bold typeface and underlined in case numbering is misidentified one skilled in the art could determine the open reading frame without undue experimentation) .
LP FVIIa DMA Secruence (1221 bp) (ORF = 1-1221) :
LP FVIIa (start (atg) and stop (tga) codons are indicated in bold typeface and underlined) .
ATGGTCTCCCAGGCCCTCAGGCTCCTCTGCCTTCTGCTTGGGCTTCAGGGCTGCCTGGCTGCAGTCTTCGTA ACCCAGGAGGAAGCCCACGGCGTCCTGCACCGGCGCCGGCGCCTGTTCTGGATTTCTTACAGTGATGGGGAC CAGTGTGCCTCAAGTCCATGCCAGAATGGGGGCTCCTGCAAGGACCAGCTCCAGTCCTATATCTGCTTCTGC CTCCCTGCCTTCGAGGGCCGGAACTGTGAGACGCACAAGGATGACCAGCTGATCTGTGTGAACGAGAACGGC GGCTGTGAGCAGTACTGCAGTGACCACACGGGCACCAAGCGCTCCTGTCGGTGCCACGAGGGGTACTCTCTG CTGGCAGACGGGGTGTCCTGCACACCCACAGTTGAATATCCATGTGGAAAAATACCTATTCTAGAAAAAAGA AATGCCAGCAAACCCCAAGGCCGAATTGTGGGGGGCAAGGTGTGCCCCAAAGGGGAGTGTCCATGGCAGGTC CTGTTGTTGGTGAATGGAGCTCAGTTGTGTGGGGGGACCCTGATCAACACCATCTGGGTGGTCTCCGCGGCC CACTGTTTCGACAAAATCAAGAACTGGAGGAACCTGATCGCGGTGCTGGGCGAGCACGACCTCAGCGAGCAC GACGGGGATGAGCAGAGCCGGCGGGTGGCGCAGGTCATCATCCCCAGCACGTACGTCCCGGGCACCACCAAC CACGACATCGCGCTGCTCCGCCTGCACCAGCCCGTGGTCCTCACTGACCATGTGGTGCCCCTCTGCCTGCCC GAACGGACGTTCTCTGAGAGGACGCTGGCCTTCGTGCGCTTCTCATTGGTCAGCGGCTGGGGCCAGCTGCTG GACCGTGGCGCCACGGCCCTGGAGCTCATGGTCCTCAACGTGCCCCGGCTGATGACCCAGGACTGCCTGCAG CAGTCACGGAAGGTGGGAGACTCCCCAAATATCACGGAGTACATGTTCTGTGCCGGCTACTCGGATGGCAGC AAGGACTCCTGCAAGGGGGACAGTGGAGGCCCACATGCCACCCACTACCGGGGCACGTGGTACCTGACGGGC ATCGTCAGCTGGGGCCAGGGCTGCGCAACCGTGGGCCACTTTGGGGTGTACACCAGGGTCTCCCAGTACATC GAGTGGCTGCAAAAGCTCATGCGCTCAGAGCCACGCCCAGGAGTCCTCCTGCGAGCCCCATTTCCCTGA
LP FVIIa (406aa) ; The underlined portion is a predicted signal sequence (Met-1 to Lys-40) . A predicted SP cleavage site is between R-38 and L-39 indicated as follows: 1 MVSQALRLLCLLLGLQGCLAAVFVTQEEAHGVLHRRRRΛLF IS 43. The light chain is LF ISYSDGDQCASSPCQNGGSCKDQLQSYICFCLPAFEGRNCETHKDDQLICVNENGGCEQYCSDHTGTKR SCRCHEGYSLLADGVSCTPTVEYPCGKIPILEKRNASKPQGR indicated below by double underling. The catalytic domain is
IVGGKVCPKGECP QVLLLVNGAQLCGGTLINTIWVVSAAHCFDKIKN RNLIAVLGΞHDLSEHDGDEQSRR VAQVIIPSTYVPGTTNHDIALLRLHQPWLTDHWPLCLPΞRTFSERTLAFVRFSLVSG GQLLDRGATALE LMVLNVPRL TQDCLQQSRKVGDSPNITEYMFCAGYSDGS DSCKGDSGGPHATHYRGT YLTGIVS GQGC ATVGHFGVYTRVSQYIEWLQKLMRSEPRPGVLLRAPFP indicated below in italics.
MVSOALRLLCLLLGLOGCLAAVFVTOEEAHGVLHRRRRLF ISYSDGDOCASSPCONGGSCKDOLOS YICFCLPAFEGRNCETHKDDOLICVNENGGCEOYCSDHTGTKRSCRCHEGYSLLADGVSCTPTVEYP CGKIPl EKW^ASKVOGRIVGGKVCPKGECPWOV LLVNGAOLCGGTLINTIWVVSAAHCFDKIKNW RNLIAVLGEHDL3EHDGDEQSRRVAQVIIPSTYVPGTTNHDIALLRLHQPWLTDHWPLCLPERTF SERTLAFVRFSLV3GWGQLLDRGATALELMVLNVPRLMTQDCLQQSRKVGDSPNITEYMFCAGYSDG 3KD3CKGD3GGPHATHYRGTWYLTGIVSWGQGCATVGHFGVYTRVSQYIEWLQKLMRSEPRPGV LR APFP ClustalW Alignment of LP FVIIa & Native Factor Vila (444aa)
Deleted portion of LP FVIIIa is indicated by underlining. 1 [ - • . . : 50
LP FVIIa MVSQALRLLCLLLGLQGCLAAVFVTQEEAHGVLHRRR
VIIafull_444aa_ MVSQALRLLCLLLGLQGCLAAVFVTQEEAHGVLHRRRRANAFLEELRPGS consensus/100% MVSQALRLLCLLLGLQGCLAAVFVTQEEAHGVLHRRR consensus/80% MVSQALRLLCLLLGLQGCLAAVFVTQEEAHGVLHRR 51 . . . . 1 100
LP FVIIa RLFWISYSDGDQCASSPCQNGGSCK VIIafull_444aa_ LERECKEEQCSFEEAREIFKDAERTKLFWISYSDGDQCASSPCQNGGSCK consensus/100% +LF ISYSDGDQCASSPCQNGGSCK consensus/80% +LF ISYSDGDQCASSPCQNGGSCK 101 . . . . 150
LP FVIIa DQLQSYICFCLPAFΞGRNCETHKDDQLICVNENGGCEQYCSDHTGTKRSC VIIafull_444aa_ DQLQSYICFCLPAFEGRNCETHKDDQLICVNENGGCEQYCSDHTGTKRSC consensus/100% DQLQSYICFCLPAFEGRNCETHKDDQLICVNENGGCEQYCSDHTGTKRSC consensus/80% DQLQSYICFCLPAFEGRNCETHKDDQLICVNENGGCEQYCSDHTGTKRSC 151 . . . . 2 200
LP FVIIa RCHEGYSLLADGVSCTPTVΞYPCGKIPILEKRNASKPQGRIVGGKVCPKG VIIafull_444aa_ RCHEGYSLLADGVSCTPTVEYPCGKIPILEKRNASKPQGRIVGGKVCPKG consensus/100% RCHΞGYSLLADGVSCTPTVEYPCGKIPILEKRNASKPQGRIVGGKVCPKG consensus/80% RCHEGYSLLADGVSCTPTVEYPCGKIPILEKRNASKPQGRIVGGKVCPKG 201 . . . . : 250
LP FVIIa ECP QVLLLVNGAQLCGGTLINTI VVSAAHCFDKIKNWRNLIAVLGEHD VIIafull_444aa_ ECPWQVLLLVNGAQLCGGTLINTIWVVSAAHCFDKIKN KNLIAVLGEHD consensus/100% ECP QVLLLVNGAQLCGGTLINTI VVSAAHCFDKIKN RNLIAVLGEHD consensus/ 80% ECP QVLLLVNGAQLCGGTLINTIWVVSAAHCFDKIKNWRNLIAVLGEHD 251 . . . . 3 300
LP FVIIa LSEHDGDEQSRRVAQVIIPSTYVPGTTNHDIALLRLHQPWLTDHWPLC VIIafull_444aa_ LSEHDGDEQSRRVAQVIIPSTYVPGTTNHDIALLRLHQPWLTDHWPLC consensus/100% LSEHDGDEQSRRVAQVIIPSTYVPGTTNHDIALLRLHQPWLTDHWPLC consensus/80% LSEHDGDEQSRRVAQVIIPSTYVPGTTNHDIALLRLHQPWLTDHWPLC 301 . . . . 350
LP FVIIa LPERTFSERTLAFVRFSLVSGWGQLLDRGATALELMVLNVPRLMTQDCLQ VIIafull_444aa_ LPERTFSERTLAFVRFSLVSG GQLLDRGATALELMVLNVPRLMTQDCLQ consensus/100% LPERTFSERTLAFVRFSLVSGWGQLLDRGATALELMVLNVPRLMTQDCLQ consensus/80% LPERTFSERTLAFVRFSLVSG GQLLDRGATALELMVLNVPRL TQDCLQ 351 . . . .. 4 400
LP FVIIa QSRKVGDSPNITEYMFCAGYSDGSKDSCKGDSGGPHATHYRGT YLTGIV VIIafull_444aa_ QSRKVGDSPNITEYMFCAGYSDGSKDSCKGDSGGPHATHYRGTWYLTGIV consensus/100% QSRKVGDSPNITEYMFCAGYSDGSKDSCKGDSGGPHATHYRGTWYLTGIV consensus/80% QSRKVGDSPNITEYMFCAGYSDGSKDSCKGDSGGPHATHYRGT YLTGIV 401 . . . . ] 444
LP FVIIa SWGQGCATVGHFGVYTRVSQYIEWLQKLMRSEPRPGVLLRAPFP VIIafull_444aa_ SWGQGCATVGHFGVYTRVSQYIE LQKL RSΞPRPGVLLRAPFP consensus/100% S GQGCATVGHFGVYTRVSQYIEWLQKLMRSEPRPGVLLRAPFP consensus/80% S GQGCATVGHFGVYTRVSQYIEWLQKL RSEPRPGVLLRAPFP II. Definitions LP polynucleotide As used herein, the term "LP polynucleotide" refers to a molecule comprising a nucleic acid sequence contained in a Table herein, in a sequence of SEQ ID NO:X or Y (where X or Y are general placeholder representations for any specific DNA or amino acid sequence identifier). For example, the polynucleotide can contain the nucleotide sequence of the full length cDNA sequence, including the 5' and 3' untranslated sequences, the coding region, with or without the signal sequence, the secreted protein coding region, as well as fragments, epitopes, domains, and variants of the nucleic acid sequence. An "LP polynucleotide" also encompasses, e.g., those polynucleotides that stably hybridize, under stringent hybridization conditions, to a sequence contained in SEQ ID NO:X, or the complement thereof. In specific embodiments, an LP polynucleotide sequence is at least 15, at least 30, at least 50, at least 100, at least 125, at least 500, or at least 1200 contiguous nucleotides but are less than or equal to 300 kb, 200 kb, 100 kb, 50 kb, 15 kb, 10 kb, 7.5 kb, 5 kb, 2.5 kb, 2.0 kb, or 1 kb, in length. In a further embodiment, an LP polynucleotide sequence comprises a portion of a coding sequence, as disclosed herein, but does not comprise all or a portion of any intron. In another embodiment, the polynucleotides comprising coding sequences do not contain coding sequences of a genomic flanking gene (i.e., 5' or 3' to the sequence of interest in the genome). In other embodiments, an LP polynucleotide sequence do not contain the coding sequence of more than 1000, 500, 250, 100, 50, 25, 20, 15, 10, 5, 4, 3, 2, or 1 genomic flanking gene(s). An LP polynucleotide sequence can be composed of any polyribonucleotide or polydeoxribonucleotide, which may be unmodified RNA or DNA or modified RNA or DNA. For example, polynucleotides can be composed of single- and double-stranded DNA, DNA that is a mixture of single- and double-stranded regions, single- and double-stranded RNA, and RNA that is mixture of single- and double-stranded regions, hybrid molecules comprising DNA and RNA that may be single-stranded or, more typically, double-stranded or a mixture of single-and double-stranded regions. In addition, the polynucleotide can be composed of triple-stranded regions comprising RNA or DNA or both RNA and DNA. A polynucleotide may also contain one or more modified bases or DNA or RNA backbones modified for stability or for other reasons. "Modified" bases can include, e.g., for example, tritylated bases and unusual bases such as inosine. A variety of modifications can be made to DNA and RNA; thus, the term "polynucleotide" embraces chemically, enzymatically, or metabolically modified forms. "Altered" nucleic acid sequences encoding LP include those sequences with deletions, insertions, or substitutions of different nucleotides, resulting in a polypeptide the same as LP or a polypeptide with at least one functional characteristic of LP. Included within this definition are polymorphisms which may or may not be readily detectable using a particular oligonucleotide probe of the polynucleotide encoding LP, and improper or unexpected hybridization to allelic variants, with a locus other than the normal chromosomal locus for the polynucleotide sequence encoding LP. "Substantial similarity" in a nucleic acid sequence comparison context means either that the segments, or their complementary strands, when compared, are identical when optimally aligned, with appropriate nucleotide insertions or deletions, in at least about 50% of the nucleotides, generally at least 56%, more generally at least 59%, ordinarily at least 62%, more ordinarily at least 65%, often at least 68%, more often at least 71%, typically at least 74%, more typically at least 77%, usually at least 80%, more usually at least about 85%, preferably at least about 90%, more preferably at least about 95 to 98% or more, and in particular embodiments, as high at about 99% or more of the nucleotides. Alternatively, substantial similarity exists when the segments will hybridize under selective hybridization conditions, to a strand, or its complement, typically using a sequence derived from SEQ ID X. Typically, selective hybridization will occur when there is at least about 55% similarity over a stretch of at least about 30 nucleotides, preferably at least about 65% over a stretch of at least about 25 nucleotides, more preferably at least about 75%, and most preferably at least about 90% over about 20 nucleotides. See Kanehisa (1984) Nuc. Acids Res. 12:203-213. The length of similarity comparison, as described, may be over longer stretches, and in certain embodiments will be over a stretch of at least about 17 nucleotides, usually at least about 20 nucleotides, more usually at least about 24 nucleotides, typically at least about 28 nucleotides, more typically at least about 40 nucleotides, preferably at least about 50 nucleotides, and more preferably at least about 75 to 100 or more nucleotides, e.g., 35, 40, 45, 50, 55, 60, 65, 70, 75, 80, 85, 90, 95, 100, 110, 120, 130, 140, 150, 160, 170, 180, 190, 200, 210, 220, 230, 240, 250, 260, 270, 280, 290, 300, 310, 320, 330, 340, 350, 360, 370, 380, 390, 400, 410, 420, 430, 440, 450, 460, 470, 480, 490, 500, 510, 520, 530, 540, 550, 560, 570, 580, 590, 600, 610, 620, 630, 640, 650,660, 670, 680, 690, 700, 710, 720, 730, 740, 750, 760, 770, 780, 790, 800, 810, 820, 830, 840, 850, 860, 870, 880, 890, 900, 910, 920, 930, 940, 950, 960, 970, 980, 990,1000, 1010, 1020, 1030, 1040, 1050, 1060, 1070, 1080, 1090, 1100, 1110, 1120, 1130, 1140, 1150, 1160, 1170, 1180, 1190, 1200, 1210, 1220, 1230, 1240 or 1250, etc. For sequence comparison, typically one sequence acts as a reference sequence, to which test sequences are compared. When using a sequence comparison algorithm, test and reference sequences are input into a computer, subsequent coordinates are designated, if necessary, and sequence algorithm program parameters are designated. The sequence comparison algorithm then calculates the percent sequence identity for the test sequence(s) relative to the reference sequence, based on the designated program parameters. Optical alignment of sequences for comparison can be conducted, e.g., by the local homology algorithm of Smith and Waterman (1981) Adv. Appl. Math. 2:482, by the homology alignment algorithm of Needlman and Wunsch (1970) J. Mol. Biol. 48:443, by the search for similarity method of Pearson and Lipman (1988) Proc. Nat'l Acad. Sci. USA 85:2444, by computerized implementations of these algorithms (GAP, BESTFIT, FASTA, and TFASTA in the Wisconsin Genetics Software Package, Genetics Computer Group, 575 Science Dr., Madison, WI), or by visual inspection (see generally Ausubel et al., supra). One example of a useful algorithm is PILEUP. PILEUP creates a multiple sequence alignment from a group of related sequences using progressive, pairwise alignments to show relationship and percent sequence identity. It also plots a tree or dendogram showing the clustering relationships used to create the alignment. PILEUP uses a simplification of the progressive alignment method of Feng and Doolittle (1987) J. Mol. Evol. 35:351-360. The method used is similar to the method described by Higgins and Sharp (1989) CABIOS 5:151-153. The program can align up to 300 sequences, each of a maximum length of 5,000 nucleotides or amino acids. The multiple alignment procedure begins with the pairwise alignment of the two most similar sequences, producing a cluster of two aligned sequences. This cluster is then aligned to the next most related sequence or cluster of aligned sequences. Two clusters of sequences are aligned by a simple extension of the pairwise alignment of two individual sequences. The final alignment is achieved by a series of progressive, pairwise alignments. The program is run by designating specific sequences and their amino acid or nucleotide coordinates for regions of sequence comparison and by designating the program parameters. For example, a reference sequence can be compared to other test sequences to determine the percent sequence identity relationship using the following parameters: default gap weight (3.00), default gap length weight (0.10), and weighted end gaps. Another example of algorithm that is suitable for determining percent sequence identity and sequence similarity is the BLAST algorithm, which is described Altschul, et al. (1990) J. Mol. Biol. 215:403-410. Software for performing BLAST analyses is publicly available through the National Center for Biotechnology Information (http:www.ncbi.nlm.nih.gov/). This algorithm involves first identifying high scoring sequence pairs (HSPs) by identifying short words of length W in the query sequence, which either match or satisfy some positive- valued threshold score T when aligned with a word of the same length in a database sequence. T is referred to as the neighborhood word score threshold (Altschul, et al., supra). These initial neighborhood word hits act as seeds for initiating searches to find longer HSPs containing them. The word hits are then extended in both directions along each sequence for as far as the cumulative alignment score can be increased. Extension of the word hits in each direction are halted when: the cumulative alignment score falls off by the quantity X from its maximum achieved value; the cumulative score goes to zero or below, due to the accumulation of one or more negative-scoring residue alignments; or the end of either sequence is reached. The BLAST algorithm parameters W, T, and X determine the sensitivity and speed of the alignment. The BLAST program uses as defaults a word length (W) of 11, the
BLOSUM62 scoring matrix (see Henikoff and Henikoff (1989) Proc. Nat'l Acad. Sci. USA 89:10915) alignments (B) of 50, expectation (E) of 10, M=5, N=4, and a comparison of both strands. In addition to calculating percent sequence identity, the BLAST algorithm also performs a statistical analysis of the similarity between two sequences (see, e.g., Karlin and Altschul (1993) Proc. Nat'l Acad. Sci. USA 90:5873-5787). One measure of similarity provided by the BLAST algorithm is the smallest sum probability (P(N)), which provides an indication of the probability by which a match between two nucleotide or amino acid sequences would occur by chance. For example, a nucleic acid is considered similar to a reference sequence if the smallest sum probability in a comparison of the test nucleic acid to the reference nucleic acid is less than about 0.1, more preferably less than about 0.01, and most preferably less than about 0.001. Many proteins (and translated DNA sequences) contain regions where the amino acid composition is highly biased toward a small subset of the available residues. For example, membrane spanning domains and signal peptides (that also are membrane spanning) typically contain long stretches where Leucine (L), Valine (V), Alanine (A), and Isoleucine (I) predominate. Poly-Adenosine tracts (polyA) at the end of cDNAs appear in forward translations as poly-Lysine (poly-K) and poly-Phenylalanine (poly-F) when the reverse complement is translated. These regions are often referred to as "low complexity" regions. Such regions can cause database similarity search programs such as BLAST to find high-scoring sequence matches that do not imply true homology. The problem is exacerbated by the fact that most weight matrices (used to score the alignments generated by BLAST) give a match between any of a group of hydrophobic amino acids (L,V and I) that are commonly found in certain low complexity regions almost as high a score as for exact matches. To compensate for this, BLASTX.2 (version 2.0 aSMP-WashU) employs filters (designated "seg" and "xnu") that "mask" the low complexity regions in a particular sequence. These filters parse the sequence for such regions, and create a new sequence in which the amino acids in the low complexity region have been replaced with the character "X". This is then used as the input sequence (sometimes referred to herein as "Query" and/or "Q") to the BLASTX program. While this regime helps to ensure that high-scoring matches represent true homology, there is a negative consequence in that the BLASTX program uses the query sequence that has been masked by the filters to draw alignments. Thus, a stretch of "X's in an alignment shown in the following application does not necessarily indicate that either the underlying DNA sequence or the translated protein sequence is unknown or uncertain. Nor is the presence of such stretches meant to indicate that the sequence is identical or not identical to the sequence disclosed in the alignment of the present invention. Such stretches may simply indicate that the BLASTX program masked amino acids in that region due to the detection of a low complexity region, as defined above. A further indication that two nucleic acid sequences of polypeptides are substantially identical is that the polypeptide encoded by the first nucleic acid is irnmunologically cross reactive with the polypeptide encoded by the second nucleic acid, as described below. Thus, a polypeptide is typically substantially identical to a second polypeptide, for example, where the two peptides differ only by conservative substitutions. Another indication that two nucleic acid sequences are substantially identical is that the two molecules hybridize to each other under stringent conditions, as described below. "Homologous" polynucleotide sequences, when compared, exhibit significant similarity (e.g., sequence identity at the nucleotide level). Generally, standards for determining homology between nucleic acid molecules (or polynucleotide sequences) use art known techniques which examine, e.g., the extent of structural similarity or sequence identity between polynucleotide sequences; and/or that determine a phylogenetic relationship (e.g., whether compared sequences are orthologs or paralogs); and/or that are based on the ability of sequences to form a hybridization complex. Hybridization conditions are described in detail herein. "Hybridization" refers to the process by which a polynucleotide strand anneals with a complementary strand through base pairing under defined hybridization conditions. Specific hybridization is an indication that two nucleic acid sequences share a high degree of similarity and/or identity. Specific hybridization complexes form under permissive annealing conditions and remain hybridized after "washing." Washing is particularly important in determining the stringency of the hybridization process, typically, with more stringent conditions allowing less non-specific binding (e.g., binding between polynucleotide sequences that demonstrate less sequence identity or similarity). Permissive conditions for annealing of nucleic acid sequences are routinely determinable by one of ordinary skill in the art and may be consistent among hybridization experiments, whereas wash conditions may be varied among experiments to achieve a desired stringency, and therefore, a particular hybridization specificity. "Stringent conditions," when referring to homology or substantial similarity and/or identity in the hybridization context, will be stringent combined conditions of salt, temperature, organic solvents, and other parameters, typically those controlled in hybridization reactions. Stringent temperature conditions will usually include temperatures in excess of about 30°C, more usually in excess of about 37°C, typically in excess of about 40°C, characteristically in excess of about 42°C, routinely in excess of about 45°C, usually in excess of about 47°C, preferably in excess of about 50°C, more typically in excess of about 55°C, characteristically in excess of about 60°C, preferably in excess of about 65°C, and more preferably in excess of about 70°C. In this context, the term "about" includes, e.g., a particularly recited temperature (e.g., 50°C), and/or a temperature that is greater or lesser than that of the stated temperature by, e.g., one, two, three, four, or five degrees Celsius (e.g., 49°C or 51°C). Stringent salt conditions will ordinarily be less than about 500 mM, usually less than about 450 mM, even more usually less than about 400 mM, more usually less than about 350 mM, even more usually less than about 300 mM, typically less than about 250 mM, even more typically less than about 200 mM, preferably less than about 100 mM, and more preferably less than about 80 mM, even down to less than about 20 mM. In this context, the term "about" includes, e.g., a particularly recited molarity (e.g., 400 mM), and/or a molarity that is greater or lesser than that of the stated molarity by, e.g., three, five, seven, nine, eleven or fifteen millimolar (e.g., 389 mM or 415 mM). It is to be remembered that the combination of parameters is more important than the measure of any single parameter (see, e.g., Wetmur and Davidson (1968) J. Mol. Biol. 31:349-370). A nucleic acid probe that binds to a target nucleic acid under stringent conditions to form a stable hybridization complex is said to be specific for said target nucleic acid. Preferably, hybridization under stringent conditions should give a signal of at least 2-fold over background, more preferably a signal of at least 3 to 5 -fold over background or more. Typically, a hybridization probe is more than 11 nucleotides in length and is sufficiently identical (or complementary) to the sequence of the target nucleic acid (over the region determined by the sequence of the probe) to bind the target under stringent hybridization conditions to form a detectable stable hybridization complex. The term "hybridization complex" refers to a complex formed between two nucleic acid molecules by virtue of the formation of hydrogen bonds between complementary bases. A hybridization complex may be formed in solution (e.g., C0t or R0t analysis) or formed between one nucleic acid sequence present in solution and another nucleic acid sequence immobilized on a solid support (such as, e.g., without limitation, paper, plastic, a membrane, a filter, a chip, a pin, glass, or any other appropriate substrate to which cells or their nucleic acids can be complexed with either covalently or non-covalently). LP protein from other mammalian species can be cloned and isolated by cross- species hybridization of closely related species (as described, e.g., herein). Similarity and/or sequence identity may be relatively low between distantly related species, and thus hybridization of relatively closely related species is advisable. Alternatively, preparation of an antibody preparation that exhibits less species specificity may be useful in an expression cloning approach. Generally, stringency of hybridization is expressed, in part, with reference to the temperature under which the wash step is carried out. Such wash temperatures are typically selected to be about 5°C to 20°C lower than the thermal melting point (Tm) for the specific sequence at a defined ionic strength and pH. The Tm is the temperature (under defined ionic strength and pH) at which 50% of the target sequence hybridizes to a perfectly matched probe. An equation for calculating Tm and conditions for nucleic acid hybridization are well known (see, e.g., Sambrook, et al. (1990) Molecular Cloning: A Laboratory Manual (cur. ed.) Vol. 1-3, Cold Spring Harbor Laboratory, Cold Spring Harbor Press, NY, which is incorporated herein by reference and hereinafter referred to as "Sambrook, et al."). A non-limiting example of a high stringency condition of the invention comprises including a wash condition of 68°C in the presence of about 0.2 x SSC and about 0.1% SDS, for 1 hour. Alternatively, temperatures of about 67°C, 63°C, 61°C, 59°C, 57°C, 53°C, 51°C, 49°C, 47°C, 43°C, or 41°C may be used. SSC concentration may be varied from about 0.1 to 2X SSC, with SDS being present at about 0.1%. Typically, blocking reagents are used to block non-specific hybridization. Such blocking reagents include, for instance, sheared, and denatured salmon sperm DNA at about 100-200 Dg/ml. Organic solvent, such as, e.g., formamide at a concentration of about 35-50% v/v, may also be used under particular circumstances, such as for a RNA:DNA hybridization. Useful variations on these wash conditions will be readily apparent to those of ordinary skill in the art. Hybridization, particularly under high stringency conditions, may be suggestive of evolutionary similarity between the nucleotides. Such similarity is indicative of a similar functional and/or biological role for the nucleotide sequence and its correspondingly encoded polypeptide sequence. Another non-limiting example of a stringent hybridization condition comprises, e.g., an overnight incubation at 42°C in a solution comprising 50% formamide, 5x SSC (750 mM NaCl, 75 mM trisodium citrate), 50 mM sodium phosphate (pH 7.6), 5x Denhardt's solution, 10% dextran sulfate, and 20 pg/ml denatured, sheared salmon sperm DNA, followed by washing the filters in 0. lx SSC at about 65°C. Also contemplated are nucleic acid molecules that hybridize to an LP polynucleotide sequence at lower stringency hybridization conditions. Changes in the stringency of hybridization and signal detection can be accomplished through the manipulation of formamide concentration (lower percentages of formamide result in lowered stringency); salt conditions, or temperature. For example, an alternate stringency condition can comprise, e.g., an overnight incubation at 37°C in a solution comprising 6X SSPE (20X SSPE = 3M NaCl; 0.2M NaH,PO, 0.02M EDTA, pH 7.4), 0.5% SDS, 30% formamide, 100ml salmon sperm blocking DNA; followed by washes at 50°C with IX SSPE, 0.1% SDS. In addition, to achieve another alternate stringency condition, washes are performed following stringent hybridization at higher salt concentrations (e.g. 5X SSC). Note that variations in the above conditions may be accomplished through the inclusion and/or substitution of alternate blocking reagents used to suppress background in hybridization experiments. Typical blocking reagents include, e.g., Denhardt's reagent, BLOTTO, heparin, denatured salmon sperm DNA, and commercially available proprietary formulations. The inclusion of specific blocking reagents may require modification of a hybridization conditions described herein. A polynucleotide that hybridizes only to polyA+ sequences (such as any 3' terminal polyA+ tract of a cDNA of the invention), or to a complementary stretch of T (or U) residues, is not included, e.g., in the definition of an "LP polynucleotide" since such a polynucleotide would hybridize to any nucleic acid molecule containing a poly (A) stretch or the complement thereof (i.e., practically any double-stranded cDNA clone generated using oligo dT as a primer). Still another non-limiting example of a stringent hybridization condition is one that employs, e.g.: low ionic strength and high temperature for washing (e.g., 15mM sodium chloride/1.5 mM sodium citrate/0.1% sodium dodecyl sulfate at 50°C); a denaturing agent (during hybridization) such as formamide (e.g., 50% (v/v) formamide with 0.1% bovine serum albumin/0.1% ficoll/0.1% polyvinylpyrrolidone/50 mM sodium phosphate buffer at pH 6.5 with 750 mM sodium chloride/75 mM sodium citrate at 42°C); or 50% formamide, 5X SSC (750 mM sodium chloride, 75 mM sodium citrate), 50 mM sodium phosphate (pH 6.8), 0.1% sodium pyrophosphate, 5X Denhardt's solution, sonicated salmon sperm DNA (50 μg/mL), 0.1% SDS, and 10% dextran sulfate at 42°C with washes at 42°C in 0.2X SSC (30 mM sodium chloride/3 mM sodium citrate) and 50% formamide at 55°C, followed by a high-stringency wash consisting of 0.1X SSC containing EDTA at 55°C. An LP polynucleotide sequence of the present invention can be composed of any polyribonucleotide or polydeoxribonucleotide, which may be unmodified RNA or DNA or modified RNA or DNA. For example, polynucleotides can be composed of single- and double-stranded DNA, DNA that is a mixture of single- and double-stranded regions, single- and double-stranded RNA, and RNA that is mixture of single- and double- stranded regions, hybrid molecules comprising DNA and RNA that may be single- stranded or, more typically, double-stranded or a mixture of single-and double-stranded regions. In addition, the polynucleotide can be composed of triple-stranded regions comprising RNA or DNA or both RNA and DNA. A polynucleotide may also contain one or more modified bases or DNA or RNA backbones modified for stability or for other reasons. "Modified" bases can include, e.g., for example, tritylated bases and unusual bases such as inosine. A variety of modifications can be made to DNA and RNA; thus, the term "polynucleotide" embraces chemically, enzymatically, or metabolically modified forms. An "isolated" nucleic acid is a nucleic acid molecule or a polynucleotide sequence (e.g., an RNA, DNA, cDNA, genomic DNA, or a mixed polymer) which is substantially separated from other biologic components that naturally accompany a native sequence (e.g., proteins and flanking genomic sequences from the originating species), and thus, is altered "by the hand of man" from its natural state. In a preferable embodiment, the isolated LP sequence is free of association with components that can interfere with diagnostic or therapeutic uses for the sequence including, e.g., enzymes, hormones, and other proteinaceous or non-proteinaceous agents. The term embraces a polynucleotide sequence removed from its naturally occurring environment. For example, an isolated polynucleotide sequence could comprise part of a vector or a composition of matter, or could be contained within a cell, and still be "isolated" because the vector, composition of matter, or cell is not the original environment of the polynucleotide sequence. Moreover, the term encompasses recombinant or cloned DNA isolates, chemically synthesized analogs, or analogs biologically synthesized using heterologous systems. Furthermore, the term includes both double-stranded and single-stranded embodiments. If single- stranded, the polynucleotide sequence may be either the "sense" or the "antisense" strand. A substantially pure molecule includes isolated forms of the molecule.
An isolated nucleic acid molecule will usually contain homogeneous nucleic acid molecules, but, in some embodiments, it will contain nucleic acid molecules having minor sequence heterogeneity. Typically, this heterogeneity is found at the polymer ends or portions of the LP sequence that are not critical to a desired biological function or activity. The term "isolated" does not refer to genomic or cDNA libraries, whole cell total or mRNA preparations, genomic DNA preparations (including those separated by electrophoresis and transferred onto blots), sheared whole cell genomic DNA preparations, or other compositions where the art demonstrates no distinguishing features of a LP polynucleotide sequence of the present invention. A "recombinant" nucleic acid or polynucleotide sequence is defined either by its method of production or its structure. In reference to its method of production, e.g., a product made by a process, the process is use of recombinant nucleic acid techniques, e.g., involving human intervention in the nucleotide sequence, typically selection or production. Alternatively, it can be a nucleic acid made by generating a sequence comprising fusion of two fragments which are not naturally contiguous to each other, but is meant to exclude products of nature, e.g., naturally occurring mutants. Thus, for example, products made by transforming cells with any non-naturally occurring vector are encompassed, as are nucleic acids comprising sequence derived using any synthetic oligonucleotide process. Such is often done to replace a codon with a redundant codon encoding the same or a conservative amino acid, while typically introducing or removing a sequence recognition site. Alternatively, it is performed to join together nucleic acid segments of desired functions to generate a single engineered entity comprising a desired combination of functions not found in the commonly available natural forms. Restriction enzyme recognition sites are often the target of such artificial manipulations, but other site-specific targets may be incorporated by design (such as, e.g., promoters, DNA replication sites, regulation sequences, control sequences, and other useful features). A similar concept is intended for a recombinant polypeptide (e.g., a fusion protein). Specifically included are synthetic nucleic acid molecules which, due to the redundancy of the genetic code, encode polypeptides similar to fragments of these antigens, and fusions of sequences from various different species variants. LP protein As used herein, an "LP protein" shall encompass, when used in a protein context, a protein or polypeptide having an amino acid sequence shown in SEQ ID NO: Y or a significant fragment of such a protein or polypeptide, preferably a natural embodiment. The term "protein" or "polypeptide" is meant any chain of contiguous amino acid residues, regardless of length or postranslation modification (e.g., glycosylation, or phosphorylation). Further, an LP protein or an LP polypeptide encompass polypeptide sequences that are pre- or pro-proteins. Moreover, the present invention encompasses a mature LP protein, including a polypeptide or protein that is capable of being directed to the endoplasmic reticulum (ER), a secretory vesicle, a cellular compartment, or an extracellular space typically, e.g., as a result of a signal sequence, however, a protein released into an extracellular space without necessarily having a signal sequence is also encompassed. Generally, the polypeptide undergoes processing, e.g., cleavage of a signal sequence, modification, folding, etc, resulting in a mature form (see, e.g., Alberts, et al. (1994) Molecular Biology of The Cell, Garland Publishing, New York, NY, pp. 557-560, 582-592.). If an LP polypeptide is released into an extracellular space, it can undergo extracellular processing to produce a "mature" protein. Release into the extracellular space can occur by many mechanisms, including, e.g., exocytosis, and proteolytic cleavage. The invention also embraces polypeptides that exhibit similar structure to an LP polypeptide (e.g., one that interacts with an LP protein specific binding composition). These binding compositions, e.g., antibodies, typically bind an LP protein with high affinity, e.g., at least about 100 nM; usually, better than about 30 nM; preferably, better than about 10 nM; and more preferably, at better than about 3 nM. The term "polypeptide" or "protein" as used herein includes a "polypeptide fragment" of an LP protein or an LP polypeptide that encompasses a stretch of contiguous amino acid residues contained in SEQ ID NO: Y, or encoded by cDNA contained in a deposited clone. Protein and/or polypeptide fragments or segments may be "free-standing," or comprised within a larger polypeptide, of which the fragment or segment forms a part or region, e.g., a single continuous region. Representative examples of polypeptide fragments of the invention, include, e.g., a fragment comprising, or alternatively consisting of, from about amino acid number 1-20, 21-40, 41 -60, 61-80, 81- 100, 102-120, 121-140, 141-160, or 161 to the end of the coding region. Moreover, an LP polypeptide fragment can be about at least: 8, 10, 12, 14, 16, 18, 20, 22, 24, 26, 28, 30, 32, 34, 36, 38, 40, 42, 44, 46, 48, 50, 52, 54, 56, 58, 60, 62, 64, 66, 68, 70, 72, 74, 76, 78, 80, 82, 84, 86, 88, 90, 92, 94, 96, 98, 100, 110, 120, 130, 140, or 150 contiguous amino acids in length. In this context "about" includes, e.g., the particularly recited ranges or values, and ranges or values larger or smaller by several amino acid residues (e.g., five, four, three, two, or one) located at either extreme or at both extremes of the segment. Polynucleotides encoding such polypeptides are also encompassed by the invention. Moreover, a polypeptide comprising more than one of the above polypeptide fragments is encompassed by the invention; including a polypeptide comprising at least: one, two, three, four, five, six, seven, eight, nine, ten, or more fragments, wherein the fragments (or combinations thereof) may be of any length described herein (e.g., a fragment of 12 contiguous amino acids and another fragment of 30 contiguous amino acids, etc.). The invention also encompasses proteins or polypeptides comprising a plurality of distinct, e.g., non-overlapping, segments of specified lengths. Typically, the plurality will be at least two, more usually at least three, and preferably four, five, six, seven, eight, nine, ten, or even more. While length minima are stipulated, longer lengths (of various sizes) may be appropriate (e.g., one of length seven, and two of lengths of twelve). Features of one of the different polynucleotide sequences should not be taken to limit those of another of the polynucleotide sequences. Preferred polypeptide fragments include, e.g., the secreted protein as well as the mature form. Further preferred polypeptide fragments include, e.g., the secreted protein or the mature form having a continuous series of deleted residues from the amino or the carboxy terminus, or both. For example, 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, or 30 amino acids can be deleted from the amino terminus of either the secreted polypeptide or the mature form. Similarly, 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, or 30, can be deleted from the carboxy terminus of the secreted protein or mature form. Furthermore, any combination of the above amino and carboxy terminus deletions are preferred. Similarly, polynucleotides encoding these polypeptide fragments are also preferred. Also preferred are polypeptide and polynucleotide fragments characterized by having structural or functional domains, such as fragments that comprise, e.g., alpha-helix and alpha-helix forming regions, beta-sheet and beta-sheet-forming regions, turn and turn-forming regions, coil and coil-forming regions, hydrophilic regions, hydrophobic regions, alpha amphipathic regions, beta amphipathic regions, flexible regions, surface- forming regions, a substrate binding region, and antigenic index regions. Polypeptide fragments of SEQ ID NO: Y falling within conserved domains are specifically contemplated by the present invention. Moreover, polynucleotides encoding these domains are also contemplated. Other preferred polypeptide fragments are biologically active fragments. A polypeptide having biological activity refers to biologically active fragments or polypeptides exhibiting activity similar, but not necessarily identical to, an activity of an LP polypeptide (or fragment thereof), 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 ten-fold less activity, and most preferably, not more than about three-fold less activity relative to the polypeptide of the present invention.). The biological activity of a fragment may include, e.g., an improved desired activity, or a decreased undesirable activity. Polynucleotides encoding such polypeptide fragments are also encompassed by the invention. Any appropriate assay described herein or otherwise known in the art may routinely be applied to measure the ability of a polypeptide of the invention and a fragment, variant, derivative, and analog thereof to elicit related biological activity related to that of the polypeptide of the invention (either in vitro or in vivo). Other methods will be known to the skilled artisan and are within the scope of the invention. Furthermore, the present invention also provides a polypeptide comprising, or alternatively, consisting of, a polypeptide sequence (or fragment thereof) of at least 12 contiguous amino acid residues of a mature polypeptide SEQ ID NO: Y and/or at least a 12 contiguous amino acid residue fragment of a mature polypeptide encoded by a cDNA contained in ATCC deposit as described herein. Polynucleotides encoding a polypeptide comprising, or alternatively consisting of a polypeptide sequence of SEQ ID NO:Y and/or a polypeptide sequence encoded by a cDNA contained in ATCC deposit as described herein are also encompassed by the invention. Polynucleotides encoding such polypeptides are also encompassed by the invention. Preferably, a polynucleotide fragment of the invention encodes a polypeptide that demonstrates a functional activity. By demonstrating a "functional activity" is meant, a polypeptide having one or more known functional activities associated with a mature protein. Such functional activities include, e.g., but are not limited to, biological activity; antigenicity (an ability to bind, or compete with a polypeptide of the invention for binding, to an antibody to a polypeptide of the invention); immunogenicity (an ability to stimulate the formation of a specific and/or selective antibody which binds to a polypeptide of the invention); an ability to form multimers with a polypeptide of the invention; and an ability to specifically and/or selectively bind a binding composition of a polypeptide of the invention. A functional activity of a polypeptide of the invention (including fragments, variants, derivatives, and analogs thereof) can be assayed by various methods. For example, in one embodiment, assaying a binding (or competitive binding) ability with a full-length polypeptide of the invention for binding to an antibody of a polypeptide of the invention, various immunoassays known in the art can be used, including but not limited to, competitive and non-competitive assay systems using techniques such as radioimmunoassays, ELISA (enzyme linked immunosorbent assay), "sandwich" immunoassays, immunoradiometric assays, gel diffusion precipitation reactions, immunodiffusion assays, in situ immunoassays (e.g., using colloidal gold, enzyme or radioisotope labels), 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 another embodiment, antibody binding, e.g., is detected by detecting a label on the primary antibody. In another embodiment, a primary antibody is detected, e.g., 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. In yet another embodiment, where a ligand for a polypeptide of the invention is identified (or the ability of a polypeptide fragment, variant, or derivative of the invention to multimerize is being evaluated) binding can be assayed, e.g., by any art known method (such as, e.g., reducing and non-reducing gel chromatography, protein affinity chromatography, and affinity blotting; see, e.g., generally, Phizicky, et al. (1995)
Microbial. Rev. 59:94-123). In still yet another embodiment, physiological correlates of binding of a polypeptide of the invention to its substrates (e.g., signal transduction) can be assayed.
Modifications An LP polypeptide can be composed of amino acids joined to each other by peptide bonds or modified peptide bonds, i.e., peptide isosteres, and may contain amino acids other than the 20 gene-encoded amino acids. The polypeptides may be modified by either natural processes, such as post-translational processing, or by chemical modification techniques that are well known in the art. Such modifications are well described in basic texts and in more detailed monographs, as well as in a voluminous research literature. Modifications can occur anywhere in a polypeptide, including the peptide backbone, the amino acid side-chains and the amino or carboxyl termini. It will be appreciated that the same type of modification may be present in the same or varying degrees at several sites in a given polypeptide. Also, a given polypeptide may contain many types of modifications. Polypeptides may be branched, for example, as a result of ubiquitination, and they may be cyclic, with or without branching. Cyclic, branched, and branched cyclic polypeptides may result from posttranslation natural processes or may be made by synthetic methods. Modifications include, e.g., acetylation, acylation, ADP- ribosylation, amidation, covalent attachment of flavin, covalent attachment of a heme moiety, covalent attachment of a nucleotide or nucleotide derivative, covalent attachment of a lipid or lipid derivative, covalent attachment of phosphotidylinositol, cross-linking, cyclization, disulfide bond formation, demethylation, formation of covalent cross-links, formation of cysteine, formation of pyroglutamate, formylation, gamma-carboxylation, glycosylation, GPI anchor formation, hydroxylation, iodination, methylation, myristoylation, oxidation, pegylation, proteolytic processing, phosphorylation, prenylation, racemization, selenoylation, sulfation, transfer-RNA mediated addition of amino acids to proteins such as arginylation, and ubiquitination (see, e.g., Creighton (1993) 2nd ed. Proteins- Structure and Molecular Properties, W. H. Freeman and Company, New York; Johnson (1983) ed. Posttranslational Covalent Modification of Proteins, Academic Press, New York, pp. 1-12; Seifter et al. (1990) Meth Enzymol 182:626-646; Rattan et al. (1992) Ann NY Acad Sci 663:48XX) . The encoded protein may also be "altered," and may contain deletions, insertions, or substitutions of amino acid residues that produce a silent change and result in a functionally equivalent SECP. Deliberate amino acid substitutions may be made based on similarity in polarity, charge, solubility, hydrophobicity, hydrophilicity, and/or the amphipathic nature of the residues, as long as the biological or immunological activity of SECP is retained. For example, negatively charged amino acids may include aspartic acid and glutamic acid, and positively charged amino acids may include lysine and arginine. Amino acids with uncharged polar side chains having similar hydrophilicity values may include: asparagine and glutamine; and serine and threonine. Amino acids with uncharged side chains having similar hydrophilicity values may include: leucine, isoleucine, and valine; glycine and alanine; and phenylalanine and tyrosine. "Substantially pure" refers to LP nucleic acid or LP protein or polypeptide that are removed from their natural environment and are isolated and/or separated from other contaminating proteins, nucleic acids, and other biologicals. Purity, or "isolation" may be assayed by standard methods, and will ordinarily be at least about 50% pure, more ordinarily at least about 60% pure, generally at least about 70% pure, more generally at least about 80% pure, often at least about 85% pure, more often at least about 90% pure, preferably at least about 95% pure, more preferably at least about 98% pure, and in most preferred embodiments, at least 99% pure. Similar concepts apply, e.g., to LP antibodies or nucleic acids of the invention. For example, it may be desirable to purify an LP polypeptide from recombinant cell proteins or polypeptides. Typical exemplary suitable purification procedures include, e.g., without limitation, fractionation on an ion-exchange column; ethanol precipitation; reversed-phase HPLC; chromatography on silica or cation- exchange resins (such as, e.g., DEAE); chromatofocusing; SDS-PAGE; ammonium sulfate precipitation; gel filtration (using, e.g., Sephadex G-75); protein A Sepharose columns (e.g., to remove contaminants such as IgG); and metal chelating columns (e.g., to bind epitope-tagged forms of an LP polypeptide). Various art known methods of protein purification may be employed (see, e.g., Deutscher, (1990) Methods in
Enzymology 182: 83-9 and Scopes, (1982) Protein Purification: Principles and Practice, Springer- Verlag, NY.) Typicaly, the purification method selected depends, e.g., on the nature of the production process used and the particular LP polypeptide produced. In another example, a chimeric LP protein comprising a heterologous moiety, which can be recognized by another molecule, can be purified using a commercially available affinity matrix. Such moieties include, without limit, glutathione S-transferase (GST), maltose binding protein (MBP), thioredoxin (Trx), calmodulin binding peptide (CBP), 6-His, FLAG, c-myc, and hemagglutinin (HA). The GST, MBP, Trx, CBP, and 6- His moieties enable purification of their cognate fusion proteins on immobilized glutathione, maltose, phenylarsine oxide, calmodulin, and metal-chelate resins, respectively. The FLAG, c-myc, and hemagglutinin (HA) moieties enable immunoaffinity purification of fusion proteins using commercially available monoclonal and polyclonal antibodies that specifically recognize these epitope tags. "Solubility" of an LP protein or polypeptide is reflected by sedimentation measured in Svedberg units, which are a measure of the sedimentation velocity of a molecule under particular conditions. The determination of the sedimentation velocity was classically performed in an analytical ultracentrifuge, but is typically now performed in a standard ultracentrifuge (see, Freifelder (1982) Physical Biochemistry (2d ed.) W.H. Freeman & Co., San Francisco, CA; and Cantor and Schimmel (1980) Biophysical Chemistry parts 1-3, W.H. Freeman & Co., San Francisco, CA). As a crude determination, a sample containing a putatively soluble polypeptide is spun in a standard full sized ultracentrifuge at about 5 OK rpm for about 10 minutes, and soluble molecules will remain in the supernatant. A soluble particle or polypeptide will typically be less than about 30S, more typically less than about 15S, usually less than about 10S, more usually less than about 6S, and, in particular embodiments, preferably less than about 4S, and more preferably less than about 3S. Solubility of a polypeptide or fragment depends upon the environment and the polypeptide. Many parameters affect polypeptide solubility, including temperature, electrolyte environment, size and molecular characteristics of the polypeptide, and nature of the solvent. Typically, the temperature at which the polypeptide is used ranges from about 4° C to about 65° C. Usually the temperature at use is greater than about 18° C and more usually greater than about 22° C. For diagnostic purposes, the temperature will usually be about room temperature or warmer, but less than the denaturation temperature of components in the assay. For therapeutic purposes, the temperature will usually be body temperature, typically about 37° C for humans, though under certain situations the temperature may be raised or lowered in situ or in vitro. The size and structure of the polypeptide should generally be in a substantially stable state, and usually not in a denatured state. The polypeptide may be associated with other polypeptides in a quaternary structure, e.g., to confer solubility, or associated with lipids or detergents in a manner which approximates natural lipid bilayer interactions. The solvent will usually be a biologically compatible buffer, of a type used for preservation of biological activities, and will usually approximate a physiological solvent. Usually the solvent will have a neutral pH, typically between about 5 and 10, and preferably about 7.5. On some occasions, a detergent will be added, typically a mild non- denaturing one, e.g., CHS (cholesteryl hemisuccinate) or CHAPS (3-[3- cholarnidopropyl)-dimethylammonio]-l -propane sulfonate), or a low enough concentration as to avoid significant disruption of structural or physiological properties of the protein.
Signal Sequence The present invention encompasses "mature" forms of a polypeptide comprising a polypeptide sequence listed in a Table herein, a polypeptide sequence of SEQ ID NO: Y, or a polypeptide sequence encoded by a cDNA in a deposited clone. Polynucleotides encoding a mature form (such as, e.g., an LP polynucleotide sequence listed in a Table herein, an LP polypeptide sequence of SEQ ID NO: X and/or a polynucleotide sequence contained in the cDNA of a deposited clone) are also encompassed by the invention. According to the signal hypothesis, proteins secreted by mammalian cells have a signal or secretary leader sequence that is cleaved off before export of the growing polypeptide chain across the rough endoplasmic reticulum has been completed. Most mammalian cells (and even insect cells) cleave secreted proteins with the same specificity. However, in some cases, cleavage of a secreted protein is not entirely uniform resulting in two or more mature species of the protein. All such forms are encompassed herein. Methods for predicting whether a protein has a signal sequence, as well as the cleavage point for that sequence, are known in the art (McGeoch, (1985) Virus Res. 3:271-286), e.g., using information from a short N-terminal charged region and a subsequent uncharged region of the complete (uncleaved) protein (von Heinje, (1986) Nucleic Acids Res. 144683-4690) using information from residues surrounding the cleavage site, typically residues -13 to +2, where + 1 indicates the amino terminus of the secreted protein, or a deduced amino acid sequence of a secreted polypeptide can be analyzed using a computer program called SignalP which predicts the cellular location of a protein based on the amino acid sequence (Henrik Nielsen et al. (1997) Protein Engineering 10: 1-6). The accuracy of predicting cleavage points of known mammalian secretory proteins typically is about 75-80%, however, not all art methods produce the same predicted cleavage point(s) for a given protein (incorporated herein by reference for these techniques) . Employing such known art methods a signal sequence for an LP polypeptide was made. However, as one of ordinary skill would appreciate, cleavage sites may vary from organism to organism and cannot be predicted with absolute certainty. Accordingly, the present invention provides secreted LP polypeptides having a sequence listed in a Table herein, or a polypeptide sequence of SEQ ID NO: Y, or a polypeptide sequence encoded by a cDNA in a deposited clone in which a particular N-terminus variant polypeptide sequence can begin within five, four, three, two, or one amino acid residues (e.g., +5, +4, +3, +2, +1, or -5, -4, -3, -2, -1) from a particular cleavage point that is designated as such herein. Similarly, it is also recognized that in some cases, cleavage of a signal sequence of a secreted protein is not uniform, resulting in more than one secreted species for a given protein (e.g., a cleavage variant). Such cleavage variant LP polypeptides, and the polynucleotides encoding them, are also encompassed by the present invention. Moreover, the signal sequence identified by the above analysis may not necessarily predict a naturally occurring signal sequence. For example, a naturally occurring signal sequence may be further upstream from a predicted signal sequence. However, it is likely that a predicted signal sequence will be capable of directing the secreted protein to the ER. Nevertheless, the present invention encompasses a mature LP polypeptide or protein produced by expression of a polynucleotide sequence listed in a Table herein, an LP polynucleotide sequence of SEQ ID NO: X, or an LP polynucleotide sequence contained in a cDNA of a deposited clone, in a mammalian cell (e.g., a COS cell, as described). These LP polypeptides (fragments thereof), and the polynucleotides encoding them, are also encompassed by the present invention.
LP Variants The present invention encompasses variants of an LP polynucleotide sequence disclosed in SEQ ID NO: X, the complementary strand thereto, and/or a cDNA sequence contained in a deposited clone. The present invention also encompasses variants of a polypeptide sequence disclosed in SEQ ID NO: Y and/or encoded by a deposited clone. The term "variant" refers to a polynucleotide or polypeptide differing from an LP polynucleotide sequence or an LP polypeptide of the present invention, but retaining essential properties thereof. Generally, variants are closely similar overall in structural and/or sequence identity, and, in many regions, identical to an LP polynucleotide or polypeptide of the present invention. The present invention encompasses nucleic acid molecules that comprise, or alternatively consist of, a polynucleotide sequence that is at least: 80%, 85%, 90%, 95%, 96%, 97%, 98%, or 99% identical to, e.g., a polynucleotide coding sequence of SEQ ID NO: X (or a strand complementary thereto); a nucleotide sequence contained in a deposited cDNA clone (or a complementary strand thereto); a nucleotide sequence encoding a polypeptide of SEQ ID NO: Y; a nucleotide sequence encoding a polypeptide encoded by the cDNA contained in a deposited clone; and/or polynucleotide fragments of any of these nucleic acid molecules (e.g., a fragment as defined herein). Polynucleotides, that stably hybridize to a polynucleotide fragment (as defined herein) under stringent hybridization conditions or lower stringency conditions, are also encompassed by the invention, as are polypeptides (or fragments thereof) encoded by these polynucleotides. The present invention is also directed to polypeptides that comprise, or alternatively consist of, an amino acid sequence that is at least: 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99% identical to, e.g., a polypeptide sequence of SEQ ID NO: Y (or fragments thereof); a polypeptide sequence encoded by a cDNA contained in a deposited clone, and/or a polypeptide fragment of any of these polypeptides (e.g., those fragments as defined herein). A polynucleotide sequence having at least some "percentage identity," (e.g., 95%) to another polynucleotide sequence, means that the sequence being compared (e.g., the test sequence) may vary from another sequence (e.g. the referent sequence) by a certain number of nucleotide differences (e.g., a test sequence with 95% sequence identity to a reference sequence can have up to five point mutations per each 100 contiguous nucleotides of the referent sequence). In other words, for a test sequence to exhibit at least 95% identity to a referent sequence, up to 5% of the nucleotides in the referent may differ, e.g., be deleted or substituted with another nucleotide, or a number of nucleotides (up to 5% of the total number of nucleotides in the reference sequence) may be inserted into the reference sequence. The test sequence may be: an entire polynucleotide sequence, e.g., as shown in Tables 1-18, the ORF (open reading frame), or any fragment, segment, or portion thereof (as described herein). As a practical matter, determining if a particular nucleic acid molecule or polynucleotide sequence exhibits at least about: 80%, 85%, 90%, 95%, 96%, 97%, 98% or 99% identity to an LP polynucleotide sequence can be accomplished using known computer programs. Typically, in such a sequence comparison, one sequence acts as a reference sequence, to which test sequences are compared. When using a sequence comparison algorithm, test and reference sequences are input into a computer, subsequent coordinates are designated, if necessary, and sequence algorithm program parameters are designated. The sequence comparison algorithm then calculates the percentage sequence identity for a test sequence(s) relative to the reference sequence, based on the parameters of a designated program. Optimal alignment of sequences for comparison can be conducted, e.g., by the local homology algorithm of Smith and Waterman (1981) Adv. Appl. Math. 2:482, by the homology alignment algorithm of Needlman and Wunsch (1970) J. Mol. Biol. 48:443, by the search for similarity method of Pearson and Lipman (1988) Proc. Nat'l Acad. Sci. USA 85:2444, by computerized implementations of these algorithms (GAP, BESTFIT, FASTA, and TFASTA in the Wisconsin Genetics Software Package, Genetics Computer Group, 575 Science Dr., Madison, WI), or by visual inspection (see generally, Ausubel, et al. supra). A typical method for determining , a best overall match (also referred to as a global sequence alignment) between a test and a referent sequence can be determined using , e.g., the FASTDB computer program based on the algorithm of Brutlag, et al. (1990) Comp. App. Biosci. 6: 237-245. In a FASTDB sequence alignment, the test and referent sequences are, e.g., both DNA sequences. An RNA sequence can be compared by converting U's to T's. The result of a global sequence alignment is given in terms of a percentage identity. Typical parameters used in a FASTDB alignment of DNA sequences to calculate percent identity are, e.g.,
Figure imgf000038_0001
k-tuple=4, Mismatch Penalty=l, Joining Penalty=30, Randomization Group Length=0, Cutoff Score=l, Gap Penalty=5, Gap Size Penalty 0.05, Window Size=500, or the length of the referent nucleotide sequence, whichever is shorter. If the referent sequence is shorter than the test sequence because of 5' or 3' 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 5' and 3' truncations of the subject sequence when calculating percent identity. For referent sequences truncated at the 5' or 3' ends, relative to the test sequence, the percentage identity is corrected by calculating the number of bases of the test sequence that are 5 ' and 3' of the subject sequence, which are not matched/aligned, as a percentage of the total bases of the test sequence. Whether a nucleotide is matched/aligned is determined by results of the FASTDB sequence alignment. This percentage is then subtracted from the percentage identity, calculated by the above FASTDB program using the specified parameters, to arrive at a final percentage identity score. The corrected score is what is used for the purposes of sequence identity for the present invention. Ordinarily, bases outside the 5' and 3' bases of the subject sequence, as displayed by the FASTDB alignment, which are not matched/aligned with the test sequence, are calculated for the purposes of manually adjusting the percent identity score. For example, a 90 base referent sequence is aligned to a 100 base test sequence to determine percentage identity. The deletions occur at the 5' end of the referent sequence and therefore, the FASTDB alignment does not show a matched/alignment of the first 10 bases at the 5' end. The 10 unpaired bases represent 10% of the sequence (number of bases at the 5' and 3' ends not matched/total number of bases in the test sequence) so 10% is subtracted from the percentage identity score calculated by the FASTDB program. If the remaining 90 bases were perfectly matched the final percentage identity would be 90%. In another example, a 90 base referent sequence is compared with a 100 base test sequence. This time the deletions are internal deletions so that there are no bases on the 5' or 3' of the subject sequence, which are not matched/aligned with the test. In this case, the percentage identity calculated by FASTDB is not manually corrected. Again, only bases 5' and 3' of the subject sequence that are not matched/aligned with the test sequence are manually corrected for. No other manual corrections are to made for the purposes of the present invention. Especially preferred are polynucleotide variants containing alterations, which produce silent substitutions, additions, or deletions, but do not alter the properties or activities of the encoded polypeptide. Nucleotide variants produced by silent substitutions due to the degeneracy of the genetic code are preferred. Polynucleotide variants can be produced for a variety of reasons, e.g., to optimize codon expression for a particular host (change codons in the human mRNA to those preferred by a bacterial host such as E. coli). A further indication that two nucleic acid sequences of polypeptides are substantially identical is that the polypeptide encoded by the first nucleic acid is immunologically cross reactive with the polypeptide encoded by the second nucleic acid, as described herein. Thus, a polypeptide is typically substantially identical to a second polypeptide, for example, where the two peptides differ only by conservative substitutions. Another indication that two nucleic acid sequences are substantially identical is that the two molecules hybridize to each other under stringent conditions, as described below. A polypeptide exhibiting or having at least about, e.g., 95% "sequence identity" to another amino acid sequence may include, e.g., up to five amino acid alterations per each 100 amino acid stretch of the test amino acid sequence. In other words, a first amino acid sequence that is at least 95% identical to a second amino acid sequence, can have up to 5% of its total number of amino acid residues different from the second sequence, e.g., by insertion, deletion, or substitution of an amino acid residue. Alterations in amino residues of a polypeptide sequence may occur, e.g., at the amino or carboxy terminal positions or anywhere between these terminal positions, interspersed either individually among residues in the sequence or in one or more contiguous amino residue sections, portions, or fragments within the sequence. As a practical matter, whether any particular polypeptide sequence exhibits at least about: 80%, 85%, 90%, 95%, 96%, 91%, 98%, or 99% similarity to another sequence, for example, such as shown in Table 1 (SEQ ID NO:Y) or to an amino acid sequence encoded by a cDNA contained in a deposited clone, can be determined conventionally by using known methods in the art, e.g., a computer algorithm such as ClustalW. A preferred method for determining the best overall match (also called a global sequence alignment) between two sequences (either nucleotide or amino acid sequences) uses the FASTDB algorithm of Brutlag, et al. (1990) Comp. App. Biosci. 6:237-245. The result of such a global sequence alignment is given as a percentage of sequence identity, e.g., with 100% representing complete sequence identity. Typical FASTDB parameters for amino acid alignments are, e.g.,:
Figure imgf000040_0001
0, k-tuple=2, Mismatch Penalty=l, Joining Penalty=20, Randomization Group Length=O, Cutoff Score=l, Window Size=sequence length, Gap Penalty=5 Gap Size Penalty=0.05, Window Size=500 or the length of the subject amino acid sequence, whichever is shorter. If the subject sequence is shorter than the test 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 test sequence, the percent identity is corrected by calculating the number of residues of the test 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 test 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 percentage- identity score. This final percentage identity score is what is used for the purposes of the present invention. Only residues to the N- and C-termini of the subject sequence, which are not matched/aligned with the test sequence, are considered for the purposes of manually adjusting the percent identity score. That is, only test residue positions outside the farthest N- and C-terminal residues of the subject sequence. For example, a 90 amino acid residue subject sequence is aligned with a 100-residue test sequence to determine percent identity. The deletion occurs at the N-terminus of the subject sequence and therefore, the FASTDB alignment does not show a matching/alignment of the first 10 residues at the N-terminus. The unpaired residues represent 10% of the sequence (number of residues at the N- and C-termini not matched/total number of residues in the test sequence) so 10% is subtracted from the percent identity score calculated by the FASTDB program. If the remaining 90 residues were perfectly matched the final percent identity would be 90%. In another example, a 90-residue subject sequence is compared with a 100-residue test sequence. This time the deletions are internal deletions so there are no residues at the N- or C-termini of the subject sequence, which are not matched/aligned with the test. In this case, the percent identity calculated by FASTDB is not manually corrected. Once again, only residue positions outside the N- and C-terminal ends of the subject sequence, as displayed in the FASTDB alignment, which are not matched/aligned with the test sequence are manually corrected for. No other manual corrections are to made for the purposes of the present invention. Variants encompassed by the present invention may contain alterations in the coding regions, non-coding regions, or both. Moreover, variants in which 1-2, 1-5, or 5-10 amino acids are substituted, deleted, or added in any combination are also preferred. Naturally occurring variants encompassed herein are "allelic variants," which refer to one of several alternate forms of a gene occupying a given locus on a chromosome of an organism. Allelic variants can vary at either the polynucleotide and/or polypeptide level and both types of variants are encompassed by the present invention. Alternatively, non-naturally occurring variants may be produced by mutagenesis techniques or by direct synthesis using known methods of protein engineering and recombinant DNA technology. Such variants may be generated to improve or alter the characteristics of an LP polypeptide (or fragment thereof). For instance, one or more amino acids can be deleted from the N-terminus or C- terminus of a secreted polypeptide of the invention (or fragment thereof) without a substantial loss of biological function. For example, Ron, et al. (1993) J. Biol. Chem. 268: 2984-2988, reported variant KGF proteins having heparin binding activity even after deleting 3, 8, or 27 ammo-teπninal amino acid residues. Similarly, interferon gamma was shown to exhibit up to ten times increased activity after 8-10 amino acid residues were deleted from the carboxy terminus (Dobeli, et al. (1988) J. Biotechnology 7:199-216) Moreover, ample evidence demonstrates that polypeptide or polynucleotide variants retain a biological activity similar to that of the naturally occurring protein. For example, Gayle, et al. (1993) J. Biol. Chem 268:22105-22111, conducted extensive mutational analysis of human cytokine IL-1 alpha using random mutagenesis to generate over 3,500 individual IL-1 alpha mutants that averaged (over the entire length of the molecule) 2.5 amino acid changes per variant. Multiple mutations were examined at every possible amino acid position. The results showed that most of the molecule could be altered with little effect on either binding or biological activity. In fact, out of more than 3,500 nucleotide sequences examined, only 23 amino acid sequences produced a protein that differed significantly in activity from the wild-type. Moreover, even if deleting one or more amino acids from the N-terminus or C-terminus of the polypeptide results in modification or loss of one or more biological functions, other biological activities may be retained. For example, antigenicity and/or immunogenicity can be retained (e.g., the ability of a deletion variant to induce and/or to bind antibodies that recognize a mature form of a polypeptide) when less than the majority of the residues of the secreted form are removed from the N-terminus or C-terminus. Whether a polypeptide lacking N- or C-terminal residues of a protein retains such activities can readily be determined by routine methods such as those described herein or known in the art. Thus, the invention also encompasses, e.g., polypeptide variants that show biological activity such as, e.g., immunogenicity, or antigenicity. Such variants include, e.g., deletions, insertions, inversions, repeats, and substitutions selected so as have little effect on activity using general rules known in the art. For example, teachings on making phenotypically silent amino acid substitutions are provided, e.g., by Bowie, et al. (1990) Science 247: 1306-1310 One technique compares amino acid sequences in different species to identify the positions of conserved amino acid residues since changes in an amino acid at these positions are more likely to affect a protein function. In contrast, the positions of residues where substitutions are more frequent generally indicates that amino acid residues at these positions are less critical for a protein function. Thus, to a first degree, positions tolerating amino acid substitutions typically may be modified while still maintaining a biological activity of a protein. A second technique uses genetic engineering to introduce amino acid changes at specific positions of a polypeptide to identify regions critical for a protein function. For example, site directed mutagenesis or alanme-scanning mutagenesis (the introduction of single alanine mutations at every residue in the molecule) can be used. (Cunningham and Wells (1989) Science 244: 1081-1085) A resulting mutein can subsequently be tested for a biological activity. These two techniques have revealed that proteins are surprisingly tolerant of amino acid substitutions and they generally indicate which amino acid changes are likely to be permissive at certain amino acid positions in a protein. For example, typically, most buried amino acid residues (those within the tertiary structure of the protein) require nonpolar side chains, whereas few features of surface side chains are generally conserved. Moreover, tolerated conservative amino acid substitutions involve replacement of the aliphatic or hydrophobic amino acids Ala, Val, Leu and He; replacement of the hydroxyl residues Ser and Thr; replacement of the acidic residues Asp and Glu; replacement of the amide residues Asn and Gin, replacement of the basic residues Lys, Arg, and His; replacement of the aromatic residues Phe, Tyr, and Trp, and replacement of the small- sized amino acids Ala, Ser, Thr, Met, and Gly. Besides using conservative amino acid substitutions, other variants of the present invention include, e.g., but are restricted to, e.g., (i) substitutions with 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) substitution with one or more of amino acid residues having a substituent group, or (iii) fusion of the mature polypeptide with another compound, such as a compound to increase the stability and/or solubility of the polypeptide (e.g., polyethylene glycol), or (iv) fusion of the polypeptide with additional amino acids, such as, e.g., an IgG Fc fusion region peptide, or leader or secretory sequence, or a sequence facilitating purification. All such variants would be within the scope of those skilled in the art of molecular biology given Applicants' teachings herein, e.g., specifying unique polynucleotide and polypeptide sequences. For example, polypeptide variants containing amino acid substitutions of charged amino acids with other charged or neutral amino acids may produce polypeptides with improved characteristics e.g., such as less aggregation. Aggregation of pharmaceutical formulations both reduces activity and increases clearance due to the aggregate's immunogenic activity (Pinckard, et al. (1967) Clin. Exp. Immunol. 2:331-340; Robbins, et al. (1987) Diabetes 36:838-845; Cleland, et al. (1993) Crit. Rev. Therapeutic Drug Carrier Systems 10:307-377). A further embodiment of the invention encompasses a protein that comprises an amino acid sequence of the present invention that contains at least one amino acid substitution, but not more than 50 amino acid substitutions, even more preferably, not more than 40 amino acid substitutions, still more preferably, not more than 30 amino acid substitutions, and still even more preferably, not more than 20 amino acid substitutions, nor more than 15 amino acid substitutions. Of course, in order of ever-increasing preference, it is highly preferable for a peptide or polypeptide to have an amino acid sequence that comprises an amino acid sequence of the present invention, which contains at least: one, but not more than: 10, 9, 8, 7, 6, 5, 4, 3, 2, or 1 amino acid substitutions. In specific embodiments, the number of additions, substitutions, and/or deletions in an polypeptide sequence of the present invention or fragments thereof (e.g., a mature form and/or other fragments described herein), is at least: 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 10-50, or 50-150; wherein conservative amino acid substitutions are more preferable than non-conservative substitutions.
LP Polynucleotide and LP Polypeptide Fragments The present invention is also directed to fragments of an LP polynucleotide. An
LP polynucleotide "fragment" encompasses a short polynucleotide of a nucleic acid molecule, which is a portion of a sequence contained in a deposited clone, or encoding a polypeptide encoded by a cDNA in a deposited clone; or a portion of a polynucleotide sequence of SEQ ID NO: X or a complementary strand thereto, or a portion of a polynucleotide sequence encoding a polypeptide of SEQ ID NO: Y (or fragment thereof). Polynucleotide fragments of the invention encompass a polynucleotide sequence that is preferably at least about 15 nucleotides, more preferably at least about: 20, 21, 22, 24, 26, or 29 nucleotides, favorably at least about: 30, 32, 34, 36, 38, or 39 nucleotides, and even more preferably, at least about: 40, 42, 44, 46, 48, or 49 nucleotides, desirably at least about: 50, 52, 54, 56, 58, or 59 nucleotides, particularly at least about 75 nucleotides, or at least about 150 nucleotides in length. A polynucleotide fragment "at least 20 nucleotides in length," e.g., is intended to include, e.g., 20 or more contiguous bases from the cDNA sequence contained in a deposited clone or the nucleotide sequence shown in SEQ ID NO: X. In this context "at least about" includes, e.g., a specifically recited value (e.g., 20nt), and a value that is larger or smaller by one or more nucleotides (e.g., 5, 4, 3, 2, or 1), at either terminus or at both termini. A polynucleotide fragment has use that includes without limit; e.g., diagnostic probes and primers as discussed herein. Larger fragments (e.g., 50, 150, 500, 600, or 2000 nucleotides) are also useful and preferred. Representative examples of various lengths of polynucleotide fragments encompassed by the invention, include, e.g., fragments comprising, or alternatively consisting of, a polynucleotide sequence of SEQ ID NO:X from about nucleotide number 1-50, 51-100, 101-150, 151-200, 201-250, 251-300, 301-350, 351-400, 401-450, 451-500, 501-550, 551-600, 651-700, 701-750, 751-800, 800-850, 851-900, 901-950, 951-1000, 1001-1050, 1051-1100, 1101-1150, 1151-1200, 1201-1250, 1251-1300, 1301-1350, 1351-1400, 1401-1450, 1451-1500, 1501-1550, 1551-1600, 1601-1650, 1651-1700, 1701-1750, 1751-1800, 1801-1850, 101851-1900, 1901-1950, 1951-2000, or 2001 to the end of SEQ ID NO:X, or a strand complementary thereto, or a cDNA contained in a deposited clone. In this context, the term "about" includes, e.g., a particularly recited polynucleotide fragment range herein, and/or ranges that have lengths that are larger or smaller by several nucleotides (e.g., 5, 4, 3, 2, or lnt), at either terminus or at both termini. Preferably, these fragments encode a polypeptide possessing biological activity as defined herein, e.g., immunogenicity, or antigenicity. More preferably, a polynucleotide fragment can be used as a probe or primer as discussed herein. Furthermore, the present invention also encompasses a polynucleotide that stably hybridizes to a polynucleotide fragment described herein under either stringent or lowered stringency hybridization conditions. Additionally incorporated are polypeptides encoded by a polynucleotide fragment or a hybridized polynucleotide stably bound to a polynucleotide fragment of the invention. Additionally encompassed by the invention is a polynucleotide encoding a polypeptide, which are specifically or selectively bound by an antibody directed to/or generated against a mature polypeptide of the invention (or fragment thereof), e.g., a mature polypeptide of SEQ ID NO: Y. In the present invention, a "polypeptide fragment or segment" encompasses an amino acid sequence that is a portion of SEQ ID NO: Y or a polypeptide segment encoded by a cDNA contained in a deposited clone. Protein and/or polypeptide fragments or segments may be "free-standing," or they may comprise part of a larger polypeptide or protein, of which the fragment or segment forms a portion or region, e.g., a single continuous region of SEQ ID NO: Y connected in a fusion protein. Representative examples of lengths of polypeptide fragments or segments encompassed by the invention, include, e.g., fragments comprising, or alternatively consisting of, from about amino acid residue number 1-20, 21-40, 41-60, 61-80, 81-100, 102-120, 121-140, 141-160, 161-170, 171-180, 181-190, 191-200, 201-210, etc., to the end of the mature coding region of a polypeptide of the invention (or fragment thereof). Preferably, a polypeptide segment of the invention can have a length of contiguous amino acids of a polypeptide of the invention (or fragment thereof) that is at least about: 7, 8, 9, 10, 12, 14, 16, 18, 20, 22, 24, 26, 28, 30, 32, 34, 36, 38, 40, 42, 44, 46, 48, 50, 52, 54, 56, 58, 60, 62, 64, 66, 68, 70, 72, 74, 76, 78, 80, 82, 84, 86, 88, 90, 92, 94, 96, 98, 100, 110, 120, 130, 140, or 150 contiguous amino acids in length. In this context "about" includes, e.g., the specifically recited ranges or values described herein, and it also encompasses values that differ from these recited values by several amino acid residues (e.g., plus or minus 5,. plus or minus 4, plus or minus 3, plus or minus 2, or; plus or minus 1 amino acid residues), at either or both ends of the fragment. Further, a polynucleotide encoding a polypeptide such a fragment is also encompassed by the invention. Moreover, the invention encompasses proteins or polypeptides comprising a plurality of said amino acid segments or fragments, e.g., nonoverlapping, segments of a specified length. Typically, a plurality will be at least two, more usually at least three, and preferably at least: four, five, six, seven, eight, nine, or more. While minimum lengths of a segment are provided, maximum lengths of various sizes are also encompassed for any specific plurality of segments, e.g., a plurality of three segments in toto could have one segment of length 7 contiguous amino acids, and two additional non- overlapping segments, each of which has a length of 12. Features of one of the different genes should not be taken to limit those of another of the genes. Preferred polypeptide fragments include, e.g., the secreted protein, as well as the mature form. Further preferred polypeptide fragments include, e.g., the secreted protein or the mature form having a continuous series of deleted residues from the amino or the carboxy terminus, or both. For example, a number of amino acids, ranging from 1-30, can be deleted from the amino terminus of either the secreted polypeptide or the mature form. Similarly, a number of amino acids, ranging from 1-30, can be deleted from the carboxy terminus of the secreted protein or mature form. Furthermore, any combination of the above amino and carboxy terminus deletions are preferred. Similarly, polynucleotides encoding these polypeptide fragments are also preferred. Also preferred are polypeptide fragments or segments (and their corresponding polynucleotide fragments) that characterize structural or functional domains, such as, fragments, or combinations thereof, that comprise e.g., alpha-helix, and alpha-helix forming regions, beta-sheet, and beta-sheet-forming regions, turn, and turn-forming regions, coil, and coil-forming regions, hydrophilic regions, hydrophobic regions, alpha amphipathic regions, beta amphipathic regions, flexible regions, loop regions, hairpin domains, beta-alpa-beta motifs, helix bundles, alpha/beta barrels, up and down beta barrels, jelly roll or swiss roll motifs, transmembrane domains, surface-forming regions, substrate binding regions, transmembrane regions, linkers, immunogenic regions, epitopic regions, and high antigenic index regions. Polypeptide fragments of SEQ ID NO: Y falling within conserved domains are specifically encompassed by the present invention. Moreover, polynucleotides encoding these domains are also encompassed. Other preferred polypeptide segments are biologically active fragments.
Biologically active fragments are those exhibiting activity similar, but not necessarily identical, to an activity of an LP polypeptide (or fragment thereof). The biological activity of the fragments may include, e.g., an improved desired activity, or a decreased undesirable activity. Polynucleotides encoding these polypeptide fragments are also encompassed by the invention. Preferably, the polynucleotide fragments of the invention encode a polypeptide that demonstrates a functional activity. The phrase "functional activity" encompasses a polypeptide segment that can accomplish one or more known functional activities associated with a full-length (complete) polypeptide of invention protein. Such functional activities include, e.g., without limitation, biological activity, antigenicity [ability to bind (or compete with a polypeptide of the invention for binding) to an antibody to a polypeptide of the invention], immunogenicity (ability to generate antibody that binds to a polypeptide of the invention), ability to form multimers with a polypeptide of the invention, and the ability to bind to a receptor or ligand of a polypeptide of the invention. The functional activity of a polypeptide of the invention (including fragments, variants, derivatives, and analogs thereof) can be assayed by various methods. For example, where one is assaying for the ability to bind or compete with a full-length polypeptide of the invention for binding to an antibody of a polypeptide of the invention, various immunoassays known in the art can be used, including, e.g., without limitation, competitive and non-competitive assay systems using techniques such as radioimmunoassays, ELISA (enzyme linked immunosorbent assay), "sandwich" immunoassays, immunoradiometric assays, gel diffusion precipitation reactions, immunodiffusion assays, in situ immunoassays (using colloidal gold, enzyme or radioisotope labels, for example), western blots, precipitation reactions, agglutination assays (e.g., gel agglutination assays, hemagglutination assays, complement fixation assays, immunofluorescence assays, protein A assays, and immunoelectrophoresis assays, etc.) In another embodiment, antibody binding is accomplished 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. In another embodiment, where a ligand for a polypeptide of the invention is identified, or the ability of a polypeptide fragment, variant or derivative of the invention to multimerize is being evaluated, binding can be assayed, e.g., by using reducing and non-reducing gel chromatography, protein affinity chromatography, and affinity blotting (see generally, Phizicky, et al. (1995) Microbial. Rev. 59:94-123). In another embodiment, physiological correlates of binding of a polypeptide of the invention to its substrates (signal transduction) can be assayed with common techniques. In addition, assays described herein (see, e.g., the "Examples" section of the application), or otherwise known in the art, can routinely be applied to measure the ability of a polypeptide of the invention (its fragments, variants derivatives and analogs thereof) to elicit a related biological activity (either in vitro or in vivo). Epitopes and Antibodies The present invention encompasses a polypeptide comprising, or alternatively consisting of, an epitope of SEQ ID NO: Y, or an epitope of a polypeptide sequence encoded by a polynucleotide contained in cDNA in a clone deposited as described herein; or encoded by a polynucleotide that stably hybridizes to form a hybridization complex, under stringent hybridization conditions (or lower stringency hybridization conditions) as defined herein, to a complement of a sequence of SEQ ID NO: X or to a sequence contained in deposited cDNA clone as described herein. The present invention further encompasses a polynucleotide sequence encoding an epitope of a polypeptide sequence of the invention (such as, e.g., the sequence disclosed in SEQ ID NO: X), a polynucleotide sequence of the complementary strand of a polynucleotide sequence encoding an epitope of the invention, and a polynucleotide sequence that stably hybridizes to a complementary strand under stringent hybridization conditions or lower stringency hybridization conditions as defined herein. The term "epitope," as used herein, refers to a portion of a polypeptide having antigenic or immunogenic activity in an animal, preferably a mammal, and most preferably in a human. In a preferred embodiment, the present invention encompasses a polypeptide comprising an epitope, as well as the polynucleotide encoding this polypeptide. An "immunogenic epitope," as used herein, is defined as a portion of a protein or a linearized polypeptide (or fragment thereof) that elicits an antibody response in an animal, as determined by any art known method (e.g., by the methods for generating antibodies described herein or otherwise known, see, e.g., Geysen, et al. (1983) Proc. Natl. Acad. Sci. USA 308 1 :3998-4002). An "antigenic epitope," as used herein, is defined as a portion of a protein or polypeptide to which a binding composition, e.g., an antibody or antibody binding fragment, selectively binds or is specifically immunoreactive with as determined by any known art method, e.g., by an immunoassay described herein. Selective binding excludes non-specific binding but does not necessarily exclude cross-reactivity with other antigens. An antigenic determinant may compete with the intact antigen (i.e., the immunogen used to elicit the immune response) for binding to an antibody. Antigenic epitopes need not necessarily be immunogenic. The phrase "specifically binds to" or is "specifically immunoreactive with", when referring to a protein or peptide, refers to a binding reaction which is determinative of the presence of a protein or fragment (e.g., an LP protein) in the presence of a heterogeneous population of proteins and/or other biological components. Typically, the interaction is dependent upon the presence of a particular structure, e.g., an antigenic determinant (or epitope) recognized by a binding composition. For example, if an antibody is specific for epitope "A," the presence of a polypeptide comprising the epitope A, or the presence of free unlabeled A, in a reaction containing free labeled A and the antibody will reduce the amount of labeled A that binds to the antibody. Thus, under designated immunoassay conditions, the specified antibodies bind to a particular protein or polypeptide sequence and do not significantly bind other proteins or other polypeptide sequences that are present in the sample. Specific binding to an antibody under such conditions may require an antibody that is selected for its specificity and/or selectivity for a particular protein. For example, antibodies raised to the protein immunogen with an amino acid sequence depicted in SEQ ID NO: Y can be selected to obtain antibodies specifically immunoreactive with LP proteins or LP polypeptides and not with other proteins or polypeptides. These antibodies will also recognize proteins or polypeptide sequences that have an above average degree of similarity or identity to an LP protein or LP polypeptide sequence. Fragments that function as epitopes can be produced by any conventional means such as, e.g., (1985) Houghten, Proc. Natl. Acad. Sci. USA 82:5131- 5135, further described in U.S. Patent No. 4,631,211. In the present invention, an antigenic or immunogenic epitope preferably contains a polypeptide sequence of at least four, at least five, at least six, at least seven, more preferably at least eight, at least nine, 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 40, at least 50, and, favorably, between about 15 to about 30 contiguous amino acids of a mature polypeptide of SEQ ID NO: Y or encoded by a polynucleotide contained in a cDNA in a clone deposited as described herein. Preferred polypeptide fragments of contiguous amino acid residues of SEQ ID NO: Y comprising immunogenic or antigenic epitopes are at least 10, 15, 20, 25, 30, 35, 40, 45, 50, 55, 60, 65, 70, 75, 80, 85, 90, 95, or 100 contiguous amino acid residues in length. Additional non-exclusive preferred antigenic epitopes include, e.g., the antigenic epitopes disclosed herein, as well as portions thereof. Antigenic epitopes are useful, e.g., to generate antibodies, including monoclonal antibodies that specifically bind the epitope. Preferred antigenic epitopes include, e.g., the antigenic epitopes disclosed herein, as well as any plurality thereof, e.g., at least: two, three, four, five or more of these antigenic epitopes in any combination or structural arrangement. Antigenic epitopes can be used as the target molecules in immunoassays (see, e.g., Wilson, et al. (1984) Cell 37:767-778; Sutcliffe, et al. (1983) Science 219:660-666). Similarly, immunogenic epitopes can be used, e.g., to induce antibodies according to any known art method (see, for instance, Sutcliffe, et al. supra; Wilson, et al. supra; Chow, et al. Proc. Natl. Acad. Sci. USA 82:910-25914; and Bittle, et al. (1985) J. Gen. Virol. 66:2347-2354. Preferred immunogenic epitopes include, e.g., an immunogenic epitope disclosed herein, as well as a plurality or any combination thereof, e.g., of at least two, three, four, five or more of these immunogenic epitopes including, e.g., repeats of a particular epitope. A polypeptide comprising a plurality of epitopes may be used to elicit an antibody response with a carrier protein, such as, e.g., an albumin, to an animal system (such as, e.g., a rabbit or a mouse), or, if a polypeptide is of sufficient length (e.g., at least about 25 amino acids), the polypeptide may be presented without a carrier. However, immunogenic epitopes comprising as few as 8 to 10 amino acids have also been shown to be sufficient to generate antibodies and to be useful since they are capable of binding to, e.g., linear epitopes in a denatured polypeptide such as in Western blotting. Polypeptides or proteins bearing an epitope of the present invention may be used to generate antibodies according to known methods including, e.g., without limitation, in vivo immunization, in vitro immunization, and phage display methods (see, e.g., Sutcliffe, et al. supra; Wilson, et al. supra, and Bittle, et al. (1985) J. Gen. Virol. 66:2347-2354. If in vivo immunization is used, animals may be immunized with free peptide; however, anti-peptide antibody titer may be boosted by coupling the peptide to a macromolecular carrier, such as keyhole limpet hemacyanin (KLH) or tetanus toxoid. For example, polypeptides containing cysteine residues may be coupled to a carrier using a linker such as maleimidobenzoyl- N-hydroxysuccinimide ester (MBS), while other polypeptides may be coupled to carriers using a more general linking agent such as glutaraldehyde. Animals such as rabbits, rats and mice are immunized with either free or carrier- coupled peptides, for instance, by intraperitoneal and/or intradermal injection of emulsions containing about 100 μg of peptide or carrier protein and Freund's adjuvant or any other adjuvant known for stimulating an immune response. Several booster injections may be needed, for instance, at intervals of about two weeks, to provide a useful titer of anti-peptide antibody that can be detected, e.g., by ELISA assay using free peptide adsorbed to a solid surface. The titer of anti-peptide antibodies in serum from an immunized animal may be increased by selection of anti-peptide antibodies, for instance, by adsorption to the peptide on a solid support and elution of the selected antibodies according to any known art method. "Binding Composition" The term "binding composition" refers to molecules that bind with specificity and/or selectivity to an LP of the invention or fragment thereof (such as, e.g., in an antibody-antigen interaction). However, other compositions (e.g., antibodies, oligonucleotides, proteins (e.g., receptors), peptides, or small molecules) may also specifically and/or selectivity associate (bind) with the LP in contrast to other molecules. Typically, the association will be in a natural physiologically relevant protein-protein interaction (either covalent or non-covalent) and it may include members of a multi- protein complex (including carrier compounds or dimerization partners). The composition may be a polymer or chemical reagent. A functional analog may be a protein with structural modifications or may be a wholly unrelated molecule (such as, e.g., one that has a molecular shape that interacts with the appropriate binding determinants). The proteins may serve as agonists or antagonists of the binding partner, see, e.g., Goodman, et al. (eds.) (1990) Goodman & Gilman's: The Pharmacological Bases of Therapeutics (cur. ed.) Pergamon Press, Tarrytown, N.Y. The LP may be used to screen for binding compositions that specifically and/or selectively bind an LP of the invention or fragment thereof (e.g., a binding composition can be a molecule, or part of one, that selectively and/or stoichiometrically binds, whether covalently or not, to one or more specific sites of an LP (or fragment thereof) such as, e.g., in an antigen-antibody interaction, a hormone-receptor interaction, a substrate- enzyme interaction, etc.). At least one and up to a plurality of test binding compositions can be screened for specific and or selective binding with the LP. In one embodiment, a binding composition thus identified is closely related to a natural ligand of an LP (such as, e.g., a ligand or fragment thereof, a natural substrate, a structural or functional mimetic, or a natural binding partner; see, e.g., Coligan, et al. (1991) Current Protocols in rmmunology l(2):_Chapter 5.) Similarly, a binding composition can be closely related to a natural ligand to which a secreted LP (or fragment thereof) binds such as, e.g., to a receptor or to at least a fragment of the receptor (e.g., the ligand binding site). In either case, a binding composition can be rationally designed using known techniques. In another embodiment, screening for binding compositions involves using an appropriate cell that expresses an LP, or fragment thereof (either as a secreted protein or complexed with a cell membrane for presentation). Preferred cells include, e.g., mammalian, yeast, insect (e.g., Drosophila), or bacterial cells (e.g., E. coli). Alternatively, an isolated LP (or fragment thereof) is immobilized on a solid phase (such as, e.g., a stable surface such as, e.g., a membrane, plastic, nylon, a pin, glass, etc.), by covalent or non-covalent attachments, to permit presentation of the LP to a test binding composition for a time sufficient to permit selective and/or specific binding to occur. In a further embodiment, a test binding composition is contacted to a presented LP (or fragment thereof) and the interaction is subsequently analyzed (e.g., to determine the presence or absence of: binding, stimulation, inhibition, agonist or antagonist activity, etc., either of the LP or the test composition). By such methods, inhibitors of a binding interaction can be identified, e.g., screening for peptide or small molecule inhibitors or agonists of a binding interaction between the LP and a binding composition.
"Binding Agent:LP Complex" The term "binding agentiLP complex," as used herein, refers to a complex of a binding agent and a LP (or fragment thereof) which is formed by specific and/or selective binding of the binding agent to the respective LP (or fragment thereof). Specific and/or selective binding of the binding agent means that the binding agent has a specific and/or selective binding site that recognizes a site on the LP protein (or fragment thereof). For example, antibodies raised against a LP protein (or fragment thereof) that recognize an epitope on the LP protein (or fragment thereof) are capable of forming a binding agentLP complex by specific and/or selective binding. Typically, the formation of a binding agentLP complex allows the measurement of LP protein (or fragment thereof) in a mixture of other proteins and/or biologies.
"Antibody:LP Complex" The phrase "antibody:LP complex" refers to an embodiment in which the binding agent, e.g., is an antibody. The antibody may be monoclonal, polyclonal, or a binding fragment of an antibody (including, without limit, e.g., Fv, Fab, or F(ab)2 fragments; diabodies; linear antibodies (Zapata, et al, (1995) Protein Engin. 8(10): 1057-62); single- chain antibody molecules; and multispecific antibodies formed from antibody fragments). Preferably, for cross-reactivity purposes, the antibody is a polyclonal antibody. Various methods such as, e.g., Scatchard analysis in conjunction with radioimmunoassay techniques may be used to assess the affinity of antibodies for LP (or a fragment thereof). Affinity is expressed as an association constant, Ka, which is defined as the molar concentration of antibody:LP complex divided by the molar concentrations of free antigen and free antibody under equilibrium conditions. The Ka determined for a preparation of polyclonal antibodies, which are heterogeneous in their affinities for multiple LP epitopes, represents the average affinity, or avidity, of the antibodies for LP. The Ka determined for a preparation of monoclonal antibodies, which are monospecific for a particular LP epitope, represents a specific measure of affinity. High-affinity antibody preparations with Ka ranging from about 109 to 1012 L/mole are preferred for use in immunoassays in which the antibody:LP complex must withstand rigorous manipulations. Low-affinity antibody preparations with Ka ranging from about 106 to 107 L/mole are preferred for use in immunopurification and similar procedures which ultimately require dissociation of LP, preferably in active form, from the antibody (Catty, (1988) Antibodies, Volume I: A Practical Approach. IRL Press, Washington DC; Liddell, J.E. and A. Cryer (1991) A Practical Guide to Monoclonal Antibodies. John Wiley & Sons, New York NY). The titer and avidity of polyclonal antibody preparations may be further evaluated to determine the quality and suitability of such preparations for certain downstream applications. For example, a polyclonal antibody preparation containing at least 1-2 mg specific antibody/ml (preferably, 5-10 mg specific antibody/ml) is generally employed in procedures requiring precipitation of antibody :LP complexes. Procedures for evaluating antibody selectivity, specificity, titer, and avidity, and guidelines for antibody quality and usage in various applications, are generally available (see, e.g., Catty, supra, and Coligan et al. supra).
Delivery of a Polynucleotide Sequence Encoding an LP Binding Composition In a specific embodiment, a recombinant vector comprising a polynucleotide sequence comprising sequence encoding an LP binding composition (e.g., an antibody or functional derivative thereof) can be administered using any appropriate known art method (e.g., by polynucleotide delivery) to modulate, treat, inhibit, ameliorate, or prevent a disease, syndrome, condition, or disorder associated with aberrant expression and/or activity of a polypeptide (or fragment thereof) of the invention. In a preferred aspect, the vector comprises polynucleotide sequence comprising sequence encoding an LP antibody, wherein the polynucleotide sequence is part of an expression vector that expresses the antibody, (or fragments, or chimeric proteins, or heavy or light chains thereof), in a suitable host. In particular, such polynucleotide sequences have promoters, operably linked to the antibody coding region, that can be either inducible or constitutive, and, optionally, e.g., tissue-specific, cell-specific or developmentally specific . Another particular embodiment, uses nucleic acid molecules comprising sequence in which the antibody coding sequences and any other desired sequences are flanked by regions that promote homologous recombination at a desired site, thus providing for targeted delivery and expression of the antibody (see, e.g., Koller and Smithies, Proc. Natl. Acad. Sci. USA 86:8932-8935 (1989); Zijlstra, et al, (1989) Nature 342:435-438. In specific embodiments, the expressed antibody molecule is a single chain antibody or alternatively, the heterologous sequence includes, e.g., sequence encoding both heavy and light chains, or fragments thereof, of an antibody. Delivery of such sequences into a cell can either be direct, (in which case a cell is directly exposed to the nucleic acid molecule or nucleic acid-carrying vectors), or indirect, (in which a case a cell is first transformed in vitro, then transplanted into a mammalian host). The two approaches are known, respectively, as in vivo or ex vivo polynucleotide delivery. III. Nucleic Acids Primate LP proteins described herein are exemplary of larger classes of structurally and functionally related proteins. The preferred embodiments, as disclosed, are useful in standard procedures to isolate similar genetic sequences from different individuals or other species (e.g., warm blooded animals, such as birds and mammals). Cross hybridization will allow isolation of related genes encoding proteins with substantially similar identity from individuals, strains, or species. A number of different approaches are available to successfully isolate a suitable nucleic acid clone based upon the information provided herein. Southern blot hybridization studies can qualitatively determine the presence of similar genetic sequences in human, monkey, rat, mouse, dog, cow, and rabbit genomes under specific hybridization conditions. Complementary sequences are useful as probes or primers. Based upon identification of the likely amino terminus, other peptides should be particularly useful, e.g., coupled with anchored vector or poly-A complementary PCR techniques or with complementary DNA of other peptides. Techniques for nucleic acid manipulation of genes encoding LP proteins, such as subcloning nucleic acid sequences encoding polypeptides into expression vectors, labeling probes, DNA hybridization, and the like are described generally in Sambrook, et al. Various methods of isolating DNA sequences encoding LP proteins can be utilized. For example, DNA is isolated from a genomic or cDNA library using labeled oligonucleotide probes having sequences identical or complementary to the sequences disclosed herein. Full-length probes may be used, or oligonucleotide probes may be generated by comparison of the sequences disclosed. Such probes can be used directly in hybridization assays to isolate DNA encoding LP proteins, or probes can be designed for use in amplification techniques such as PCR, for the isolation of DNA encoding LP proteins. To prepare a cDNA library, mRNA is isolated from cells which expresses a LP protein. cDNA is prepared from the mRNA and ligated into a recombinant vector. The vector is transfected into a recombinant host for propagation, screening, and cloning. Methods for making and screening cDNA libraries are well known. See Gubler and Hoffman (1983) Gene 25:263-269 and Sambrook, et al. For a genomic library, the DNA is extracted from tissue and either mechanically sheared or enzymatically digested to yield fragments of about 12-20 kb. The fragments are then separated by gradient centrifugation and cloned in bacteriophage lambda vectors. These vectors and phage are packaged in vitro, as described in Sambrook, et al. Recombinant phage are analyzed by plaque hybridization as described in Benton and Davis (1977) Science 196:180-182. Colony hybridization is carried out as generally described in e.g., Grunstein, et al. (1975) Proc. Natl Acad. Sci. USAΛ 72:3961-3965. DNA encoding an LP (or fragment thereof) can be identified in either cDNA or genomic libraries by its ability to hybridize with the nucleic acid probes described herein, e.g., in colony or plaque hybridization assays. The corresponding DNA regions are isolated by standard methods familiar to those of skill in the art (see, e.g., Sambrook, et al). Various methods of amplifying target sequences, such as the polymerase chain reaction, can also be used to prepare DNA encoding LP proteins. Polymerase chain reaction (PCR) technology is used to amplify such nucleic acid sequences directly from mRNA, from cDNA, and from genomic libraries or cDNA libraries. The isolated sequences encoding LP proteins may also be used as templates for PCR amplification. Typically, in PCR techniques, oligonucleotide primers complementary to two 5' regions in the DNA region to be amplified are synthesized. The polymerase chain reaction is then carried out using the two primers (see Innis, et al. (eds.) (1990) PCR Protocols: A Guide to Methods and Applications Academic Press, San Diego, CA.). Primers can be selected to amplify the entire regions encoding a full-length LP protein or to amplify smaller DNA segments as desired. Once such regions are PCR-amplified, they can be sequenced and oligonucleotide probes can be prepared from sequence obtained using standard techniques. These probes can then be used to isolate DNA's encoding LP proteins. Oligonucleotides for use as probes are usually chemically synthesized according to the solid phase phosphoramidite triester method first described by Beaucage and Carruthers (1983) Tetrahedron Lett. 22(20):1859-1862, or using an automated synthesizer, as described in Needham-VanDevanter, et al. (1984) Nucleic Acids Res. 12:6159-6168. Purification of oligonucleotides is performed e.g., by native acrylamide gel electrophoresis or by anion-exchange HPLC as described in Pearson and Regnier (1983) J. Chrom. 255:137-149. The sequence of the synthetic oligonucleotide can be verified using, e.g., the chemical degradation method of Maxam, A.M. and Gilbert, W. in Grossman, L. and Moldave (eds.) (1980) Methods in Enzymology 65:499-560 Academic Press, New York. LP proteins of the invention exhibit limited similarity to portions other intracellular proteins. In particular, beta-sheet and alpha-helix residues can be determined using, e.g., RASMOL program, see Sayle and Milner-White (1995) TIBS 20:374-376; or Gronenberg, et al. (1991) Protein Engineering 4:263-269; and other structural features are defined in Lodi, et al. (1994) Science 263:1762-1767. This invention provides isolated DNA or polynucleotide fragments to encode an
LP protein described herein. In addition, this invention provides isolated or recombinant DNA that encodes a protein or polypeptide which is capable of hybridizing under appropriate conditions, e.g., high stringency, with the DNA sequences described herein. Said biologically active protein or polypeptide can be an intact protein, or fragment, and have an amino acid sequence as disclosed in SEQ ID NO: Y (particularly natural embodiments), or as listed in a Table herein, or as encoded in a described deposited clone. Preferred embodiments are full-length natural sequences. Further, this invention contemplates the use of isolated or recombinant DNA, or fragments thereof, which encode proteins that have sequence similarity (or identity) to an LP protein or which were isolated using cDNA encoding a LP protein as a probe. The isolated DNA can have the respective regulatory sequences in the 5' and 3' flanks, e.g., promoters, enhancers, poly-A addition signals, and others. Also embraced are methods for making expression vectors with these sequences, or for making, e.g., expressing and purifying, protein products. A DNA sequence that codes for an LP protein is particularly useful to identify genes, mRNA, and cDNA specie that code for related or similar proteins, as well as DNAs that code for homologous and/or proteins from different species that share sequence similarity or identity. There are likely homologs (e.g., orthologs and paralogs) and/or similar sequences (e.g., gene duplications) in other species, including primates, rodents, canines, felines, and birds. Various homologous LP proteins are encompassed herein. However, even proteins that have a more distant evolutionary relationship to an LP antigen can readily be isolated under appropriate conditions using these sequences if they are sufficiently structurally similar. Of particular interest, are primate LP proteins. Recombinant clones derived from the genomic sequences, e.g., containing introns, will be useful for transgenic studies, including, e.g., transgenic cells and organisms, and for gene therapy (see, e.g., Goodnow (1992) "Transgenic Animals" in Roitt (ed.) Encyclopedia of Immunology, Academic Press, San Diego, pp. 1502-1504; Travis (1992) Science 256:1392-1394; Kuhn, et al. (1991) Science 254:707-710; Capecchi (1989) Science 244:1288; Robertson (1987)(ed.) Teratocarcinomas and Embryonic Stem Cells: A Practical Approach, IRL Press, Oxford; and Rosenberg (1992) J. Clinical Oncology 10:180-199.).
V. Antibodies Antibodies can be raised to various LP proteins, including individual, polymorphic, allelic, strain, or species variants, and fragments thereof, both in their naturally occurring (full-length) forms and in their recombinant forms. Additionally, antibodies can be raised to LP proteins in either their active forms or in their inactive forms. Anti-idiotypic antibodies may also be used. Antibodies of the invention include, e.g., without limitation, polyclonal, monoclonal, multispecific, human, humanized or chimeric antibodies, single chain antibodies, Fab fragments, F(ab') fragments, fragments produced by a Fab expression library, anti-idiotypic (anti-Id) antibodies (including, e.g., anti-Id antibodies to antibodies of the invention), and an epitope-binding fragment of any of the above. The term "antibody," as used herein refers to immunoglobulin compositions and immunologically active portions of immunoglobulin compositions, e.g., a molecule that contains an antigen binding site that specifically binds an antigen. An immunoglobulin composition of the invention can be of any type (e.g., IgG, IgE, IgM, IgD, IgA, and IgY), class (e.g., IgGl, IgG2, IgG3, IgG4, IgAl, and IgA2) or a subclass of an immunoglobulin molecule. Preferably an antibody is a human antigen-binding antibody fragment of the present invention such as, e.g., without limitation, Fab, Fab' and F(ab')2, Fd, 6 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: a hinge region, a CHI, a CH2, or a CH3 domain or combinations thereof. Also included in the invention is, e.g., without limitation, an antigen-binding fragment that also can comprise any combination of variable region(s) with a hinge region, e.g., such as a CHI, CH2, or a CH3 domain or combinations thereof. An antibody of the invention may be from any animal origin including birds and mammals. Preferably, the antibodies are human, murine (e.g., mouse and rat), donkey, rabbit, goat, guinea pig, camel, horse, or chicken. As used herein, the phrase "human antibodies" includes, e.g., without limitation, antibodies having an amino acid sequence of a human immunoglobulin including, e.g., without limitation, an antibody isolated from a human immunoglobulin library or from an animal transgenic for one or more human immunoglobulins and that do not express endogenous immunoglobulins, as described herein or, as taught, e.g., in U.S. Patent No. 5,939,598. An antibody of the present invention may be monospecific, bispecific, trispecific or of greater multispecificity. Multispecific antibodies may be specific for different epitopes of an LP polypeptide (or fragment thereof) or may be specific for both a polypeptide of the present invention as well as for a heterologous epitope, such as a heterologous polypeptide or solid support material (see, e.g., WO 2093/17715; WO
92/08802; WO 91/00360; WO 92/05793; Tutt, et al. (1991) J. Immunol. 147:60-69; U.S. Patent Nos. 4,474,893; 4,714,681; 4,925,648; 5,573,920; or 5,601,819; or Kostelny, et al. (1992) J. Immunol. 148:1547-1553 . An antibody of the present invention may be described or specified in terms of an epitope(s) or portion(s) of an LP polypeptide (or fragment thereof) that it recognizes or selectively binds. An epitope(s) or polypeptide portion(s) may be specified as described herein, e.g., by N-terminal and C-terminal positions, by size in contiguous amino acid residues, or as listed in an accompanying Table and/or Figure. Additionally, an antibody that specifically binds an epitope, polypeptide, protein, or fragment of a polypeptide or protein of the present invention, may also be specifically excluded from this invention. For instance, Applicants reserve the right to proviso out any antibody that specifically binds an epitope, polypeptide, protein, or fragment of a polypeptide or protein of the present invention. Accordingly, the present invention can encompass a first (or other) antibody that specifically binds a polypeptide or protein, or fragment thereof, of the present invention, and, at the same time, it can exclude a second (or other) antibody that may also selectively bind the same protein or polypeptide, or fragment thereof, e.g., by binding a different epitope. Antibodies of the present invention may also be described or specified in terms of their cross-reactivity. Antibodies that do not bind any other analog, ortholog, paralog, or homolog of an LP polypeptide (or fragment thereof) are included. Antibodies that bind polypeptides with at least 95%, at least 90%, at least 85%, at least 80%, at least 75%, at least 70%, at least 65%, at least 60%, at least 55%, and at least 50% identity (as calculated using known methods or as described herein) to an LP polypeptide (or fragment thereof) are also included. Specific embodiments include, e.g., antibodies of the present invention cross-react with murine, rat and/or rabbit homologs of human proteins, and the corresponding epitopes thereof. Antibodies that do not bind polypeptides with less than 95%, less than 90%, less than 85%, less than 80%, less than 75%, less than 70%, less than 65%, less than 60%, less than 55%, and less than 50% identity (as calculated using known methods or as described herein) to an LP polypeptide (or fragment thereof) are also included. Specific embodiments include, e.g., the above-described cross- reactivity with respect to any single specific antigenic or immunogenic polypeptide, or combination(s) of 2, 3, 4, 5, or more of the specific antigenic and or immunogenic polypeptides disclosed herein. Further encompassed by the present invention is an antibody that selectively binds a polypeptide, which is encoded by a polynucleotide that stably hybridizes, under stringent hybridization conditions (as described herein), to an LP polynucleotide sequence. An antibody of the present invention may also be characterized or specified in terms of its binding affinity to a protein or polypeptide (fragment thereof), or epitope of the invention. A preferred binding affinity of a binding composition, e.g., an antibody or antibody binding fragment, includes, e.g., a binding affinity that demonstrates a dissociation constant or Kd of less than about: 5 X 10"2M, 10"2M, 5 X 10"3M, 10"3M, 5 X lO^M, 10"4M, 5 X 10"5M, 10'5M, 5 X 10_6M, 10"6M, 5 X 10"7M, 10"7M, 5 X 10"8M, 10"8M, 5 X 10"9M, 10"9M, 5 X 10-10M, 10-10M, 5 X 10_11M, 10M, 5 X 10"12M, 10"12M, 5 X 10" 13M, 10_13M, 5 X 10"14M, 10'14M, 5 X 10"15M, or 10"15M. The invention also encompasses antibodies that competitively inhibit binding of a binding composition to an epitope of the invention as determined by any known art method for determining competitive binding, e.g., 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%. Antibodies of the present invention may act as agonists or antagonists of an LP polypeptide (or fragment thereof). For example, an antibody or binding composition of present invention can disrupt, e.g., an interaction, either partially or completely, of a polypeptide of the invention with its cognate receptor/ligand. Preferably, antibodies of the present invention bind an antigenic epitope disclosed herein, or a portion thereof. The invention encompasses both receptor-specific antibodies and ligand-specific antibodies.
It also encompasses receptor-specific antibodies that do not prevent ligand binding but prevent receptor activation. Receptor activation (e.g., signaling) can be determined by techniques described herein or otherwise known in the art. For example, receptor activation can be determined by detecting phosphorylation
(e.g., tyrosine or serine/threonine) of a receptor or its substrate by immunoprecipitation followed by western blot analysis (e.g., as described herein). 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 an activity in absence of the antibody. The invention also features receptor-specific antibodies that 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 encompassed by the invention, are neutralizing antibodies that bind a ligand and prevent it binding to a receptor. Similarly encompassed are ligand-binding antibodies that inhibit receptor activation without inhibiting receptor binding. Alternatively, ligand-binding antibodies that activate a receptor are also included.
Antibodies of the invention may act as receptor agonists, e.g., by potentiating or activating either all or a subset of the biological activities of the ligand-mediated receptor activation, e.g., by inducing dimerization of a receptor. The antibodies may be specified as agonists, antagonists, or inverse agonists for biological activities comprising the specific biological activities of a peptide of the invention disclosed herein. An antibody agonist can be made using known methods art (see, e.g., WO 96/40281; U.S. Patent No.
5811,097; Deng, et al, Blood 92(6):1981-1988 (1998); Chen, et al., Cancer Res. 58(16):3668-3678 (1998); Harrop, et al., J. Immunol. 161(4):1786-1794 (1998); Zhu, et al., Cancer Res. 58( 15):3209-3214 (1998); Yoon, et al., J. Immunol. 160(7): 3170-53179 (1998); Prat, et al., J. Cell. Sci. 11 l(Pt2):237-247 (1998); Pitard, et al., J. Immunol. Methods 205(2): 177-190 (1997); Liautard, et al., Cytokine 9(4):233-241 (1997); Carlson, et al., J. Biol. Chem. 272(17): 11295-1 1301 (1997); Taryman, et al., Neuron 14(4):755- 762 (1995); Muller, et al., Structure 6(9): 1153-1 167 (1998); Bartunek, et al., Cytokine 8(1): 14-20 (1996) (all incorporated by reference for these teachings). Antibodies of the present invention may be used, e.g., without limitation, to purify, detect, or target a polypeptide (or fragment thereof) of the present invention for, e.g., in vitro and/or in vivo diagnostic and therapeutic methods. For example, the antibodies have use in immunoassays for qualitatively and/or quantitatively measuring levels of a polypeptide (or fragment thereof) of the present invention in a biological sample (see, e.g., Harlow, et al., Antibodies: A Laboratory Manual, Cold Spring Harbor Laboratory Press, cur. ed.; incorporated by reference). As discussed in more detail herein, an antibody of the present invention may be used either alone or in combination with other compositions. Furthermore, an antibody may be recombinantly fused to a heterologous polypeptide at the N- or C-terminus, or chemically conjugated (including covalently and non-covalently conjugations) to a polypeptide or other compositions. For example, antibodies of the present invention may be recombinantly fused or conjugated to molecules useful as labels in detection assays and effector molecules such as heterologous polypeptides, drugs, radionuclides, or toxins (see, e.g., WO 92/08495; WO 91/14438; WO 89/12624; U.S. Patent No. 5,314,995; and EP 396,387). An antibody of the invention includes, e.g., derivatives that are modified, e.g., by the covalent attachment of any type of molecule to the antibody such that the covalent attachment does not prevent the antibody from generating an anti-idiotypic response. For example, but not by way of limitation, an antibody derivative includes, e.g., antibodies that have been modified, e.g., by glycosylation, acetylation, pegylation, phosphylation, 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, e.g., but not limited to, specific chemical cleavage, acetylation, formulation, metabolic synthesis of tunicamycin, etc. Additionally, a derivative may contain one or more non-classical amino acids. An antibody of the present invention may be generated by any suitable known art method. Polyclonal antibodies to an antigen-of-interest can be produced by various procedures known in the art. For example, a polypeptide of the invention can be administered to various host animals including, e.g., without limitation, rabbits, mice, rats, etc., to induce the production of sera containing polyclonal antibodies specific for the antigen. Various adjuvants may be used to increase an immunological response depending on the host species, these include, e.g., without limitation, Freund's (complete and incomplete), mineral gels such as e.g., aluminum hydroxide, surface active substances such as e.g., lysolecithin, plutonic polyols, polyanions, peptides, oil emulsions, keyhole limpet hemocyanins, dinifrophenol, and potentially useful human adjuvants such as, e.g., BCG (bacille Calmette-Guerin) and corynebacterium parvum. Such adjuvants are known in the art. Monoclonal antibodies can be prepared using a variety of art known techniques including, e.g., the use of hybridoma, recombinant, and phage display technologies, or combinations thereof. For example, monoclonal antibodies can be produced using hybridoma techniques including those known in the art and taught, e.g., in Harlow, et al., Antibodies: A Laboratory Manual, (Cold Spring Harbor Laboratory Press, current edition); Hammerling, et al., in: Monoclonal Antibodies and T-Cell Hybridomas 563-68 1 (Elsevier, N.Y., 1981) (each of which are incorporated by reference for theses teachings). 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. Methods for producing and screening for specific antibodies using hybridoma technology are routine and known in the art and are discussed in detail herein (e.g., in the Example Section). In a non-limiting example, mice are immunized with a polypeptide of the invention or a cell expressing such a polypeptide. 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 known techniques to any suitable myeloma cells; e.g., SP20 cells (available from the ATCC). Hybridomas are then selected and cloned by limited dilution. The hybridoma clones are then assayed by art known methods to discover cells that secrete antibodies that bind an LP polypeptide (or fragment thereof). Ascites fluid, which generally contains high levels of antibodies, can be generated by immunizing mice with positive hybridoma clones. Accordingly, the present invention provides methods of generating monoclonal antibodies as well as antibodies produced by the method comprising culturing a hybridoma cell secreting an antibody of the invention 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 an LP polypeptide. Antibody fragments that recognize specific epitopes may be generated by known techniques. For example, Fab and F(ab')2 fragments of the invention may be produced by proteolytic cleavage of immunoglobulin molecules, using enzymes such as papain (to produce Fab fragments) or pepsin (to produce F(ab')2 fragments). F(ab')2 fragments contain the variable region, the light chain constant region and the CHI domain of the heavy chain. For example, an antibody of the present invention can also be generated using various phage display methods known in the art in which functional antibody domains are displayed on the surface of phage particles, which carry a polynucleotide sequence encoding them. In a particular embodiment, a phage display method is used to display antigen- binding domains expressed from a repertoire or combinatorial antibody library (e.g., human or murine). Phage that express an antigen binding domain that binds an 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. Typically, phage used in these methods are filamentous phage including, e.g., fd and M 13 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 contemplated for use include, e.g., those of Brinkman, et al., J. Immunol. Methods 182:41-50 (1995); Ames, et al., J. Immunol. Methodsl84:177-186 (1995); Kettleborough, et al, Eur. J. Immunol. 24:952-958 (1994); Persicet, et al., Gene 1879-18 (1997); Burton, et al, Advances in rmmunology 57:191-280(1994); PCT application No. PCT/GB91/01134; WO 90/02809; WO 91/10737; WO 92/01047; WO 9208619; WO 93/1 1236; WO 95/15982; WO95/20401; 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 for these teachings). After phage selection, antibody coding regions from a phage are 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 herein and in the literature. For example, techniques to recombinantly produce Fab, Fab' and F(ab')2 fragments can also be employed using art known methods such as, e.g., WO 92122324; 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) (which are incorporated by reference for these teachings). Examples of producing single-chain Fvs and antibodies include, e.g., U.S. Patents 4,946,778 and 5,258,498; Huston, et al., Methods m Enzymology 203:46-88 (1991); Shu, et al., Proc. Natl. Acad. Sci. USA 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 chimera, 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 for these teachings). Humanized antibodies are antibody molecules from non-human species that bind a desired antigen having one or more complementarity determining regions (CDRs) from a non-human species and a framework region from a human immunoglobulin molecule. Often, framework residues of the human framework regions are substituted with a corresponding residue from a CDR donor antibody to alter, preferably improve, antigen binding. These framework substitutions are identified by any known art method, e.g., by (1) modeling the interactions of a CDR and framework residues to identify framework residues important for antigen binding and/or (2) by sequence comparison to identify unusual framework residues at particular positions (see, e.g., U.S. Patent No. 5,585,089, Riechmann, et al., Nature 332:323 (1988), which are incorporated herein by reference for these teachings). Antibodies can be humanized using a variety of known techniques including, e.g., CDR-grafting (see, e.g., EP 239,400; WO 91/09967; U.S. Patent Nos.
5,225,539; 5,530,101; and 5,585,089), veneering or resurfacing (see, e.g., EP 592,106; EP 519,596; Padlan, Molecular Immunology 28(4/5):489-498 (1991); Studnicka, et al, Protein Engineering 7(6):805-814 (1994); Roguska, et al., Proc. Natl. Acad. Sci. USA 91:969-973 (1994)), and chain shuffling (see, e.g., U.S. Patent No. 5,565,332). Completely human antibodies are particularly desirable for therapeutic treatment of human patients. Human antibodies can be made using a variety of known methods including, e.g., phage display methods described herein using antibody libraries derived from human immunoglobulin sequences (see e.g., U.S. Patent Nos. 4,444,887 and4,716,ll l; and WO 98/46645, WO 98150433, WO 00/58513104 WO 98124893, WO 98116654, WO 96134096, WO 96133735, and WO 91/10741; each of which is incorporated herein by reference for its teachings on human antibodies). Human antibodies can also be produced using transgenic mice that are incapable of expressing functional endogenous immunoglobulins, but which can express human immunoglobulin genes. Generally, human heavy and light chain immunoglobulin gene complexes may be introduced randomly or by homologous recombination into mouse embryonic stem cells. Alternatively, human variable regions, constant regions, and diversity regions may be introduced into mouse embryonic stem cells in addition to the human heavy and light chain genes. The mouse heavy and light chain immunoglobulin genes may be rendered non-functional (separately or simultaneously) with the introduction of human immunoglobulin loci by homologous recombination. In particular, homozygous deletion of the JH region prevents endogenous antibody production. The modified embryonic stem cells are expanded and microinjected into blastocysts to produce chimeric mice. The chimeric mice are then bred to produce homozygous offspring that express human antibodies. The transgenic mice are immunized in the normal fashion with a selected antigen, e.g., all or a portion of a (or fragment thereof) polypeptide of the invention. Monoclonal antibodies directed against the antigen can be obtained from an immunized, transgenic mouse 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, it is possible to produce therapeutically useful IgG, IgA, IgM, and IgE antibodies. For an overview of the technology for producing human antibodies, see, e.g.,
Lonberg and Huszar, Int. Rev. Immunol. 13:65-93 (1995). A more detailed discussion on producing human antibodies and human monoclonal antibodies including protocols can be found, e.g., in WO 98/24893; WO 92/01047; WO 96/34096; WO 96133735; 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; and 5,939,598, all of which are incorporated by reference for teachings of this technology. In addition, commercial companies such as, e.g., Abgenix, Inc. (Freemont, CA) and Genpharm (San Jose, CA) can be hired to produce human antibodies. Completely human antibodies that recognize a selected epitope can be generated by "guided selection." In this method, 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 (see, e.g., Jespers, et al. (1988) BioTechnology 12:899- 903). Further, antibodies of the invention can, in turn, be used to generate anti-idiotype antibodies that "mimic" a polypeptide (or fragment thereof) of the invention using known techniques (see, e.g., Greenspan & Bona, FASEB J. 7(5):437-444; (1989) and Nissinoff, J. (1991) Immunol. 147(8):2429-2438). For example, antibodies that bind and competitively inhibit polypeptide multimerization and/or competitively inhibit binding of a polypeptide of the invention to a ligand can be used to generate anti-idiotypes that "mimic" the polypeptide multimerization and or binding domain and, as a consequence, bind to and neutralize a polypeptide and/or its ligand. Such neutralizing anti-idiotypes, or Fab fragments of such anti-idiotypes, can be used in therapeutic regimens to neutralize a polypeptide ligand. For example, such anti-idiotypic antibodies can be used to bind a polypeptide of the invention (or fragment thereof) and/or to bind its ligand/receptor, and thereby block its biological activity. Polynucleotides Encoding Antibodies. The invention further provides a nucleic acid molecule comprising a polynucleotide sequence encoding an antibody of the invention and/or a fragment thereof. The invention also encompasses polynucleotides that hybridize under stringent or lower stringency hybridization conditions, e.g., as defined herein, to a polynucleotide that encodes an antibody, preferably, that specifically and/or selectively binds a mature polypeptide or protein of the invention, preferably, an antibody that binds to a mature polypeptide of SEQ ID NO:Y. The polynucleotides may be obtained, and the nucleotide sequence of the polynucleotides determined, by any known art method. For example, if the nucleotide sequence of the antibody is known, a polynucleotide encoding the antibody may be assembled from chemically synthesized oligonucleotides (e.g., as described in Kutmeier, et al., (1994) BioTechniques 17:242), which, briefly described, involves synthesizing overlapping oligonucleotides containing portions of the sequence encoding the antibody, annealing, and ligating those oligonucleotides, then, amplifying the ligated oligonucleotides using a polymerase chain reaction. Alternatively, a polynucleotide encoding an antibody can be generated from nucleic acid of any suitable source. If a clone containing a nucleic acid molecule encoding a particular antibody is not available, but, however, the sequence of the antibody molecule is known, then a nucleic acid encoding the immunoglobulin may be chemically synthesized or obtained from a suitable source. For example the source may be an antibody cDNA library, or a cDNA library generated from poly A+ RNA, isolated from any tissue or cell expressing the antibody of interest, such as, e.g., a hybridoma cells selected to express an antibody of the invention by PCR amplification using synthetic primers hybridizable to the 3' and 5' ends of a polynucleotide sequence of interest 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, a nucleic acid molecule for the antibody can be generated. Amplified nucleic acids may be cloned into replicable cloning vectors using any known art method. Once the nucleotide and corresponding amino acid sequence of the antibody are deteirnined, the nucleotide sequence of the antibody may be manipulated using any known art method, e.g., recombinant DNA techniques, site directed mutagenesis, PCR, etc. to generate antibodies having a different amino acid sequence to create amino acid substitutions, deletions, and/or insertions (see, e.g., Sambrook, et al., and Ausubel, et al., eds., cur. ed., Current Protocols in Molecular Biology, John Wiley & Sons, NY, both incorporated by reference for these teachings). In a specific embodiment, the amino acid sequence of the heavy and/or light chain variable domains may be inspected to identify the sequences of complementarity determining regions (CDRs) by known methods, e.g., by comparing known amino acid sequences of other heavy and chain light variable regions to determine regions of sequence hypervariability. Using routine recombinant DNA techniques, one or more CDRs may be inserted within framework regions, e.g., into human framework regions to humanize a non-human antibody. The framework regions may be naturally occurring or consensus framework regions, and are preferably human framework regions (for a listing of human framework regions see, e.g., Chothia, et al. (1998) J. Mol. Biol. 278: 457-479). Preferably, the polynucleotide generated by the combination of the framework regions and CDRs encodes an antibody that specifically binds a polypeptide of the invention (or fragment thereof). Preferably, as discussed herein, one or more amino acid substitutions may be made within the framework regions to improve binding of the antibody to its antigen. Additionally, such methods may be used to make amino acid substitutions or deletions of one or more variable-region, cysteine residues participating in an intrachain disulfide bond to generate antibody molecules lacking one or more intrachain disulfide bonds. Other alterations to the polynucleotide are encompassed by the present invention and within the skill of an ordinary artisan, e.g., such as a molecular biologist. In addition, "chimeric antibody" techniques can be used by, e.g., splicing genes from a mouse antibody molecule of appropriate antigen specificity together with genes from a human antibody molecule of appropriate biological activity (see, e.g., Morrison, et al., Proc. Natl. Acad. Sci. 81:851-855 (1984); Neuberger, et al., Nature 312: 604-608 (1984); Takeda, et al., Nature 314:452-454 (1985)). A chimeric antibody, e.g., humanized antibodies, is a molecule in which different portions are derived from different animal species, such as those having a variable region, derived from a murine mAb and constant region from a human immunoglobulin, e.g., humamzed antibodies. Alternatively, techniques can be adapted to produce single chain antibodies (see, e.g., U.S. Patent No. 4,946,778; Bird, Science 242:423-42 (1988); Huston, et al., Proc. Natl. Acad. Sci. USA 85:5879-5883 (1988); and Ward, et al., Nature 334:544-54 (1989)). Single chain antibodies are formed by linking the heavy and light chain fragments of the Fv region via an ammo 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. (1988) Science 242: 1038- 1041). Methods of Producing Antibodies An antibody or binding composition of the invention can be produced by any known art method, in particular, by chemical synthesis or preferably, by recombinant expression techniques. Recombinant expression of an antibody of the invention, or fragment, derivative or analog thereof, (e.g., a heavy or light chain of an antibody of the invention or a single chain antibody of the invention), requires construction of an expression vector containing a polynucleotide sequence that encodes the antibody. Once a polynucleotide sequence 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 known recombinant DNA technology techniques. Methods known in the art can be used to construct expression vectors containing antibody coding sequences and appropriate transcriptional and translational control signals. These methods include, e.g., without limitation, in vitro recombinant DNA techniques, synthetic techniques, and in vivo genetic recombination. The invention, thus, provides replicable vectors comprising a nucleotide sequence encoding an antibody molecule of the invention, or a heavy or light chain thereof, or a heavy or light chain variable domain, operably-linked to a promoter. Such vectors may include, e.g., the nucleotide sequence encoding the constant region of the antibody molecule (see, e.g., WO 86/05807; WO 89/01036; or 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. Generally speaking, an expression vector is transferred to a host cell by conventional techniques and the transfected cells are then cultured to produce an antibody. Thus, the invention includes, e.g., host cells containing a polynucleotide encoding an antibody of the invention, or a heavy or light chain thereof, or a single chain antibody of the invention, operably linked to a heterologous promoter. In preferred embodiments for the expression of double-chained antibodies, vectors encoding both the heavy and light chains may be co-expressed in a host cell for expression of the entire immunoglobulin molecule, as detailed herein or known the art. A variety of host-expression vector systems may be utilized to express antibody molecules of the invention. Such host-expression systems represent vehicles by which any coding sequence of interest may be produced and subsequently purified. However, when transformed or transfected with an appropriate nucleotide coding sequence, host- expression system cells may also represent an antibody molecule of the invention in situ. These cells include, e.g., without limitation, microorganisms such as bacteria (e.g., E. coli, B. suhtilis) transformed with recombinant bacteriophage DNA, plasmid DNA, or cosmid DNA expression vectors containing antibody coding sequences; yeast (e.g., Saccharomyces, Pichia) transformed with recombinant yeast expression vectors containing antibody coding sequences; insect cell systems infected with recombinant virus expression vectors (e.g., baculovirus) containing antibody coding sequences; plant cell systems infected with recombinant virus expression vectors (e.g., cauliflower mosaic virus, CaMV; tobacco mosaic virus, TMV) or transformed with recombinant plasmid expression vectors (e.g., Ti plasmid) containing antibody coding sequences; or mammalian cell systems (e.g., COS, CHO, BHK, 293, 3T3 cells) harboring recombinant expression constructs containing promoters derived from the genome of mammalian cells (e.g., metallothionein promoter) or from mammalian viruses (e.g., the adenovirus late promoter; the vaccinia virus 7.5K promoter). Preferably, bacterial cells such as Escherichia coli, and more preferably, eukaryotic cells, especially for the expression of whole recombinant antibody molecule, are used for the expression of a recombinant antibody molecule. For example, mammalian cells such as Chinese hamster ovary cells (CHO), in conjunction with a vector such as the major intermediate early gene promoter element from human cytomegalovirus is an effective expression system for antibodies (Foecking, et al. (1986) Gene 45:101; Cockett, et al. (1990) Bio/Technology 8:2). In bacterial systems, a number of expression vectors may be advantageously selected depending upon the intended use of the expressed antibody molecule. For example, when a large quantity of protein is to be produced, like, e.g., for the generation of pharmaceutical compositions of an antibody molecule, then vectors that direct the expression of high levels of fusion protein products, which are readily purified may be desirable. Such vectors include, e.g., without limitation, the E. coli expression vector pUR278 (Ruther, et al., ΕMBO J. 2: 1791 (1983)), in which the antibody coding sequence may be ligated individually into the vector in frame with the lac Z coding region so that a fusion protein is produced; pIN vectors (Inouye and Inouye, Nucleic Acids Res. 13:3 101- 3 109 (1985); Van Heeke and Schuster, J. Biol. Chem. 24:5503-5509 (1989)); and the like. pGΕX 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 pGΕX vectors are designed to include, e.g., thrombin, or factor Xa protease cleavage sites so that the cloned target gene product can be released from a GST moiety. One insect system used as a vector to express a foreign gene, is the Autographa californica nuclear polyhedrosis virus (AcNPV) system. The AcNPV virus grows in Spodopteru frugiperda cells. The antibody coding sequence may be cloned individually into non-essential regions (e.g., the polyhedrin gene) of the virus and placed under control of an AcNPV promoter (e.g., the polyhedrin promoter). In mammalian host cells, a number of viral-based expression systems may be utilized. In cases where an adenovirus is used as an expression vector, the antibody coding sequence of interest may be ligated to an adenovirus transcription/translation control complex, e.g., the late promoter and tripartite leader sequence. This chimeric gene may then be inserted in the adenovirus genome using in vitro or in vivo recombination. Insertion in a non-essential region of the viral genome (e.g., region Εl or Ε3) results in a recombinant virus that is viable and capable of expressing the antibody molecule in infected hosts (see, e.g., Logan and Shenk, (1984) Proc. Natl. Acad. Sci. USA 8 1 :355-359). Specific initiation signals may be required for efficient translation of inserted antibody coding sequences. These signals include, e.g., 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 proper 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, e.g., Bittner, et al., Methods in Enzymol. 153:5 1-544 (1987)). In addition, a host cell strain may be chosen that modulates the expression of the inserted sequence, or modifies and processes the gene product in the specific fashion desired. Such modifications (e.g., glycosylation) and processing (e.g., cleavage) of protein products may be important for the function of a protein. Different host cells have characteristic and specific mechanisms for the post- translational processing and modification of proteins and gene products. Appropriate cell lines or host systems can be chosen to ensure the correct modification and processing of a foreign protein that is expressed. To this end, eukaryotic host cells that possess the cellular machinery for proper: processing of the primary transcript, glycosylation, and phosphorylation may be used. Such mammalian host cells include, e.g., without limitation, CHO, VERY, BHK, Hela, COS, MDCK, 293, 3T3, W138, and, in particular, breast cancer cell lines such as, e.g., BT483, Hs578T, HTB2, BT20 and T47D, and normal mammary gland cell line such as, e.g., CRL7030 and Hs578Bst. For long-term, high-yield production of recombinant proteins, stable expression is preferred. For example, cell lines that stably express an antibody molecule may be engineered. Rather than using expression vectors that contain viral origins of replication, host cells can be transformed with controlled by appropriate expression control elements (e.g., promoter, a polynucleotide sequence enhancer sequences, transcription terminators, polyadenylation sites, etc.), and a selectable marker. After introducing foreign polynucleotide sequence, engineered cells can grow for 1 - 2 days in an enriched media, before switching to a selective media. A selectable marker in a recombinant plasmid confers resistance to selection and allows cells to stably integrate the plasmid into their chromosomes and to form foci that can be subsequently cloned and expanded into cell lines. This method can be used to engineer cell lines that express an antibody molecule. Such engineered cell lines are particularly useful in screening and evaluating compounds that interact either directly or indirectly with an antibody molecule of the invention. A number of selection systems can be used, including, e.g., without limitation, herpes simplex virus thymidine kinase (Wigler, et al., Cell 11 :223 (1977)) in the cells, hypoxanthine-guanine phosphoribosyltransferase in hgprt- cells (Szybalska and Szybalski, Proc. Natl. Acad. Sci. USA 48:202 (1992)), or adenine phosphoribosyltransferase in hgprt-cells (Lowy, et al., Cell 22:817 (1980)). Also, anti- metabolite resistance can be used as the basis of selection for the following genes: dhfr — which confers resistance to methotrexate (Wigler, et al., Natl. Acad. Sci. USA 77:357 (1980); O'Hare, et al, Proc. Natl. Acad. Sci. USA 78:1527 (1981)); gpt— which confers resistance to mycophenolic acid (Mulligan and Berg, Proc. Natl. Acad. Sci. USA 78:2072 (1981)); neo — which confers resistance to the aminoglycoside G-20418 (Clinical Pharmacy 12:488-505; Wu and Wu, Biotherapy 3:87-95 (1991)); Tolstoshev, Ann. Rev. Pharmacol. Toxicol. 32:573-596 (1993); Mulligan, Science 260:926-932 (1993); and Morgan and Anderson, Ann. Rev. Biochem. 62: 19 1-2 17 (1993); May, 1993, TJB TECH 1 1(5): 155-215); and hygro — which confers resistance to hygromycin (Santerre, et al., Gene 30: 147 (1984)). Known art methods can be routinely applied to select a desired recombinant clone
(see, e.g., in Ausubel, et al. (eds.), Current Protocols in Molecular Biology, John Wiley & Sons, NY (1993); Kriegler, Gene Transfer and Expression, A Laboratory Manual, Stockton Press, NY (1990); and in Chapters 12 and 13, Dracopoli, et al. (eds.), Current Protocols in Human Genetics, John Wiley & Sons, NY (1994); Colberre-Garapin, et al., J. Mol. Biol. 150: 1 (198 1), each of which are incorporated by reference for these teachings). Expression levels of an antibody molecule can be increased by vector amplification (see, e.g., Bebbington and Hentschel, Vol. 3 (Academic Press, New York, 1987)). When a marker in the vector-system expressing antibody is amplifϊable, increasing the level of inhibitor present in the host cell culture will increase the number of copies of the marker gene. Since the amplified region is associated with the antibody gene, production of the antibody also increases (Crouse, et al., Mol. Cell. Biol. 3:257 (1983)). 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 that enable equal expression of heavy and light chain polypeptides. Alternatively, a single vector may be used that encodes, and is capable of expressing, both heavy and light chain polypeptides. In such situations, the light chain should be placed before the heavy chain to avoid an excess of toxic free heavy chain (Proudfoot, Nature 322:52 (1986); Kohler, Proc. Natl. Acad. Sci. USA 77:2197 (1980)). Coding sequences for heavy and light chains may comprise cDNA or genomic DNA. Once an antibody molecule of the invention has been (produced by, e.g., an animal, chemically synthesized, or recombinantly expressed), it may be purified by any known method, e.g., by chromatography (e.g., ion exchange, affinity, particularly by affinity for the specific antigen after Protein A, and sizing column chromatography), centrifugation, differential solubility, or any other technique for protein purification. In addition, an antibody of the present invention or fragments thereof can be fused to heterologous polypeptide sequences to facilitate purification using any art known method or one described herein. The present invention encompasses antibodies recombinantly fused or chemically conjugated (including both covalent and non-covalent conjugations) to a polypeptide (or portion thereof, preferably comprising at least: 10, 20, 30, 40, 50, 60, 70, 80, 90, or 100 contiguous amino acids of a polypeptide of SED ID NO:X) of the present invention to generate fusion proteins. The fusion does not necessarily need to be direct, but may occur through linker sequences. The antibodies may be specific for antigens other than a polypeptide of the inventor (or portion thereof, preferably at least: 10, 20, 30, 40, 50, 60, 70, 80, 90, or 100 contiguous amino acids) of the present invention. For example, antibodies may be used to target an LP polypeptide (or fragment thereof) to particular cell types, either in vitro or in vivo, by fusing or conjugating a polypeptide (or fragment thereof) of the present invention to an antibody specific for a particular cell surface receptor. Antibodies fused or conjugated to a polypeptide of the invention may also be used in in viti'o irnmunoassays and in purification methods using known art methods (see e.g., Harbor, et al, supra, and WO 9312 1232; EP 439,095; Naramura et al. (1994) Immunol. Lett. 39:9 1-99; U.S. Patent No. 5,474,981; Gillies, et al. (1992) Proc. Natl. Acad. Sci. USA 89:1428-1432; Fell, et al. (1991) J. Immunol. 146: 2446-2452, each of which are incorporated by reference for these teachings). The present invention further includes compositions comprising a polypeptide of the invention (or fragment thereof) fused or conjugated to an antibody domain other than a variable region. For example, a polypeptide of the invention (or fragment thereof) may be fused or conjugated to an antibody Fc region, or portion thereof. The antibody portion that is fused to a polypeptide of the invention (or fragment thereof) may comprise a constant region, a hinge region, a CHI domain, a CH2 domain, and/or a CH3 domain or any combination of whole domains or portions thereof. A polypeptide of the invention (or fragment thereof) may also be fused or conjugated to an antibody portion described herein to form multimers. For example, Fc portions fused to a polypeptide of the invention (or fragment thereof) can form dimers through disulfide bonding between the Fc portions. Higher multimeric forms can be made by fusing the polypeptides to portions oflgA and lgM. Methods for fusing or conjugating a polypeptide of the invention (or fragment thereof) to an antibody portion are known (see, e.g., U.S. Patent Nos. 5,336,603; 5,622,929; 5,359,046; 5,349,053; 5,447,851; 5,112,946; EP 307,434; EP 367,166; WO 96/04388; WO9106,570; Ashkenazi, et al. (1991) Proc. Natl. Acad. Sci. USA 88: 10535- 10539; Zheng, et al. (1995) J. Immunol. 154:5590-5600; and Vie, et al. (1992) Proc. Natl. Acad. Sci. USA 89: 11337- 11341; which are hereby incorporated by reference for these teachings). As discussed herein, a polypeptide, polypeptide fragment, or a variant of SEQ ID NO:Y (or part thereof) may be fused or conjugated to an antibody portion described herein or known in the art to increase the in vivo half-life. Further, a polypeptide, polypeptide fragment, or a variant of SEQ ID NO: Y (or part thereof) may be fused or conjugated to an antibody portion to facilitate purification. One example uses chimeric proteins comprising the first two domains of the human CD4-polypeptide and various domains of the constant regions of the heavy or lighl chains of mammalian immunoglobulins. (see, e.g., EP 394,827; Traunecker, et al. (1988) Nature 33 1 :84-86).S A polypeptide, polypeptide fragment, or a variant of SEQ ID NO: Y (or part thereof) fused or conjugated to an antibody having disulfide-linked dimeric structures (due to the IgG) may also be more efficient in binding and neutralizing other molecules, than the monomeric secreted protein or protein fragment alone (see, e.g., Fountoulakis, et al. (1995) 3. Biochem. 270: 3958-3964). In many cases, the Fc part of a fusion protein is beneficial in therapy and diagnosis, and thus can result in, e.g., improved pharmacokinetic properties (see, e.g., EP A232, 262). Alternatively, deleting the Fc part after the fusion protein has been expressed, detected, and purified, can be favored. For example, the Fc portion may hinder therapy and diagnosis if the fusion protein is used as an antigen for immunizations. In drug discovery, e.g., human proteins, such as hIL-5, have been fused with Fc portions for the purpose of high-throughput screening assays to identify antagonists of hIL-5 (see, e.g., Bennett, et al. (1995) J. Molecular Recognition 8:52-58; Johanson, et al. (1995) J. Biol. Chem. 270:9459-9471). Moreover, an antibody of the present invention (or fragment thereof) can be fused to marker sequences, such as a peptide to facilitate purification. In preferred embodiments, the marker amino acid sequence is a hexa-histidine peptide, such as the tag provided in a pQE vector (QIAGEN, Inc., Chatsworth, CA), among others, many of which are commercially available. Hexa-histidine provides for convenient purification of a fusion protein (Gentz, et al. (1989) Proc. Natl. Acad. Sci. USA 86:821-824). Other peptide tags useful for purification include, e.g., the "HA" tag, which corresponds to an epitope derived from the influenza hemagglutinin protein (Wilson, et al. (1984) Cell 37:767) and the "flag" tag.
VI. Making LP proteins; Mimetics DNAs which encode a LP protein or fragments thereof can be obtained by chemical synthesis, screening cDNA libraries, or by screening genomic libraries prepared from a wide variety of cell lines or tissue samples. Methods for doing so, or making expression vectors are either art known or are described herein. These DNAs can be expressed in a wide variety of host cells for the synthesis of a full-length protein or fragments which can in turn, e.g., be used to generate polyclonal or monoclonal antibodies; for binding studies; for construction and expression of modified molecules; and for structure/function studies. Each LP protein or its fragments can be expressed in host cells that are transformed or transfected with appropriate expression vectors. By "transformed" is meant a cell into which (or into an ancestor of which) a DNA molecule has been introduced, by means of recombinant techniques, which encodes an LP polypeptide or fragment thereof. Heterologously expressed LP polypeptides can be substantially purified to be free of protein or cellular contaminants, other than those derived from the recombinant host, and therefore are particularly useful in pharmaceutical compositions when combined with a pharmaceutically acceptable carrier and/or diluent. The antigen, e.g., LP protein, or portions thereof, may be expressed as fusions with other proteins or possessing an epitope tag. Expression vectors are typically self-replicating DNA or RNA constructs containing the desired antigen gene or its fragments, usually operably linked to appropriate genetic control elements that are recognized in a suitable host cell. The specific type of control elements necessary to effect expression depends on the host cell used. Generally, genetic control elements include a prokaryotic promoter system or a eukaryotic promoter expression control system, and typically include a transcriptional promoter, an optional operator to control the onset of transcription, transcription enhancers to elevate the level of mRNA expression, a sequence that encodes a suitable ribosome binding site, and sequences that terminate transcription and translation. All of the associated elements both necessary and sufficient for the production of LP polypeptide are in operable linkage with the nucleic acid encoding the LP polypeptide (or fragment thereof). Usually, expression vectors also contain an origin of replication that allows the vector to replicate independently of the host cell. An expression vector will preferably include, e.g., at least one selectable marker. Such markers include, e.g., without limit, dihydrofolate reductase, G418, or neomycin resistance for eukaryotic cell culture and tetracycline, kanamycin or ampicillin resistance genes for culturing in E. coli and other bacteria. The vectors of this invention contain DNAs which encode an LP protein, or a fragment thereof, typically encoding, e.g., a biologically active polypeptide, or protein. The DNA can be under the control of a viral promoter and can encode a selection marker. This invention further contemplates use of expression vectors capable of expressing eukaryotic cDNA coding for a LP (or fragment) in a prokaryotic or eukaryotic host, where the vector is compatible with the host and where the eukaryotic cDNA coding for the protein is inserted into the vector such that growth of the host containing the vector expresses the cDNA in question. Usually, expression vectors are designed for stable replication in their host cells or for amplification to greatly increase the total number of copies of the desirable gene per cell. It is not always necessary to require that an expression vector replicate in a host cell, e.g., it is possible to effect transient expression of the protein or its fragments in various hosts using vectors that do not contain a replication origin that is recognized by the host cell. It is also possible to use vectors that cause integration of an LP protein gene or its f agments into the host DNA by recombination, or to integrate a promoter that controls expression of an endogenous gene. Vectors, as used herein, encompass plasmids, viruses, bacteriophage, integratable DNA fragments, and other vehicles that enable the integration of DNA fragments into the genome of the host. Expression vectors are specialized vectors that contain genetic control elements that effect expression of operably linked genes. Plasmids are the most commonly used form of vector, but many other forms of vectors that perform an equivalent function are also suitable for use (see, e.g., Pouwels, et al. (1985 and Supplements) Cloning Vectors: A Laboratory Manual Elsevier, N.Y.; and Rodriquez, et al. (eds.) (1988) Vectors: A Survey of Molecular Cloning Vectors and Their Uses Buttersworth, Boston, MA). Generally, a plasmid vector is introduced in a precipitate, such as, e.g., a calcium phosphate precipitate, or in a complex with a charged lipid. If the vector is a virus, it may be packaged in vitro using an appropriate packaging cell line and then transduced into host cells. The polynucleotide insert should be operatively linked to an appropriate promoter, such as, e.g., without limit, the phage lambda PL promoter, the E. coli lac, trp, phoA, and tat promoters, the SV40 early or late promoters, and promoters of retroviral LTRs. Other suitable promoters are known to a skilled artisan. Suitable host cells include prokaryotes, lower eukaryotes, and higher eukaryotes. Prokaryotes include both gram negative and gram positive organisms, e.g., E. coli and B. subtilis. Lower eukaryotes include yeasts, e.g., S. cerevisiae and Pichia, and species of the genus Dictyostelium. Higher eukaryotes include established tissue culture cell lines from animal cells, both of non-mammalian origin, e.g., insect cells, and birds, and of mammalian origin, e.g., human, primates, and rodents. Prokaryotic host- vector systems include a variety of vectors for many different species. As used herein, E. coli and its vectors will be used generically to include equivalent vectors used in other prokaryotes. A representative vector for amplifying DNA is pBR322 or its derivatives. Vectors that can be used to express these proteins or protein fragments include, but are not limited to, such vectors as those containing the lac promoter (pUC-series); trp promoter (pBR322-trp); Ipp promoter (the pIN-series); lambda-pP or pR promoters (pOTS); or hybrid promoters such as ptac (pDR540). See Brosius, et al. (1988) "Expression Vectors Employing Lambda-, trp-, lac-, and Ipp- derived Promoters," in Rodriguez and Denhardt (eds.) Vectors: A Survey of Molecular Cloning Vectors and Their Uses 10:205-236 Buttersworth, Boston, MA. Other representative bacterial vectors include, e.g., without limit, pQE70, pQE60, and pQE-9, (available from QIAGEN, Inc.); pBluescript vectors, Phagescript vectors, pNH8A, pNH16a, pNH18A, pNH46A,, (available from Stratagene Cloning Systems, Inc.); and ptrc99a, pKK223-3, pKK233-3, pDR540, pRIT5 (available from Pharmacia Biotech, Inc). Lower eukaryotes, e.g., yeasts and Dictyostelium, may be transformed with LP protein sequence containing vectors. For purposes of this invention, the most common lower eukaryotic host is the baker's yeast, Saccharomyces cerevisiae. It will be used generically to represent lower eukaryotes although a number of other strains and species are also available. Yeast vectors typically consist of a replication origin (unless of the integrating type), a selection gene, a promoter, DNA encoding the desired protein or its fragments, and sequences for translation termination, polyadenylation, and transcription termination. Suitable expression vectors for yeast include such constitutive promoters as 3-phosphoglycerate kinase and various other glycolytic enzyme gene promoters or such inducible promoters as the alcohol dehydrogenase 2 promoter or metallothionine promoter. Suitable vectors include derivatives of the following types: self-replicating low copy number (such as the YRp-series), self-replicating high copy number (such as the YEp-series); integrating types (such as the Ylp-series), or mini-chromosomes (such as the YCp-series). Additional representative yeast expression vectors include, e.g., without limit, pYES2, pYDl, pTEFl/Zeo, pYES2/GS, pPIGZ, pGAPZ, pGAPZalph, pPIC9, pPIC3.5, PHIL-D2, PHIL-SI, pPIC3.5K, pPIC9K, and PA0815 (available from Invifrogen, Carlsbad, CA). Other suitable vectors will be readily apparent to the skilled artisan. Higher eukaryotic tissue culture cells are typically the preferred host cells for expression of the functionally active LP protein. In principle, many higher eukaryotic tissue culture cell lines may be used, e.g., insect baculovirus expression systems, whether from an invertebrate or vertebrate source. However, mammalian cells are preferred to achieve proper processing, both co-translationally and post-translationally. Transformation or transfection and propagation of such cells are routine in the art. Useful cell lines include HeLa cells, Chinese hamster ovary (CHO) cell lines, baby rat kidney (BRK) cell lines, insect cell lines, bird cell lines, and monkey (COS) cell lines. Expression vectors for such cell lines usually include an origin of replication, a promoter, a translation initiation site, RNA splice sites (e.g., if genomic DNA is used), a polyadenylation site, and a transcription termination site. These vectors also may contain a selection gene or amplification gene. Suitable expression vectors may be plasmids, viruses, or retroviruses carrying promoters derived, e.g., from such sources as from adenovirus, SV40, parvoviruses, vaccinia virus, or cytomegalovirus. Representative examples of suitable expression vectors include pCDNAl; pCD (Okayama, et al. (1985) Mol. Cell Biol. 5:1136-1142); pMClneo Poly-A, (Thomas, et al. (1987) CeU 51:503- 512); and a baculovirus vector such as pAC 373 or pAC 610. Additional representative eukaryotic vectors include, e.g., without limit, pWLNEO, pSV2CAT, pOG44, pXTl and pSG (available from Stratagene); and pSVK3, pBPV, pMSG and pSVL (available from Pharmacia Biotech, Inc.). Introduction of the construct into a host cell can be effected by, e.g., without limit, by calcium phosphate transfection, DEAE-dextran mediated transfection, cationic lipid- mediated transfection, electroporation, transduction, infection, or other methods, (see, e.g., Davis, et al. (1986) Basic Methods in Molecular Biology). It is specifically contemplated that a polypeptide (or fragment thereof) of the present invention may in fact be expressed by a host cell lacking a recombinant vector. The polypeptide can be recovered and purified from recombinant cell cultures by known methods including, e.g., without limit, ammonium sulfate or ethanol precipitation, acid extraction, anion or cation exchange chromatography, phosphocellulose chromatography, hydrophobic interaction chromatography, affinity chromatography, hydroxylapatite chromatography and pectin chromatography. Most preferably, high performance liquid chromatography ("HPLC") is employed for purification of the polypeptide. A polypeptide (or fragment thereof) of the present invention, and preferably, a mature and/or secreted form, can also be recovered from natural sources, including, e.g., without limit, bodily fluids, tissues, and cells, (whether directly isolated or cultured); products of chemical synthetic procedures; and products produced by recombinant techniques from a prokaryotic or eukaryotic host (including, e.g., bacterial, yeast, higher plant, insect, and mammalian cells). It is likely that LP proteins need not be glycosylated to elicit biological responses.
However, it will occasionally be desirable to express an LP protein or LP polypeptide in a system that provides a specific or defined glycosylation pattern. In this case, the usual pattern will be that provided naturally by the expression system. However, the pattern will be modifiable by exposing the polypeptide, e.g., in unglycosylated form, to appropriate glycosylating proteins introduced into a heterologous expression system. For example, the LP protein gene may be co-transformed with one or more genes encoding mammalian or other glycosylating enzymes. It is further understood that over glycosylation may be detrimental to LP protein biological activity, and that one of skill may perform routine testing to optimize the degree of glycosylation which confers optimal biological activity. In addition, an LP polypeptide (or fragments thereof) may also include, e.g., an initial modified methionine residue (in some cases because of host-mediated processes). Typically, the N-terminal methionine encoded by the translation initiation codon removed with high efficiency from any protein after translation in all eukaryotic cells. While the N-terminal methionine on most proteins is also efficiently removed in most prokaryotes, for some proteins depending on the nature of the amino acid to which the N-terminal methionine is covalently linked, the removal process is inefficient. In one embodiment, the yeast Pichia pastoris is used to express a polypeptide of the present invention(or fragment thereof) in an eukaryotic system. Pichia pastoris is a methylotrophic yeast, which can metabolize methanol as its sole carbon source. A main step in the methanol metabolization pathway is the oxidation of methanol to formaldehyde catalyzed by the alcohol oxidase. To metabolize methanol as its carbon source, Pichiu pustoris must generate high levels of alcohol oxidase due, in part, to the relatively low affinity of alcohol oxidase for O2. Consequently, in a growth medium using methanol as a primary 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 AOX1 gene comprises up to approximately 30% of the total soluble protein in Pichiu pastoris (see, e.g., Ellis, et al., Mol. Cell. Biol. 5:1111-21 (1985); Koutz, et al., Yeast 5: 167-77 (1989); Tschopp, et al., Nucl. Acids Res. 15:3859-76 (1987)). Thus, a heterologous coding sequence, such as, e.g., an LP polynucleotide sequence, (or fragment thereof) under the transcriptional regulation of all or part of the AOXl regulatory sequence is expressed at exceptionally high levels in Pichia yeast grown in the presence of methanol. In one example, the plasmid vector pPIC9K is used to express polynucleotide sequence encoding a polypeptide of the invention, (or fragment thereof) as set forth herein, in a Pichea yeast system essentially as described in "Pichia Protocols: Methods in Molecular Biology," D.R. Higgins and J. Cregg, eds. The Humana Press, Totowa, NJ, 1998. This expression vector allows expression and secretion of a protein of the invention by virtue of the strong AOXl promoter linked to the Pichia pastoris alkaline phosphatase (PHO) secretory signal peptide located upstream of a multiple cloning site. Many other yeast vectors could be used in place of pPIC9K, such as, e.g., pYES2, pYDl, pTEFl/Zeo, pYES2/GS, pPICZ, pGAPZ, pGAPZalpha, pPIC9, pPIC3.5, PHIL-D2, PHIL- Sl, pPIC3.5K, and, PA08, as a skilled in the artisan would appreciate, as long as the proposed expression construct provides appropriately located and operably linked signals for transcription, translation, secretion (if desired), and the like, (including an in-frame stop codon as required). In another embodiment, high-level expression of a heterologous coding sequence, such as, e.g., a polynucleotide sequence of the present invention, may be achieved by cloning a heterologous polynucleotide of the invention (or fragment thereof) into an expression vector such as, e.g., pGAPZ or pGAPZ alpha, and growing the yeast culture in the absence of methanol. In addition to encompassing host cells containing vector constructs discussed herein, the invention also encompasses primary, secondary, and immortalized host cells of vertebrate origin, particularly mammalian origin, and more particularly, human origin, which have been engineered to delete or replace endogenous genetic material (e.g., coding sequence), and/or to include, e.g., genetic material (e.g., heterologous polynucleotide sequences) in operable linkage with a polynucleotide (or fragment thereof) of the invention, and which activate, alter, and/or amplify an endogenous polynucleotide(s). For example, known art techniques may be used to operably associate heterologous control regions (e.g., promotes and/or enhances) and an endogenous polynucleotide sequence(s) via, e.g., homologous recombination, resulting in the formation of a new transcription unit (see, e.g., U.S. Patent No. 5641,670; U.S. Patent No. 5,733,761; WO 96/29411; WO 94/12650; Roller, et al. (1989) Proc. Natl. Acad. Sci. USA 86:8932-8935; and Zijlstra, et al. (1989) Nature 342:435-438, which are incorporated by reference for their teachings on operably associated heterologous control regions). Furthermore, heterologously expressed proteins or polypeptides can also be expressed in plant cells. For plant cells viral expression vectors (e.g., cauliflower mosaic virus and tobacco mosaic virus) and plasmid expression vectors (e.g., TI plasmid) are suitable. Such cells are available from a wide range of sources (e.g., the American Tissue Type Culture Collection, Rockland, MD; also, see for example, Ausubel, et al. (cur. ed. and Supplements; expression vehicles may be chosen from those provided e.g., in Pouwels, et al. (Cur. ed..) Cloning Vectors, A Laboratory Manual). A LP protein, or a fragment thereof, may be engineered to be phosphatidyl inositol (PI) linked to a cell membrane, but can be removed from membranes by treatment with a phosphatidyl inositol cleaving enzyme, e.g., phosphatidyl inositol phospholipase-C. This releases the antigen in a biologically active form, and allows purification by standard procedures of protein chemistry (see, e.g., Low (1989) Biochem. Biophvs. Acta 988:427-454; Tse, et al. (1985) Science 230:1003-1008; and Brunner, et al. (1991) J. Cell Biol. 114:1275-1283). Now that LP proteins have been characterized, fragments or derivatives thereof can be prepared by conventional processes for synthesizing peptides. These include processes such as are described in Stewart and Young (1984) Solid Phase Peptide Synthesis Pierce Chemical Co., Rockford, IL; Bodanszky and Bodanszky (1984) The Practice of Peptide Synthesis Springer-Verlag, New York, NY; and Bodanszky (1984) The Principles of Peptide Synthesis Springer-Verlag, New York, NY. For example, an azide process, an acid chloride process, an acid anhydride process, a mixed anhydride process, an active ester process (for example, p-nitrophenyl ester, N-hydroxysuccinimide ester, or cyanomethyl ester), a carbodiimidazole process, an oxidative-reductive process, or a dicyclohexylcarbodiimide (DCCD)/additive process can be used. Solid phase and solution phase syntheses are both applicable to the foregoing processes. The prepared protein and fragments thereof can be isolated and purified from the reaction mixture by means of peptide separation, for example, by extraction, precipitation, electrophoresis and various forms of chromatography, and the like. An LP protein of this invention can be obtained in varying degrees of purity depending upon its desired use. Purification can be accomplished by use of known protein purification techniques or by the use of the antibodies or binding partners herein described (e.g., in immunoabsorbant affinity chromatography). Immunoabsorbant affinity chromatography is carried out by first linking the antibodies to a solid support and then contacting the linked antibodies with solubilized lysates of appropriate source cells, lysates of other cells expressing the protein, or lysates or supernatants of cells producing the LP proteins as a result of known recombinant DNA techniques. Multiple cell lines may be screened for one cell line that expresses an LP protein (or fragment thereof) at a high level when compared to other cells. Various cell lines, are screened and one is selected for its favorable handling properties. Natural LP proteins can be isolated from natural sources, or by expression from a transformed cell using an appropriate expression vector. Purification of the expressed protein is achieved by standard procedures, or may be combined with engineered means for effective purification at high efficiency from cell lysates or supernatants. Epitope or other tags, e.g., FLAG or His6 segments, can be used for such purification features. VII. Physical Variants The invention also encompasses proteins or peptides having substantial amino acid sequence similarity with an amino acid sequence of an LP protein described herein. Natural variants include individual, polymorphic, allelic, strain, or species variants. Amino acid sequence similarity, or sequence identity, is determined by optimizing residue matches, if necessary, by introducing gaps as required. This changes when considering conservative substitutions as matches. Conservative substitutions typically include substitutions within the following groups: gly cine, alanine; valine, isoleucine, leucine; aspartic acid, glutamic acid; asparagine, glutamine; serine, threonine; lysine, arginine; and phenylalanine, tyrosine. Homologous amino acid sequences include natural polymorphic, allelic, and interspecies variations in each respective protein sequence.
Typical homologous proteins or peptides will have from 50-100% similarity (if gaps can be introduced), to 75-100% similarity (if conservative substitutions are included) over fixed stretches of amino acids with the amino acid sequence of the LP protein. Similarity measures will be at least about 50%, generally at least 65%, usually at least 70%, preferably at least 75%, and more preferably at least 90%, and in particularly preferred embodiments, at least 96% or more. See also Needleham, et al. (1970) J. Mol. Biol. 48:443-453; Sankoff, et al. (1983) Time Warps. String Edits, and Macromolecules: The Theory and Practice of Sequence Comparison Chapter One, Addison- Wesley, Reading, MA; and software packages from IntelliGenetics, Mountain View, CA; and the University of Wisconsin Genetics Computer Group, Madison, WI. Stretches of amino acids will be at least about 10 amino acids, usually about 20 amino acids, usually 50 amino acids, preferably 75 amino acids, and in particularly preferred embodiments at least about 100 amino acids. Identity can also be measures over amino acid stretches of about 98, 99, 110, 120, 130, etc. Nucleic acids encoding mammalian LP proteins will typically hybridize to the nucleic acid sequence of SEQ ID NO: X under stringent conditions. For example, in one embodiment nucleic acids encoding human LP proteins will normally hybridize to a nucleic acid of SEQ ID NO: X under stringent hybridization conditions (as described herein). Generally, stringent conditions are selected to be about 10° C lower than the thermal melting point (Tm) for the probe sequence at a defined ionic strength and pH. The Tm is the temperature (under defined ionic strength and pH) at which 50% of the target sequence hybridizes to a perfectly matched probe. Typically, stringent conditions will be those in which the salt concentration is about 0.2 molar at pH 7 and the temperature is at least about 50° C. Other factors may significantly affect the stringency of hybridization, including, among others, base composition and size of the complementary strands, the presence of organic solvents such as formamide, and the extent of base mismatching. One preferred embodiment includes nucleic acids that bind to a disclosed sequence in 50% formamide and 200 mM NaCl at 42° C. Nucleic acids that hybridize to an LP nucleic acid of the invention are useful as cloning probes, primers (e.g., a PCR primer), or diagnostic probes. Hybridizing nucleic acids can be splice variants encoded by one of the LP genes described herein. Thus, the hybridizing nucleic acids may encode a polypeptide that is shorter or longer than the various forms of LP described herein. Hybridizing nucleic acids may also encode proteins that are related to LP (e.g., polypeptides encoded by genes that include a portion having a relatively high degree of identity to a LP gene described herein). An isolated LP protein encoding DNA can be readily modified by nucleotide substitutions, nucleotide deletions, nucleotide insertions, and short inversions of nucleotide stretches. Such modifications result in novel DNA sequences, which encode LP protein antigens, their derivatives, or proteins having highly similar physiological, immunogenic, or antigenic activity. Modified sequences can be used to produce mutant antigens or to enhance expression. Enhanced expression may involve gene amplification, increased transcription, increased translation, and other mechanisms. Such mutant LP protein derivatives include predetermined or site-specific mutations of the respective protein or its fragments. "Mutant LP protein" encompasses a polypeptide otherwise falling within the homology definition of a human LP protein as set forth herein (or in a deposited clone), but having an amino acid sequence which differs from that of a LP protein as found in nature, whether by way of deletion, substitution, or insertion. In particular, "site specific mutant LP protein" generally includes proteins having significant similarity with a protein having a sequence of SEQ ID NO: Y, e.g., natural embodiments, and as sharing various biological activities, e.g., antigenic or immunogenic, with those sequences, and in preferred embodiments contain most or all of the disclosed sequence. This applies also to polymorphic variants from different individuals. Similar concepts apply to different LP proteins, particularly those found in various warm-blooded animals (e.g., mammals and birds). As stated before, it is emphasized that descriptions are generally meant to encompass other LP proteins, not limited to the human embodiments specifically discussed. The invention encompasses, but is not limited to, LP proteins and polypeptides that are functionally related to an LP encoded by the specific sequence identifiers of the present application. Functionally related proteins and polypeptides include any protein or polypeptide sharing a functional characteristic with LP of the present invention (e.g., the ability to stimulate a Janus family tyrosine kinase). Such functionally related LP polypeptides include, without limitation, additions or substitutions of amino acid residues within the amino acid sequence encoded by the LP sequences described herein; particularly, those that result in a silent change, thus producing a functionally equivalent LP polypeptide. Amino acid substitutions may be made based on similarity in polarity, charge, solubility, hydrophobicity, hydrophilicity, and/or the amphiphatic nature of the residues involved. For example, nonpolar (hydrophobic) amino acids include alanine, leucine, isoleucine, valine, proline, phenylalanine, tryptophan, and methionine; polar neutral amino acids include glycine, serine, threonine, cysteine, tyrosine, asparagine, and glutamine; positively charged (basic) amino acids include arginine, lysine, and histidine; and negatively charged (acidic) amino acids include aspartic acid and glutamic acid. Furthermore, non-classical amino acids or chemical amino acid analogs may be substituted or added into an LP polypeptide sequence. Non-classical amino acids include, e.g., without limitation, D-isomers of the common amino acids; 2,4-diaminobutyric acid; a-amino isobutyric acid, 4-aminobutyric acid, Abu, 2-amino butyric acid, g-Abu, e-Ahx, 6-amino hexanoic acid, Aid, 2-amino isobutyric acid, 3 -amino propionic acid, omithine, norleucine, norvaline, hydroxyproline, sarcosine, citrulline, homocitrulline, cysteic acid, t-butylglycine, t-butylalanine, phenylglycine, cyclohexylalanine, b-alanine, fluoro-amino acids, designer amino acids such as b-methyl amino acids, Ca-methyl amino acids, Na- methyl amino acids, and amino acid analogs in general. Furthermore, the amino acid can be either dextrorotary(D ) or levorotary (L). While random mutations can be made to an LP nucleic acid molecule (using well known random mutagenesis techniques) and the resulting LP polypeptides can be tested for activity, site-directed mutations of LP coding sequences can be engineered (using well known site-directed mutagenesis techniques) to generate mutant LP with increased function (e.g. greater inhibition (or stimulation) of a kinase activity, greater resistance to degradation, increased or decreased binding affinity). To design functionally related and functionally variant LP polypeptides, it is useful to distinguish between conserved and variable amino residues using the homology comparison tables provided herein. To preserve LP function, it is preferable that conserved residues remain unaltered and that the conformational folding of the LP functional sites be preserved. Preferably, alterations of non-conserved residues are carried out with conservative alterations (e.g., a basic amino acid is replaced by a different basic amino acid). To produce altered function variants, it is preferred to make non-conservative changes at variable and or conserved residues. Deletions at conserved and variable residues can also be used to create altered function variants. Although site-specific mutation sites are predetermined, mutants need not be site- specific. LP protein mutagenesis can be conducted by making amino acid insertions or deletions. Substitutions, deletions, insertions, or any combinations may be generated to arrive at a final construct. Insertions include amino- or carboxyl- terminal fusions, e.g. epitope tags. Random mutagenesis can be conducted at a target codon and the expressed mutants can then be screened for the desired activity. Methods for making substitution mutations at predetermined sites in DNA having a known sequence are well known in the art (e.g., by Ml 3 primer mutagenesis or polymerase chain reaction (PCR) techniques; see also, Sambrook, et al. (cur. ed.) and Ausubel, et al. (cur. ed., and Supplements). The mutations in the DNA normally should not place coding sequences out of reading frames and preferably do not create complementary regions that could hybridize to produce secondary mRNA structure such as loops or hairpins. Recombinant Proteins An LP polypeptide, or fragment thereof, can be used to generate a fusion protein. For example, when fused to a second polypeptide, an LP polypeptide, or fragment thereof, can be used as an antigenic tag or an immunogen. Antibodies raised against an LP polypeptide (or fragment thereof) can be used to indirectly detect a second protein by binding thereto. In one embodiment, if an LP protein has amino acid sequence portion that targets a cellular location (e.g., based on trafficking signals), that portion of the polypeptide can be used by fusing it to another protein (or fragment) to target a protein. Examples of domains that can be fused to an LP polypeptide (or fragment thereof) include, e.g., not only heterologous signal sequences, but also other heterologous functional regions. A fusion does not necessarily need to be direct, but may occur, e.g., through linker sequences. Moreover, fusion proteins may also be engineered to improve characteristics of an LP polypeptide. For instance, a region of additional amino acids, particularly charged amino acids, may be added to the N-terminus of the polypeptide to improve stability and persistence during purification from a host cell or during subsequent handling and storage. In addition, peptide moieties can be added to the polypeptide to facilitate purification. Such regions may be removed before final preparation of the polypeptide. Additions of peptide moieties to facilitate handling are familiar and routine art techniques. Moreover, an LP polypeptide (including any fragment thereof, and specifically an epitope) can be combined with parts of the constant domain of an immunoglobulin e.g., (IgA, IgE, IgG, IgM) portions thereof (CH 1, CH2, CH3), and any combination thereof including both entire domains and portions thereof), resulting in a chimeric polypeptide. Such fusion proteins can facilitate purification and often are useful to increase the in vivo half-life of the protein. For example, this has been demonstrated for chimeric proteins comprising the first two domains of a human CD4 polypeptide and various domains of the constant regions of the heavy or light chains of mammalian immunoglobulins (EP 394,827, Traunecker, et al. (1988) Nature, 331:84-86). Fusion proteins with disulfide-linked dimeric structures (due to the IgG domain) can also be more efficient in binding and neutralizing other molecules than a monomeric secreted protein or sole protein fragment (Fountoulakis, et al. (1995) J. Biochem.15 270:3958-3964). Enhanced delivery of an antigen across an epithelial barrier to the immune system has been demonstrated for antigens (e.g., insulin) conjugated to an FcRn binding partner such as IgG or Fc fragments (see, e.g., WO 96/22024 and WO 99/104813). IgG fusion proteins that have a disulfide-linked dimeric structure due to the IgG portion disulfide bonds have also been found more efficient in binding and neutralizing other molecules than monomeric polypeptides or fragments thereof alone (Fountoulakis, et al. (1995) J. Biochem. 270:3958-3964). Additionally, a fusion protein can comprise various portions of the constant region of an immunoglobulin molecule together with a human protein (or part thereof) EP-A-O 464 533 (Canadian counterpart 2045869). In many cases, the Fc part in a fusion protein is beneficial in therapy and diagnosis, and thus, can result in, e.g., improved pharmacokinetic properties (EP-A 0232262.). Alternatively, deleting the Fc part after the fusion protein has been expressed, detected, and purified, may be desired. For example, the Fc portion may hinder therapy and/or diagnosis if the fusion protein is used as an immunogen for immunizations. In drug discovery for example, human proteins, such as hIL-5, have been fused with Fc portions for the purpose of high-throughput screening assays to identify hIL-5 antagonists (Bennett, et al. (1995) I. Molecular Recognition 8:52-58; and Johanson, et al. (1995) J. Biol. Chem. 270:9459-9471). Furthermore, new constructs may be made by combining similar functional domains from other proteins. For example, protein-binding or other segments may be "swapped" between different new fusion polypeptides or fragments (see, e.g., Cunningham, et al. (1989) Science 243:1330-1336; and O'Dowd, et al. (1988) J. Biol. Chem. 263:15985-15992). Thus, new chimeric polypeptides exhibiting new combinations of specificities will result from the functional linkage of protein-binding specificities and other functional domains. Moreover, an LP polypeptide (or fragment thereof) can be fused to a marker sequence, such as a peptide, to facilitate purification. In preferred embodiments, the marker amino acid sequence is a hexa-histidine peptide, such as, e.g., the tag provided in a pQE vector (QIAGEN, Inc., Chatsworth, CA, 91311), which provides for convenient purification of the fusion protein (Gentz, et al. (1989) Proc. Natl. Acad. Sci. USA 86:821- 824). Another useful peptide-purification tag is the "HA" tag, which corresponds to an epitope derived from an influenza hemagglutinin protein (Wilson, et al. (1984) Cell 37:767). Nucleic acid molecules containing LP polynucleotide sequences encoding an LP epitope can also be recombined with a gene of interest as an epitope tag (e.g., the "HA" or flag tag) to aid in detection and purification of the expressed polypeptide. For example, one system purifies non-denatured fusion proteins expressed in human cell lines (Janknecht, et al. (1991) Proc. Natl. Acad. Sci. USA 88:8972-897). In this system, a gene of interest is subcloned into a vaccinia recombination plasmid such that the open reading frame of the sequence of interest is translationally fused to an ammo-terminal tag consisting of six histidine residues. The tag serves as a matrix-binding domain for the fusion protein. Extracts from cells infected with the recombinant vaccinia virus are loaded onto Ni2+ nitriloacetic acid-agarose column and histidine-tagged proteins can be selectively eluted with imidazole-containing buffers. Additionally, LP fusion constructions may be generated through the techniques of gene-shuffling, motif-shuffling, exon shuffling, and/or codon shuffling (collectively referred to as "DNA shuffling"). DNA shuffling may be employed to modulate an activity of an LP polypeptide. Such methods can be used to generate LP polypeptides (or fragments thereof) with altered activity, as well as agonists and antagonists of an LP polypeptide (see, e.g., U.S. Patent Nos. 5,605,793; 5,811,238; 5,830,721; 5,834,252; and 5,837,458, and Patten, et al. (1997) Cur. Opinion Biotechnol. 8:724-33 30; Harayama, (1998) Trends Biotechnol. 16(2):76-82; Hansson, et al. (1999) J. Mol. Biol. 287:265-76; and Lorenzo and Blasco, (1998) Biotechniques 24(2): 308-13; each of which is incorporated by reference for these DNA shuffling teachings). In another embodiment, an LP polynucleotide, or its encoded LP polypeptide or fragment thereof, may be altered using random mutagenesis by error-prone PCR, random nucleotide insertion, or other methods before recombination. Similar concepts apply to heterologous nucleic acid sequences. Thus, any of type fusion described herein, can be easily engineered using an LP polynucleotide sequence (or fragment thereof) or an LP polypeptide (or fragment thereof) of the present invention. VIII. Functional Variants The blocking of physiological response to an LP protein may result from the inhibition of binding of the protein to its binding partner (e.g., through competitive inhibition). Thus, in vitro assays of the present invention will often use isolated protein, membranes from cells expressing a recombinant membrane associated LP protein, soluble fragments comprising binding segments of these proteins, or fragments attached to solid phase substrates. These assays also allow for the diagnostic determination of the effects either of binding segment mutations and modifications, or of protein mutations and modifications (e.g., protein analogs). This invention also contemplates the use of competitive drug screening assays, e.g., where neutralizing antibodies to antigen or binding partner fragments compete with a test compound for binding to the protein. In this manner, the antibodies can be used to detect the presence of a polypeptide which shares one or more antigenic binding sites of the protein and can also be used to occupy binding sites on the protein that might otherwise interact with a binding partner. "Derivatives" of LP protein antigens include amino acid sequence mutants, glycosylation variants, and covalent or aggregate conjugates with other chemical moieties. Covalent derivatives can be prepared by linkage of functionalities to groups which are found in LP protein amino acid side chains or at the N- or C- termini, by any art known means. These derivatives can include, without limitation, aliphatic esters or amides of the carboxyl terminus, or of residues containing carboxyl side chains, O-acyl derivatives of hydroxyl group-containing residues, and N-acyl derivatives of the amino terminal amino acid or amino-group containing residues, e.g., lysine or arginine. Acyl groups are selected from the group of alkyl-moieties including C3 to C18 normal alkyl, thereby forming alkanoyl aroyl species. Covalent attachment to carrier proteins may be important when immunogenic moieties are haptens. In particular, glycosylation alterations are also encompassed by this invention (e.g., by modifying the glycosylation patterns of a polypeptide during its synthesis and processing, or in further processing steps). Particularly preferred means for accomplishing this are by exposing the polypeptide to glycosylating enzymes derived from cells that normally provide such processing (e.g., mammalian glycosylation enzymes). Deglycosylation enzymes are also contemplated. Also embraced are versions of the same primary amino acid sequence that have other minor modifications, including phosphorylated amino acid residues (e.g., phosphotyrosine, phosphoserine, or phosphothreonine, or other moieties, including ribosyl groups or cross-linking reagents). The invention encompasses a polypeptide of the invention (or fragment thereof) that is differentially modified during or after translation, e.g., by glycosylation, acetylation, phosphorylation, amidation, derivatization by using protecting/blocking groups, proteolytic cleavage, linkage to an antibody molecule, or linkage to another cellular ligand, etc. Any chemical modification may be carried out using known art techniques, including, e.g., without limit, chemical cleavage by, e.g., cyanogen bromide, trypsin, chymotrypsin, papain, V8 protease, or NaBH; acetylation, formulation, oxidation, reduction; metabolic synthesis in the presence of tunicamycin; etc. Additional post- translational modifications encompassed by the invention include, e.g., without limit, N- linked, or O-linked carbohydrate chains, processing of N-terminal or C-terminal ends, attachment of chemical moieties to the amino acid backbone, chemical modifications of N-linked or O-linked carbohydrate chains, and addition or deletion of an N-terminal methionine residue, e.g., because of prokaryotic host cell expression. The polypeptides or fragments thereof may also be modified with a detectable label, such as an enzymatic, fluorescent, isotopic, or affinity label to permit detection and/or isolation. Also provided by the invention is a chemically modified derivative of a polypeptide of the invention (or fragment thereof) that may provide additional advantages such as increased solubility, increased stability increased circulating time, or decreased immunogenicity or antigenicity (see U.S. Patent no: 4,179,337). A chemical moieties for derivatization may be selected from water soluble polymers such as, e.g., polyethyleneglycol, ethylene glycol, propylene glycol, copolymers, carboxymethylcellulose, dextran, polyvinyl alcohol, etc. A polypeptide of the invention, (or fragment thereof) may be modified at random or at predetermined positions within the molecule and may include, e.g., one, two, three, or more attached chemical moieties. The polymer may be of any molecular weight, and may be branched or unbranched. For polyethylene glycol, a preferred molecular weight is between about 1 kDa and about 100 kDa (the term "about" means that in polyethylene glycol preparations, some molecules will weigh more and some will weigh less, than the stated molecular weight). Other sizes may be used, depending on the desired effect (e.g., the [period of sustained release, the effects, if any, on biological activity, ease in handling, the degree or lack of antigenicity, and other known effects of polyethylene glycol on a protein, polypeptide or an analog). Polyethylene glycol molecules (or other chemical moieties) should be attached with consideration of the effect on functional, immunogenic, and/or antigenic domains of a polypeptide (or fragment thereof). Attachment methods include; e.g., without limit, (coupling PEG to G-CSF); EP 0 401 384, pegylating GM-CSF using tresyl chloride (Malik, et al. (1992) Exp. Hematol. 20:1028-1035). For example, polyethylene glycol may be covalently bound through amino acid residues via a reactive group, such as, e.g., a free amino or carboxyl group. Reactive groups are those to which an activated polyethylene glycol molecule may be bound. Amino acid residues having a free amino group may include, e.g., lysine residues, and N-terminal amino acid residue. Amino acid residues having a free carboxyl group may include, e.g., aspartic acid residues, glutamic acid residues, and C-terminal amino acid residues. Sulfhydryl groups may also be used to attach to a polyethylene glycol molecule. For human, a preferred attachment is at an amino group, such as, e.g., an attachment at the N-terminus or a lysine group. One may specifically desire a protein, or a polypeptide (or fragment thereof) that is 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 a protein (polypeptide) molecule in the reaction mix, the type of pegylation reaction to be performed, and the method of obtaining the selected N-terminally pegylated, e.g., polypeptide. The method of obtaining an N-terminally pegylated preparation (by, e.g., separating this moiety from other monopegylated moieties if necessary) may be by purification of the N-terminally pegylated material from a population of pegylated protein molecules. Selective protein chemical modification at the N-terminus 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 appropriate reaction conditions, substantially selective derivatization of a protein or polypeptide (or fragment thereof) at the N-terminus with a carbonyl-group-containing-polymer is achieved. A maj or group of derivatives are covalent conjugates of an LP protein (or fragments thereof) with other proteins or polypeptides. These derivatives can be synthesized in recombinant culture such as N- or C-terminal fusions or by the use of agents known in the art for their usefulness in cross-linking proteins through reactive side groups. Preferred protein derivatization sites with cross-linking agents are at free amino groups, carbohydrate moieties, and cysteine residues. Fusion polypeptides between LP protein and other homologous or heterologous proteins are also provided. Heterologous polypeptides may be fusions between different surface markers, resulting in, for example, a hybrid protein exhibiting binding partner specificity. Likewise, heterologous fusions may be constructed which would exhibit a combination of properties or activities of the derivative proteins. Typical examples are fusions of a reporter polypeptide, e.g., luciferase, with a segment or domain of a protein, e.g., a segment involved in binding partner interaction, so that the presence or location of the fused protein may be easily determined (see, e.g., Dull, et al., U.S. Patent No. 4,859,609). Other gene fusion partners include bacterial D-galactosidase, trpE, Protein A, β-lactamase, alpha amylase, alcohol dehydrogenase, and yeast alpha mating factor (see, e.g., Godowski, et al. (1988) Science 241 :812-816). The fusion partner can be constructed such that it can be cleaved off such that a protein of substantially natural length is generated. Such polypeptides may also have amino acid residues that have been chemically modified by phosphorylation, sulfonation, biotinylation, or the addition or removal of other moieties, particularly those that have molecular shapes similar to phosphate groups. In some embodiments, the modifications will be useful labeling reagents, or serve as purification targets, e.g., affinity proteins. This invention also encompasses the use of derivatives of an LP protein other than variations in amino acid sequence or glycosylation. Such derivatives may involve covalent or aggregative association with chemical moieties. Generally, these derivatives fall into the three classes: (1) salts, (2) side chain and terminal residue covalent modifications, and (3) adsorption complexes (e.g., with cell membranes). Such covalent or aggregative derivatives are useful as immunogens, as reagents in immunoassays, or in purification methods such as for affinity purification of proteins or other binding proteins. For example, a LP protein antigen can be immobilized by covalent bonding to a solid support such as cyanogen bromide-activated SEPHAROSE, by methods which are well known in the art, or adsorbed onto polyolefin surfaces, with or without glutaraldehyde cross-linking, for use in an assay or purification of anti-LP protein antibodies or its respective binding partner. An LP protein can also be labeled for use in diagnostic assays with a detectable group (such as, e.g., radioiodinated by the chloramine T procedure; covalently bound to rare earth chelates; or conjugated to another fluorescent moiety). Purification of an LP protein may be effected by immobilized antibodies or a binding partner. Isolated LP protein genes will allow transformation of cells lacking expression of corresponding LP protein (e.g., either species types or cells that lack corresponding proteins and exhibit negative background activity). Expression of transformed genes will allow isolation of antigenically pure cell lines, with defined or single specie variants. This approach allows for detection that is more sensitive and discrimination of the physiological effects of LP binding proteins. Subcellular fragments, e.g., cytoplasts or membrane fragments, can also be isolated and used. A polypeptide of the invention (or fragment thereof) may be as a monomer or a multimer (e.g., a dimer, a trimer, a tetramer, or a higher multimer). Accordingly, the present invention encompasses monomers and multimers of a polypeptide of the invention, (or fragment thereof) including, e.g., their preparation, and compositions (preferably, therapeutic compositions) containing them. In specific embodiments, the polypeptides and/or fragments of the invention are monomers, dimers, trimers, tetramers or higher multimers. In additional embodiments, a multimer of the invention is at least a dimer, at least a trimer, or at least a tetramer. Multimers encompassed by the invention may be homomers or heteromers. As used herein, the term "homomer," refers to a multimer containing only a specific polypeptide (or fragment thereof) corresponding to an amino acid sequence of SEQ ID NO:Y or encoded by a cDNA contained in a deposited clone (including fragments, variants, splice variants, and fusion proteins, corresponding to these polypeptides as described herein). A homomer may, contain a polypeptide having identical or different amino acid sequences. In a specific embodiment, a homomer of the invention is a multimer containing only polypeptides (or fragments thereof) having identical amino acid sequences. In another specific embodiment, a homomer of the invention is a multimer containing polypeptides having different amino acid sequences. In specific embodiments, a multimer of the invention is a homodimer (e.g., containing polypeptides having identical and/or different amino acid sequences) or a homotrimer (e.g., containing polypeptides having identical and/or different amino acid sequences). In additional embodiments, the homomeric multimer of the invention is at least a homodimer, at least a homotrimer, or at least a homotetramer. As used herein, the term "heteromeric," refers to a multimer containing one or more heterologous polypeptides. In a specific embodiment, a multimer of the invention is a heterodimer, a heterofrimer, or a heterotetramer. In additional embodiments, the heteromeric multimer of the invention is at least a heterodimer, at least a heterofrimer, or at least a heterotetramer. Multimers of the invention may be the result of hydrophobic, hydrophilic, ionic and/or covalent associations and/or may be indirectly linked, by e.g., liposome formation. Thus, in one embodiment, a multimer of the invention, such as, e.g., homodimers or homotrimers, are formed when polypeptides of the invention (or fragments thereof) contact one another in solution. In another embodiment, a heteromultimer of the invention, such as, e.g., a heterofrimer or a heterotetramer, is formed when, e.g., a polypeptide of the invention contacts an antibody (generated against a polypeptide; or fragment thereof of the invention (including antibodies to the heterologous polypeptide sequence in a fusion protein of the invention)) in solution. In other embodiments, a multimer of the invention is formed by covalent association with and/or between a polypeptide and a binding partner such as mentioned herein (or fragment thereof). Such covalent associations may involve one or more amino acid residues contained in a polypeptide sequence (e.g., as recited in a sequence listing herein, or contained in a polypeptide encoded by a deposited clone specified herein). In one instance, a covalent association is a cross-link, e.g., between cysteine residues. In another instance, the covalent associations are the consequence of chemical or recombinant manipulation. Alternatively, such covalent associations may involve one or more amino acid residues contained in a heterologous polypeptide sequence such as, e.g., a fusion protein of the invention. In one example, covalent associations form with a heterologous sequence contained in a fusion protein of the invention (see, e.g., US Patent No. 5,478,925). In a specific example, a covalent association is between a heterologous sequence contained in an Fc fusion protein of the invention (as described herein). In another specific example, a covalent association of a fusion protein of the invention is with a heterologous polypeptide sequence such as, e.g., oseteoprotegerin (see, e.g., WO 98149305, incorporated by reference for these teachings). In another embodiment, two or more polypeptides of the invention (or fragment thereof) are joined through peptide linkers. Examples include, e.g., peptide linkers described in U.S. Pat. No. 5,073,627 (incorporated by reference for these teachings). A protein comprising multiple polypeptides of the invention that are separated by peptide linkers may be produced using conventional recombinant DNA technology. Another method for preparing multimer polypeptides of the invention involves fusing a polypeptide of the invention (or fragment thereof) to a leucine zipper or an isoleucine zipper polypeptide sequence. Leucine zipper and isoleucine zipper domains promote multimerization of polypeptides in which they are found. Leucine zippers were originally identified in several DNA-binding proteins (Landschulz, et al., Science 240: 1759, (1988)), and have been found since in a variety of different proteins. Among known leucine zippers are naturally occurring peptides and derivatives thereof that dimerize or trimerize. Examples of leucine zipper domains suitable for producing soluble, multimeric polypeptides of the invention are those described in, e.g., WO 94/10308, (incorporated by reference for these teachings). Recombinant fusion proteins comprising a polypeptide of the invention (or fragment thereof) fused to a polypeptide sequence that dimerizes or trimerizes in solution can be expressed in a suitable host cell. The resulting soluble multimeric fusion protein can be recovered from a supernatant using any art known technique or method described herein. Trimeric polypeptides of the invention may offer an advantage of enhanced biological activity (as defined herein). Preferred leucine zipper moieties and isoleucine moieties are those that preferentially form trimers. An example is a leucine zipper derived from lung surfactant protein D (SPD), as described in Hoppe, et al. FEBS Letters 344: 19 1,15(1994) and in U.S. patent application Ser. No. 08/446,922, (each hereby incorporated by reference for these teachings). Other peptides derived from naturally occurring trimeric proteins may be employed when preparing a trimeric polypeptide of the invention. In another example, polypeptides or proteins of the invention are associated by interactions with a Flag polypeptide sequence (e.g., contained in a fusion protein of the invention having a Flag sequence). In a further embodiment, a protein or a polypeptide of the invention is associated by an interaction with a heterologous polypeptide sequence (contained in a Flag fusion protein of the invention) and an anti-Flag antibody. A multimer of the invention may be generated using chemical art known techniques. For example, polypeptides (or fragments thereof) desired to be contained in a multimer of the invention may be chemically cross-linked using a linker molecule e.g., linker molecules and linker molecule length optimization techniques are known in the art; see, e.g., US Patent No. 5,478,925, which is incorporated by reference for such teachings. Additionally, a multimer of the invention may be generated using techniques known in the art to form one or more inter-molecule cross-links between the cysteine residues (see, e.g., US Patent No. 5,478,925, incorporated by reference for these teachings). Further, a polypeptide of the invention modified by the addition of cysteine or biotin to the C or N- terminus of a polypeptide can be generated by art known methods (see, e.g., US Patent No. 5,478,925, incorporated by reference for these teachings). Additionally, a multimer of the invention can be generated by art known methods (see, e.g., US Patent No. 5,478,925, incorporated by reference for these teachings). Alternatively, a multimer of the invention can be generated using other commonly known genetic engineering techniques. In one embodiment, a polypeptide contained in a multimer of the invention is produced recombinantly with fusion protein technology described herein or otherwise known in the art (see, e.g., US Patent No. 5,478,925, incorporated by reference for these teachings). In a specific embodiment, a polynucleotide encoding a homodimer of the invention can be generated by ligating a polynucleotide sequence encoding a polypeptide (or fragment thereof) of the invention to another sequence encoding a linker polypeptide and then subsequently, further to a synthetic polynucleotide encoding the translated product of the polypeptide in the reverse orientation from the original C-terminus to the N-terminus (lacking the leader sequence) (see, e.g., US Patent No. 5,478,925, incorporated by reference for these teachings). In another embodiment, recombinant techniques described herein or otherwise known in the art can be applied to generate a recombinant polypeptide of the invention (or fragment thereof) that contains a transmembrane domain (or hyrophobic or signal peptide) and that can be incorporated by membrane reconstitution techniques into a liposome (see, e.g., US Patent No. 5,478,925, incorporated by reference for these teachings).
Uses of an LP polynucleotide sequence An LP polynucleotide sequence (or fragment thereof) can be used in numerous ways, e.g., such as a reagent. The following descriptions (using known art techniques) are non-limiting examples of ways to use an LP polynucleotide sequence (or fragment thereof). For example, an LP polynucleotide sequence (or fragment thereof) is useful for chromosome identification. There exists an ongoing need to identify new chromosome markers, since few chromosome-marking reagents, based on actual sequence data (repeat polymorphisms), are presently available. Each polynucleotide of the present invention can therefore, be used as a chromosome marker. Briefly, sequences can be mapped to a chromosome by preparing PCR primers (preferably 15-25 bp in length) from a sequence taught herein, e.g., a polynucleotide shown in SEQ ID NO:X. Primers can be selected using computer analysis so that they do not span more than one predicted exon in genomic DNA. These primers are then used for PCR screening of somatic cell hybrids containing individual chromosomes such as, e.g., human chromosomes. Only hybrids containing a polynucleotide sequence corresponding to a polynucleotide sequence of the invention, e.g., a sequence of SEQ ID NO: X will yield an amplified fragment. Similarly, somatic hybrids provide a rapid method of PCR mapping a polynucleotide sequence of the invention (or fragment thereof) to a particular chromosome without undue experimentation. Moreover, sub-localization of an LP polynucleotide sequence (or fragment thereof) can be achieved using panels of specific chromosome fragments. Other gene mapping strategies that can be used include, e.g., in situ hybridization, prescreening with labeled flow-sorted chromosomes, and pre-selection by hybridization to construct chromosome specific-cDNA libraries. Precise chromosomal location of a polynucleotide can also be achieved using fluorescence in situ hybridization (FISH) of a metaphase chromosomal spread. The FISH technique can use a polynucleotide sequence of about 500-600 bases in length; however, a polynucleotide length of 2,000-4,000 bp is preferred (see, e.g., Verma, et al. (1988) "Human Chromosomes: a Manual of Basic Techniques," Pergamon Press, New York). For chromosome mapping, an LP polynucleotide sequence (or fragment) can be used individually (to mark a single chromosome or a single site on that chromosome) or in panels (for marking multiple sites and/or multiple chromosomes). Preferably an LP polynucleotide sequence (or fragment thereof) corresponds to a non-coding region since coding sequences are more likely to be conserved within gene families, thus increasing the chance of non-specific cross hybridization during chromosome mapping. Once an LP polynucleotide sequence (or a fragment thereof) has been mapped to a precise chromosomal location, the physical position of the polynucleotide on the chromosome can be used in linkage analysis. Linkage analysis can be used to establish correlation between a chromosomal location and disease, syndrome, disorder or presentation of a particular condition (e.g., diseases associated with chromosomal mapping can be found, e.g., in V. McKusick, Mendelian Inheritance in Man (available on line through Johns Hopkins University
Welch Medical Library)). Assuming a one megabase mapping resolution and one gene per 20 kb of length of DNA, a cDNA precisely localized to a chromosomal region associated with a disease, syndrome, disorder or condition could be one of approximately 50-500 potential causative genes. Once correlation is established, differences in a polynucleotide sequence and a corresponding gene can be examined between an affected individual and an individual unaffected by a particular disease, syndrome, disorder, or condition. Typically, visible structural alterations in a chromosome, such as, e.g., deletions or translocations, are examined in chromosome spreads or by using PCR. If no structural alterations are found, then the presence or absence of point mutations is determined. A mutation in a sequence of interest that correlates with some or all individuals affected with a particular disease, syndrome, disorder or condition but that is not found in individuals without the disease, syndrome disorder or condition suggests that the mutation in the sequence may be the cause of the disease, syndrome, disorder or condition. Furthermore, expression of an LP polynucleotide sequence (or fragment thereof) in an individuals as compared to another individual can be accomplished by the present invention to screen for individuals that have a particular condition, disorder, syndrome or disease state. A typical alteration (e.g., altered expression, chromosomal rearrangement, or mutation) can be used as a diagnostic or prognostic marker. Thus, the invention provides a method useful during diagnosis of a disease, syndrome, disorder or condition involving measuring the level of a polynucleotide mRNA, fragment, or degradation product of the present invention (or fragment thereof) in, e.g., a cell, tissue, sample, or fluid from an individual and comparing, e.g., a polynucleotide mRNA, fragment, or degradation product level with a corresponding standard level, whereby an increase or decrease in a level compared to a standard indicates or prognosticates a disease, syndromes, disorder or condition, or tendency to develop such a disease, syndromes, disorder or condition. In still another embodiment, the invention encompasses a kit, e.g., for analyzing a sample for the presence of a polynucleotide associated with a proliferative disease, syndrome, disorder, or condition. In a general embodiment, the kit includes, e.g., at least an LP polynucleotide sequence (or fragment thereof) probe containing a polynucleotide sequence that hybridizes with an LP polynucleotide sequence(or fragment thereof) and directions, e.g., such as for disposal. In another specific embodiment, a kit includes, e.g., two polynucleotide probes defining an internal region of an LP polynucleotide sequence, where each probe has one strand containing a 31 mer-end internal to a region the polynucleotide. In a further embodiment, a probe may be useful as a primer for amplification using a polymerase chain reaction (PCR). Where a diagnosis of a disease, syndrome, disorder or condition has already been made according to conventional methods, the present invention is useful as a prognostic indicator, for a subject exhibiting an enhanced or diminished expression of an LP polynucleotide sequence (or fragment thereof) by comparison to a subject expressing the polynucleotide of the present invention (or fragment thereof) at a level nearer a standard level. The phrase, "measuring level of a composition of the present invention" is intended to mean herein measuring or estimating (either qualitatively and/or quantitatively) a level of, e.g., a polypeptide (or fragment thereof), or a polynucleotide (or fragment thereof) including, e.g., mRNA, DNA, or cDNA, in a first sample (e.g., preferably a biological sample) either directly (e.g., by determining or estimating an absolute protein or mRNA level) or relatively (e.g., by comparing to a polypeptide or mRNA level in a second sample). In one embodiment, the level in the first sample is measured or estimated from an individual having, or suspected of having, a disease, syndrome, disorder or condition and comparing that level to a second level, wherein the second level is obtained from an individual not having and/or not being suspected of having a disease, syndrome, disorder or condition. Alternatively, the second level is determined by averaging levels from a population of individuals not having or suspected of having a disease, syndrome, disorder, or condition. As is appreciated in the art, once a standard level is determined, it can be used repeatedly as a standard for comparison. A "biological sample" is intended to mean herein any sample comprising biological material obtained from, using, or employing, e.g., an organism, body fluid, exudate, lavage product, waste product, cell (or part thereof), cell line, organ, biopsy, tissue culture, or other source originating from, or associated with, a living cell, tissue, organ, or organism, which contains, e.g., a polypeptide (or fragment thereof), a protein (or fragment thereof), a mRNA (or fragment thereof), or polynucleotide sequence (or fragment thereof) of the present invention, including, e.g., without limitation, a sample such as from, e.g., hair, skin, blood, saliva, semen, vomit, synovial fluid, amniotic fluid, breast milk, lymph, pulmonary sputum, urine, fecal matter, a lavage product, etc. As indicated, a biological sample can include, e.g., without limitation, body fluids
(e.g., such as semen, lymph, sera, plasma, urine, synovial fluid and spinal fluid) that contain a polypeptide (or fragment thereof), mRNA (or fragment thereof), a protein (or fragment thereof), or polynucleotide (or fragment thereof) of the present invention, by product, or, waste product; and/or other tissue source found to express a polypeptide (or fragment thereof), mRNA (or fragment thereof), or nucleic acid (or fragment thereof), by product, or, waste product; of the present invention. Methods for obtaining biological samples, e.g., tissue biopsies, body fluids, cells, or waste products from mammals are known in the art. Where the biological sample is to include, e.g., mRNA, a tissue biopsy is a preferred source. The method(s) provided herein may preferably be applied in a diagnostic method and/or a kit in which a polynucleotide and/or an LP polypeptide (or fragment thereof) are attached to a solid support. In an exemplary method, a support may be a "gene chip" or a "biological chip" as described in, e.g., US Patents 5,837,832; 5,874,219; 5,856,174; 5,700,637 and European Patent 0-373-203 (each of which is incorporated by reference herein for these teachings). Moreover, such a gene chip comprising an LP polynucleotide sequence(or fragment thereof) may be used, e.g., to identify polymorphisms between a polynucleotide sequence from one source, and a polynucleotide from a second, third, or multiple sources.
Knowledge of such polymorphisms (e.g., their location, as well as their extent) is useful to identify, e.g., a location associated with a disease, syndromes, disorder, or condition including, e.g., a cell proliferative disease (see e.g., US Patents 5,858,659, and 5,856,104; each incorporated by reference herein for these teachings). The present invention further encompasses an LP polynucleotide sequence (or fragment thereof) that is chemically synthesized, or reproduced as a peptide nucleic acid (PNA) using art known methods. The use of a PNA is preferred if a polynucleotide (or a fragment thereof) is incorporated, e.g., onto a solid support, or genechip. For the purposes of the present invention, a peptide nucleic acid (PNA) is a polyamide type of polynucleotide analog in which, generally, e.g., the monomeric units for adenine, guanine, thymine and cytosine are available commercially (see, e.g., Perceptive Biosystems). Certain components of a polynucleotide, such as DNA, like phosphorus, phosphorus oxides, or deoxyribose derivatives, are not present in a PNA. Generally, PNAs bind specifically and tightly to complementary DNA strands and are not degraded by nucleases (Nielsen, et al. (1993) Nature 365: 666). In fact, a PNA binds more strongly to DNA than DNA binds to itself, probably, as there is no electrostatic repulsion between PNA/DNA; furthermore, the PNA polyamide backbone is more flexible than DNA. Because of this, PNA/DNA duplexes can bind under a wider range of stringency conditions than DNA/DNA duplexes thus, making it easier to perform multiplex hybridizations. Moreover, smaller probes can be used with PNA than with DNA due to the strong binding. In addition, it is more likely that single base mismatches can be determined using a PNA/DNA hybridization since, e.g., a single mismatch in a PNA/DNA 15-mer lowers the melting point (Tm) by 8°-20°C, versus lowering the melting point 4°-16°C for the DNA/DNA 15-mer duplex. In addition, the absence of charge groups in a PNA molecule means that hybridizations can be done at low ionic strengths and the absence of charge groups with the DNA reduces possible interference by salt. The present invention is also useful for detecting a cell proliferative condition, e.g., such as cancer, in a mammal. In particular the invention is useful during diagnosis of pathological cell proliferative neoplasias like, e.g., without limit: acute myelogenous leukemias including acute monocytic leukemia, acute myeloblastic leukemia, acute promyelocytic leukemia, acute myelomonocytic leukemia, acute erythroleukemia, acute megakaryocyticleukemia, and acute undifferentiated leukemia, etc.; and chronic myelogenous leukemias including chronic myelomonocytic leukemia, chronic granulocyticleukemia, etc. Typically, a preferred mammal includes, e.g., a primate, a monkey, a cat, a dog, a cow, a pig, a sheep, a goat, a horse, and a rabbit. Particularly preferred are human primates. Pathological cell proliferative diseases, disorders, syndromes, and/or conditions are often associated with inappropriate activation of proto-oncogenes. Neoplasias can result from, e.g., a qualitative alteration of a normal cellular gene product, or from a quantitative modification of nucleic acid expression by insertion of a viral sequence, by chromosomal translocation of a polynucleotide sequence to a more actively transcribed region, or by some other mechanism. It is likely that mutated or altered expression of a specific polynucleotide sequence e.g., is involved in the pathogenesis of some leukemias. Indeed, the human counterparts of oncogenes involved in some animal neoplasias have been amplified or translocated in some cases of human leukemia and carcinoma (see, e.g., Gelmann, et al. (1985) "The Etiology of Acute Leukemia: Molecular Genetics and Viral Oncology," inNeoplastic Diseases of the Blood, Vol. 1., Wiemik, et al. eds. pp. 161-182). For example, c-myc expression is highly amplified in the non-lymphocytic leukemia cell line HL-60. When HL-60 cells are chemically induced to stop proliferation, the level of c-myc is found to be down-regulated (WO91/15580). However, exposure of HL-60 cells to a DNA construct that is complementary to the 5' end of c-myc or c-myb blocks translation of the corresponding mRNAs which down regulates expression of the c-myc or c-myb proteins and causes arrest of cell proliferation and differentiation of the treated cells (see, e.g., WO 91115580; Wickstrom, et al (1988) Proc. Natl. Acad. Sci. 85:1028 ; Anfossi, et al. (1989) Proc. Natl. Acad. Sci. 86:3379). However, in light of the numerous cells and cell types of varying origins that are known to exhibit a proliferative phenotype, a skilled artisan would appreciate that the present invention's usefulness is not limited to the treatment of proliferative diseases, disorders, syndromes, and/or conditions of hematopoietic cells and tissues. In addition to the foregoing, an LP polynucleotide sequence (or fragment thereof) can be used to control polynucleotide expression through triple helix formation, or antisense DNA, or antisense RNA, in which binding of a polynucleotide sequence is to a complementary stretch of DNA or RNA. Preferably, the polynucleotide sequence used to contour expression is an oligonucleotide about 20-40 bases in length that is typically complementary to a target region of a polynucleotide sequence involved in transcription. Typically, triple helix formation blocks RNA transcription from DNA, while antisense RNA hybridization blocks mRNA translation. Either technique can be used to design antisense or triple helix polynucleotides to treat, prevent, or ameliorate a disease or condition associated with cell proliferation when coupled with the sequence information disclosed herein, (see, e.g., J. Okano, (1991) Neurochem. 56:560;"Oligodeoxynucleotides as Antisense Inhibitors of Gene Expression CRC Press, Boca Raton, FL (1988); Lee, et al. (1979) Nucleic Acids Research 6: 3073; Cooney, et al. (1988) Science 241 : 456; and Dervan, et al. (1991) Science 251:1360). An LP polynucleotide sequence (or fragment thereof) is also useful in polynucleotide delivery. One goal of polynucleotide delivery is to insert a polynucleotide sequence into an organism so that it is stably expressed. Polynucleotides of the invention (or fragments thereof), offer a means, e.g., of targeting a genetic defect in a highly accurate manner. Another goal is to insert a polynucleotide sequence that is not normally present in a host genome. An LP polynucleotide sequence is also useful to identify an individual from a sample such as, e.g., a biological sample. For instance, the United States military is considering the use of restriction fragment length polymorphism (RFLP) analysis to identify specific military personnel. In this technique, genomic DNA from a sample is digested with one or more restriction enzymes, and subsequently probed on a Southern blot to yield unique bands that can correspond to a specific individual. This method is an improvement over current identification means, e.g., "Dog Tags" which can be lost, switched, or stolen, making positive individual identification difficult. A polynucleotide sequence (or fragment thereof) of the present invention can also be used as an additional DNA marker for RFLP analysis. A polynucleotide sequence (or fragment thereof) of the present invention can also be used as an alternative to RFLP, by determining the actual base-by-base DNA sequence of selected portions of an individual's genome. Such polynucleotide sequences can be used to prepare PCR primers for amplifying and isolating selected portions of DNA, which can then be sequenced. Using a RFLP technique, an individual can be identified because each individual has a unique set of DNA sequences. Once an ID database is established for a particular individual, then a positive identification of a biological sample from that individual (living or dead) can be made. Forensic techniques can also benefit from using a DNA-based identification technique as disclosed herein. A polynucleotide sequence taken from a very small biological sample such as a tissue, (e.g., hair or skin), or a body fluid, (e.g., blood, saliva, semen, synovial fluid, amniotic fluid, breast milk, lymph, pulmonary sputum or surfactant, urine), or fecal matter, etc., can be amplified using a polymerase chain reactor. In a prior art technique, polynucleotide sequences amplified from polymorphic loci, such as a DQa class II HLA gene, can be used to identify an individual.(see, e.g., Erlich (1992) "PCR Technology", Freeman and Co.). Once a specific polymorphic loci is amplified, it can be digested with one or more restriction enzymes, yielding a specific set of identifying bands on a Southern blot when probed with DNA sequence corresponding to a DQa class II HLA gene. Similarly, an LP polynucleotide sequence (or fragment thereof) can be used as a polymorphic marker for identification purposes. There is also a need for reagents capable of identifying the source of a particular sample, e.g., a tissue. Such need arises, e.g., in a forensic investigation when, e.g., a tissue sample is of unknown origin. An appropriate reagent can comprise, e.g., a DNA probe, or primer that is specific to a particular tissue, which is prepared from a polynucleotide sequence of the present invention. Panels of such reagents can then be used to identify tissue by, e.g., species and/or by tissue or organ type. In a similar fashion, such reagents can be used to screen tissue cultures for contamination by, e.g., a non-specific tissue. Furthermore, an LP polynucleotide sequence can be used to create a unique polynucleotide sequence identifier, which can be placed in a material that needs future verification or authentication, e.g., in clothing, explosives, food stuffs, seed lots, etc. A reliable, duplication-proof means of authenticating goods is needed as counterfeit goods in the United States amount to approximately $200 billion a year. Using an LP polynucleotide sequence (or fragment thereof) as a template, a unique sequence can be amplified, e.g., using PCR techniques, to supply sufficient quantities of the unique sequence identifier so that it can be embedded in a material for future identification, validation, and/or authentication. For example, an ink or similar marker can be laced with a unique DNA sequence(s) to insure authenticity and to identify counterfeiting in areas such as, e.g., pharmaceuticals or cosmetics, fine arts, sports collectibles, or to secure documents and financial instruments, including, e.g., passports, currency, and ID cards (see, e.g., DNA Technologies of Los Angeles, USA). Additionally, an LP polynucleotide sequence (or fragment thereof) can be used as a molecular weight marker e.g., on a Southern gel, as a diagnostic probe for the presence of a specific mRNA in a particular cell type, as a probe to "subtract-out" known sequences in the process of discovering novel polynucleotides, for selecting and making oligomers for attachment to a "gene chip" or other support, as an immunogen, e.g., to raise anti-DNA antibodies using DNA immunization techniques, or as an antigen to elicit an immune response.
Uses of LP Polypeptides An LP polypeptide (or fragment thereof), can be used in numerous ways. The following descriptions are non-limiting, exemplars that use art known techniques. A polypeptide (or fragment thereof) can be used to assay a protein level, e.g., of a secreted protein, in a sample, e.g., such as a bodily fluid by using antibody-based techniques. For example, protein expression in a tissue can be studied by an immunohistological method (see, e.g., Jalkanen, et al. (1985) J. Cell Biol. 101:976-985; Jalkanen, et al. (1987) J. Cell Biol. 105:3087-303096). Another useful antibody-based method for detecting protein or polypeptide expression includes, e.g., an immunoassay like an enzyme linked immunosorbent assay or a radioimmunoassay (RIA). Suitable labels for an antibody assay are known in the art and include without limit, e.g., enzyme labels, such as e.g., glucose oxidase, and radioisotopes, such as, e.g., iodine (1251, 13 1), carbon (14C), sulfur (35S), tritium (3H), indium (112In), and technetium (99Tc); and fluorescent labels, such as, e.g., fluorescein, rhodamine, or biotin. In addition to assaying, e.g., the level of a secreted protein in a sample, a protein can also be detected by in vivo imaging. Antibody labels or markers for in vivo imaging of a protein (or polypeptide) include, e.g., those detectable by X-radiography, NMR or ESR. A suitable label for X-radiography, includes, e.g., a radioisotope such as barium or cesium, which emits detectable radiation but is not detrimental to a subject. Suitable markers for NMR and ESR include, e.g., those with a detectable characteristic spin, such as, e.g., deuterium, which may be incorporated into an antibody by labeling, e.g., the nutrients of a particular hybridoma. A protein-specific antibody or antibody fragment that has been labeled with an appropriate detectable imaging moiety, such as a radioisotope (e.g., 1311, 112In, 99Tc), a radio-opaque substance, or a material detectable by nuclear magnetic resonance, can be introduced into a subject (e.g., parenterally, subcutaneously, or intraperitoneally). The subject's size and the imaging system used will both effect the amount of an imaging moiety that is needed to produce a diagnostic image. Typically, for a human subject using a radioisotope moiety, the quantity of the imaging moiety ranges from about 5 to 20 millicuries of label, e.g., 99Tc. A labeled antibody or antibody fragment preferentially accumulates at the location of cells that contain the targeted protein or polypeptide (see, e.g., Burchiel, et al. (1982) "l munopharmacokinetics of Radiolabeled Antibodies and Their Fragments." (Chapter 13 in Tumor Imaging: The Radiochemical Detection of Cancer, S.W. Burchiel and B. A. Rhodes, eds., Masson Publishing Inc.) Thus, the invention provides a means for detecting, marking, locating or diagnosing a disease, syndrome, syndrome, disorder, and/or condition comprising assaying the expression of a polynucleotide (or fragment thereof), or a polypeptide (or fragment thereof), of the present invention that is in a sample, e.g., cells or body fluid of an individual by comparing one level of expression with another level of expression, e.g., a standard level of expression to indicate, e.g., a disease, syndrome, disorder, and/or condition, (or predilection to the same), or to make a prognosis or prediction. With respect to a cell proliferation condition, e.g., such as cancer, the presence of a high level of expression in a sample relative to another lower level or lower standard level may indicate a predisposition for development of a disease, syndrome, or it may provide a means for condition, or state detecting a pre-clinical disease, condition, syndrome, state, or disorder before the appearance of clinical symptoms by other means. Such a use may be beneficial by allowing early intervention thereby preventing and/or ameliorating the development or further progression of the condition. Furthermore, an LP polypeptide (or fragment thereof)can be used to treat, prevent, modulate, ameliorate, and/or diagnose a disease, syndrome, condition, and/or a disorder. For example, a subject can be administered a polypeptide (or fragment thereof) of the invention to replace absent or decreased levels of a polynucleotide or polypeptide (e.g., insulin); to supplement absent or decreased levels of a different polynucleotide or polypeptide (e.g., hemoglobin S for hemoglobin B; SOD to catalyze DNA repair proteins); to inhibit the activity of a polynucleotide or polypeptide (e.g., an oncogene or tumor suppressor); to activate a polynucleotide or polypeptide (e.g., by binding to a receptor), to reduce activity of a membrane bound receptor by competing with the receptor for free ligand (e.g., soluble TNF receptors can be used to reduce inflammation), or to bring about a desired response (e.g., blood vessel growth inhibition, enhancement of an immune response to proliferating cells or to an infectious agent). Similarly, an antibody directed to a polypeptide (or fragment thereof) of the present invention can also be used to treat, prevent, modulate, ameliorate, and/or diagnose a condition, syndrome, state, disease or disorder. For example, administration of an antibody directed to an LP polypeptide (or fragment thereof)can bind and reduce the level of the targeted polypeptide. Similarly, administration of an antibody can activate an LP polypeptide (or fragment thereof), such as by binding to the polypeptide that is bound to a membrane (e.g., a receptor). Polypeptides of the present invention can also be used as molecular weight markers on SDS-PAGE gels or on molecular sieve gel filtration columns using methods known to those of skill in the art. Both the naturally occurring and the recombinant forms of the LP proteins of this invention are particularly useful in kits and assay methods which are capable of screening compounds for binding activity to the proteins. Several methods of automating assays have been developed in recent years to permit screening of tens of thousands of -Ill- compounds in a short period. See, e.g., Fodor, et al. (1991) Science 251:161-113, and other descriptions of chemical diversity libraries, which describe means for testing of binding affinity by a plurality of compounds. The development of suitable assays can be greatly facilitated by the availability of large amounts of purified, soluble LP protein as provided by this invention. For example, antagonists can normally be found once the protein has been structurally defined. Testing of potential protein analogs is now possible upon the development of highly automated assay methods using a purified binding partner. In particular, new agonists and antagonists will be discovered by using screening techniques described herein. Of particular importance are compounds found to have a combined binding affinity for multiple LP protein binding components, e.g., compounds which can serve as antagonists for species variants of a LP protein. This invention is particularly useful for screening compounds by using recombinant protein in a variety of drug screening techniques. The advantages of using a recombinant protein in screening for specific binding partners include: (a) improved renewable source of the LP protein from a specific source; (b) potentially greater number of binding partners per cell giving better signal to noise ratio in assays; and (c) species variant specificity (theoretically giving greater biological and disease specificity). One method of drug screening uses eukaryotic or prokaryotic host cells, which are stably transformed with recombinant DNA molecules expressing a LP protein-binding counterpart. Cells may be isolated which express a binding counterpart in isolation from any others. Such cells, either in viable or fixed form, can be used for standard protein binding assays. See also, Parce, et al. (1989) Science 246:243-247; and Owicki, et al. (1990) Proc. Natl. Acad. Sci. USA 87:4007-4011, which describe sensitive methods to detect cellular responses. Competitive assays are particularly useful, where the cells (source of LP protein) are contacted and incubated with a labeled binding partner or antibody having known binding affinity to the protein, such as ^^I-antiboάy, and a test sample whose binding affinity to the binding composition is being measured. The bound and free-labeled binding compositions are then separated to assess the degree of protein binding. The amount of test compound bound is inversely proportional to the amount of labeled binding partner binding to the known source. Any one of numerous techniques can be used to separate bound from free protein to assess the degree of protein binding. This separation step could typically involve a procedure such as adhesion to filters followed by washing, adhesion to plastic followed by washing, or centrifugation of the cell membranes. Viable cells could also be used to screen for the effects of drugs on LP protein mediated functions, e.g., second messenger levels, i.e., cell proliferation; inositol phosphate pool changes, transcription using a luciferase-type assay; and others. Some detection methods allow for elimination of a separation step, e.g., a proximity-sensitive, detection system. Another method uses membranes from transformed eukaryotic or prokaryotic host cells as the source of a LP protein. These cells are stably transformed with DNA vectors directing the expression of a LP protein, e.g., an engineered membrane bound form. Essentially, the membranes would be prepared from the cells and used in a protein- binding assay such as the competitive assay set forth above. Still another approach is to use solubilized, unpurified or solubilized, purified LP protein from transformed eukaryotic or prokaryotic host cells. This allows for a "molecular" binding assay with the advantages of increased specificity, the ability to automate, and high drug test throughput. Another technique for drug screening involves an approach which provides high throughput screening for compounds having suitable binding affinity to a LP protein antibody and is described in detail in Geysen, European Patent Application 84/03564, published on September 13, 1984. First, large numbers of different small-peptide test compounds are synthesized on a solid substrate, e.g., plastic pins or some other appropriate surface, see Fodor, et al., supra. Then all the pins are reacted with solubilized- unpurified or solubilized-purifϊed LP protein antibody, and washed. The next step involves detecting bound LP protein antibody. Rational drug design may also be based upon structural studies of the molecular shapes of the LP protein and other effectors or analogs. See, e.g., Methods in Enzymology vols. 202 and 203. Effectors may be other proteins that mediate other functions in response to protein binding, or other proteins that normally interact with the binding partner. One means for determining which sites interact with specific other proteins is a physical structure determination, e.g., x-ray crystallography or 2 dimensional NMR techniques. These will provide guidance as to which amino acid residues form molecular contact regions. For a detailed description of protein structural determination, see, e.g., Blundell and Johnson (1976) Protein Crystallography Academic Press, NY. A purified LP protein can be coated directly onto plates for use in the aforementioned drug screening techniques. However, non-neutralizing antibodies to these proteins can be used as capture antibodies to immobilize the respective protein on the solid phase. At least one and up to a plurality of test compounds (such as, e.g., antibodies, oligonucleotides, proteins (e.g., receptors), or small molecules) may be screened for specific binding to an LP polypeptide (or fragment thereof). In another embodiment, the identified compound is closely related to the natural ligand of LP, e.g., a ligand or fragment thereof, a natural substrate, a structural or functional mimetic, or a natural binding partner. (See, e.g., Coligan, et al. (1991) Current Protocols in Immunology l(2):Chapter 5.) Similarly, the compound can be closely related to, e.g., a natural receptor to which LP (or fragment thereof) binds, or to at least a fragment of the receptor, e.g.,. the ligand binding site. In either case, the compound can be rationally designed using known techniques. An assay may test binding of a test compound to the polypeptide, wherein binding is detected by, e.g., a fluorophore, radioisotope, enzyme conjugate, or other detectable label. For example, the assay may comprise combining at least one test compound with an LP polypeptide (or fragment thereof), either in solution or affixed to a solid support, and detecting binding of LP to the compound. Alternatively, the assay may detect or measure binding of a test compound in the presence of a labeled competitor. Additionally, the assay may be carried out using cell-free preparations, chemical libraries, or natural product mixtures, and the test compound(s) may be free in solution or affixed to a solid support. An LP polypeptide (or fragment thereof) may also be used to screen for compounds that modulate the activity of an LP polypeptide (or fragment thereof). Such compounds may include agonists, antagonists, or partial or inverse agonists. In one embodiment, an assay is performed under conditions permissive for an LP polypeptide (or fragment thereof) activity, wherein an LP polypeptide (or fragment thereof) is combined with at least one test compound, and the activity of an LP polypeptide (or fragment thereof) in the presence of a test compound is compared with the activity of LP in the absence of the test compound. A change in the activity of LP in the presence of the test compound is indicative of a compound that modulates the activity of LP. Alternatively, a test compound is combined with an in vitro or cell-free system comprising an LP polypeptide (or fragment thereof) under conditions suitable for LP activity, and the assay is performed. In either of these assays, a test compound which modulates an activity of an LP polypeptide (or fragment thereof) may do so indirectly and need not come in direct contact with the test compound. At least one and up to a plurality of test compounds may be screened. In addition, one could identify a molecule that binds an LP polypeptide (or fragment thereof) by using a beta-pleated sheet region(s) contained in the LP sequence. Accordingly, a specific embodiment of the invention is directed to an LP polynucleotide sequence encoding the corresponding polypeptide comprising, or alternatively consisting of, an amino acid sequence of a beta pleated sheet region in a disclosed polypeptide sequence of the invention. Additional embodiments of the invention are directed to a polynucleotide (or fragment thereof), encoding a polypeptide (or fragment thereof), that comprises, or alternatively consists of all of the beta pleated sheet regions contained in a polypeptide sequence of the invention or any combination thereof. Additional preferred embodiments of the invention are directed to a polypeptide that comprises, or alternatively consists of, an amino acid sequence comprising one, two, three, four, five, six, seven, eight, nine, ten, or more beta-pleated sheets of an LP polypeptide (or any combination thereof.)
Therapeutic Uses This invention also provides reagents with significant therapeutic value. An LP protein or polypeptide (naturally occurring or recombinant), fragments thereof, and antibodies thereto, along with compounds identified as having binding affinity to an LP, are useful in the treatment of conditions associated with abnormal physiology or development, including abnormal proliferation, e.g., cancerous conditions, or degenerative conditions. Abnormal proliferation, regeneration, degeneration, and atrophy may be modulated by appropriate therapeutic treatment using a composition(s) provided herein. For example, a disease or disorder associated with abnormal expression or abnormal signaling by a LP protein is a target for an agonist or antagonist of the protein. Other abnormal developmental conditions are known in cell types shown to possess LP mRNA by northern blot analysis (see, e.g., Berkow (ed.) The Merck Manual of Diagnosis and Therapy, Merck & Co., Rahway, N.J.; Thom et al. Harrison's Principles of Internal Medicine, McGraw-Hill, N.Y.; and Rich (ed.) Clinical Immunology; Principles and Practice, Mosby, St. Louis (cur. ed.); and below). Developmental or functional abnormalities, (e.g., of the neuronal, immune, or hematopoetic system) cause significant medical abnormalities and conditions which may be susceptible to prevention or treatment using compositions provided herein. Recombinant LP or LP antibodies can be purified and administered to a subject for treatment. These reagents can be combined for use with additional active or inert ingredients, e.g., in conventional pharmaceutically acceptable carriers or diluents, e.g., immunogenic adjuvants, along with physiologically innocuous stabilizers and excipients. These combinations can be sterile filtered and placed into dosage forms as by lyophilization in dosage vials or storage in stabilized aqueous preparations. This invention also contemplates use of antibodies or binding fragments thereof, including forms which are not complement binding. Another therapeutic approach included within the invention involves direct administration of reagents, formulations, or compositions by any conventional administration techniques (such as, e.g., without limit, local injection, inhalation, or systemic administration) to a subject. The reagents, formulations, or compositions included within the bounds and metes of the invention may also be targeted to a cell by any of the methods described herein (e.g., polynucleotide delivery techniques). The actual dosage of reagent, formulation, or composition that modulates a disease, disorder, condition, syndrome, etc., depends on many factors, including the size and health of an organism, however one of one of ordinary skill in the art can use the following teachings describing methods and techniques for determining clinical dosages (see, e.g., Spilker (1984) Guide to Clinical Studies and Developing Protocols, Raven Press Books, Ltd., New York, pp. 7-13, 54-60; Spilker (1991) Guide to Clinical Trials. Raven Press, Ltd., New York, pp. 93-101; Craig and Stitzel (eds. 1986) Modem Pharmacology, 2d ed., Little, Brown and Co., Boston, pp. 127-33; Speight (ed. 1987) Avery's Drug Treatment: Principles and Practice of Clinical Pharmacology and Therapeutics, 3d ed., Williams and Wilkins, Baltimore, pp. 50-56; Tallarida, et al. (1988) Principles in General
Pharmacology, Springer-Verlag, New York, pp. 18-20; and U.S. Pat. Nos. 4,657,760; 5,206,344; and 5,225,212.). Generally, in the range of about between 0.5 fg/ml and 500 Dg/ml inclusive final concentration are administered per day to a human adult in any pharmaceutically acceptable carrier. Furthermore, animal experiments provide reliable guidance for the determination of effective does for human therapy. Interspecies scaling of effective doses can be performed following art known principles (e.g., see, Mordenti and Chappell (1989) "The Use of Interspecies Scaling in Toxicokinetics," in
Toxicokinetics and New Drug Development; Yacobi, et al. (eds.) Pergamon Press, NY). Effective doses can also be extrapolated using dose-response curves derived from in vitro or animal-model test systems. For example, for antibodies a dosage is typically 0.1 mg/kg to 100 mg/kg of a recipients body weight. Preferably, a dosage is between 0.1 mg kg and 20 mg/kg of a recipients body weight, more preferably 1 mg/kg to 10 mg/kg of a recipients body weight. Generally, homo-specific antibodies have a longer half-life than hetero-specific antibodies, (e.g., human antibodies last longer within a human host than antibodies from another species, e.g., such as a mouse, probably, due to the immune response of the host to the foreign composition). Thus, lower dosage of human antibodies and less frequent administration is often possible if the antibodies are administered to a human subject. Furthermore, 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) by using modifications such as, e.g., lipidation. The invention also provides a pharmaceutical pack or kit comprising one or more containers filled with one or more of the ingredients of the compositions of the invention and instructions such as, e.g., for disposal (typically, in a form prescribed by a governmental agency regulating the manufacture, use or sale of pharmaceuticals or biological products). The quantities of reagents necessary for effective treatment will depend upon many different factors, including means of administration, target site, physiological state of the patient, and other medicaments administered. Thus, treatment dosages should be titrated to optimize safety and efficacy. Typically, dosages used in vitro may provide useful guidance in the amounts useful for in situ administration of these reagents. Animal testing of effective doses for treatment of particular disorders will provide further predictive indication of human dosage. Various considerations are described, e.g., in Gilman, et al. (eds.) (1990) Goodman and Gilman's: The Pharmacological Bases of Therapeutics (8th ed.) Pergamon Press; and (1990) Remington's Pharmaceutical Sciences (17th ed.) Mack Publishing Co., Easton, PA. Methods for administration are discussed therein and below, e.g., for oral, intravenous, intraperitoneal, or intramuscular administration, transdermal diffusion, and others. Pharmaceutically acceptable carriers will include water, saline, buffers, and other compounds described, e.g., in the Merck Index, Merck & Co., Rahway, NJ. Dosage ranges would ordinarily be expected to be in amounts lower than 1 mM concentrations, typically less than about 10 μM concentrations, usually less than about 100 nM, preferably less than about 10 pM (picomolar), and most preferably less than about 1 fM (femtomolar), with an appropriate carrier. Slow release formulations, or a slow release apparatus will often be utilized for continuous administration. LP protein, fragments thereof, and antibodies to it or its fragments, antagonists, and agonists, may be administered directly to the host to be treated or, depending on the size of the compounds, it may be desirable to conjugate them to carrier proteins such as ovalbumin or serum albumin prior to their administration. Therapeutic formulations may be administered in any conventional dosage formulation. While it is possible for the active ingredient to be administered alone, it is preferable to present it as a pharmaceutical formulation. Formulations typically comprise at least one active ingredient, as defined above, together with one or more acceptable carriers thereof. Each carrier should be both pharmaceutically and physiologically acceptable in the sense of being compatible with the other ingredients and not injurious to the patient. Formulations include those suitable for oral, rectal, nasal, or parenteral (including subcutaneous, intramuscular, intravenous and intradermal) administration. The formulations may conveniently be presented in unit dosage form and may be prepared by any methods well known in the art of pharmacy. See, e.g., Gilman, et al. (eds.) (1990) Goodman and Gilman's: The Pharmacological Bases of Therapeutics (8th ed.) Pergamon Press; and (1990) Remington's Pharmaceutical Sciences (17th ed.) Mack Publishing Co., Easton, PA; Avis, et al. (eds.) (1993) Pharmaceutical Dosage Forms: Parenteral Medications Dekker, NY; Lieberman, et al. (eds.) (1990) Pharmaceutical Dosage Forms: Tablets Dekker, NY; and Lieberman, et al. (eds.) (1990) Pharmaceutical Dosage Forms: Disperse Systems Dekker, NY. The freatment ofthis invention may be combined with or used in association with other therapeutic agents. The present invention also provides a pharmaceutical composition. Such a composition comprises, e.g., a therapeutically effective amount of a composition of the invention in a pharmaceutically acceptable carrier. As used herein, the term "pharmaceutically acceptable carrier" means a carrier approved by a federal regulatory agency of the United States of America, or a regulatory/administrative agency of a state government of the United States or a carrier that is listed in the U.S. Pharmacopeia or other pharmacopeia; which is generally recognized by those in the art for use in an animal, e.g., a mammal, and, more particularly, in a primate, e.g., a human primate. The term "carrier" as used herein refers to a diluent, adjuvant, excipient, or vehicle that is administered with a composition of the invention. A pharmaceutical carrier typically can be a sterile liquid, such as water or oils, (including those of petroleum, animal, vegetable, or synthetic origin, e.g., such as peanut oil, soybean oil, mineral oil, sesame oil and the like). Typically, sterile water is a preferred carrier when a 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, e.g., without limit, starch, glucose, lactose, sucrose, gelatin, malt, rice, flour, chalk, silica gel, sodium stearate, glycerol monostearate, talc, sodium chloride, dried skim milk, glycerol, propylene, glycol, water, ethanol and the like. A composition of the invention, if desired, can also contain minor amounts of wetting or emulsifying agents, or pH buffering agents. A composition of the invention can be in a solution, suspension, emulsion, tablet, pill, capsule, powder, sustained-release formulation, etc., or it can be formulated as a suppository (with traditional binders, and/or carriers, e.g., such as triglycerides). Oral formulations encompassed include, e.g., without limit, standard carriers such as pharmaceutical grades of mannitol, lactose, starch, magnesium stearate, sodium saccharine, cellulose, magnesium carbonate, etc. Additional examples of suitable pharmaceutical carriers are described in the current edition of "Remington's Pharmaceutical Sciences" by E.W. Martin. Such formulations will contain a therapeutically effective amount of a composition of the invention, preferably in purified form, together with a suitable amount of carrier to provide for proper administration to a subject. Traditionally, a formulation will suit the mode of administration. In a preferred embodiment, a composition is formulated in accordance with routine procedures as a pharmaceutical composition adapted for intravenous administration to, e.g., a human. Typically, compositions for intravenous administration are solutions in sterile isotonic aqueous buffer. Where necessary, the composition may also include, e.g., a solubilizing agent and a local anesthetic such as lidocaine to promote comfort at the injection site. Generally, ingredients are supplied either separately or mixed together in unit dosage form, e.g., as a dry lyophilized powder or water free concentrate in a hermetically sealed container (such as an ampoule or sachet indicating the quantity of active agent). Where a composition is to be administered by infusion, it can be dispensed using an infusion bottle containing sterile pharmaceutical grade water or saline. Where a composition is administered by injection, an ampoule of sterile water for injection or saline can be provided so that the ingredients may be mixed before administration. Compositions of the invention can be formulated as neutral or salt forms. Pharmaceutically acceptable salts include, e.g., without limit, anionic salts (such as those derived from hydrochloric, phosphoric, acetic, oxalic, tartaric acids, etc.,) and cationic salts, (e.g., such as those derived from sodium, potassium, ammonium, calcium, ferric hydroxides, isopropylamine, triethylamine, 2-ethylamino ethanol, histidine, procaine, etc). The amount of the composition of the invention that will be effective in the modulation treatment, inhibition, amelioration, or prevention of a disease, syndrome, condition, or disorder associated with aberrant expression and/or activity of a polypeptide (or fragment thereof), or a polynucleotide (or fragment thereof) of the invention can be determined without undue experimentation by the ordinary artisan using standard clinical techniques. In addition, in vitro assays may optionally be employed to help identify optimal dosage ranges. Dosage requirements in a circumstance typically will depend on, e.g., the route of administration, the seriousness of the disease, condition, syndrome, or disorder; and the judgment of the practitioner or clinician. Another therapeutic approach included within the invention involves direct administration of a composition of the invention by any conventional administration technique (such as, e.g., without limit, local injection, inhalation, or systemic aάjninistration), to a subject with e.g., an infectious, a microbial, a bacterial, a viral or a fungal condition. A composition or formulation may also be targeted to a specific cell or a receptor by any method described herein or known in the art. A use of truncated FVIIa (LP FVIIa) is strongly implicated in modulating inflammatory and angiogenic conditions. For example, the FVIIa-TF pathway has been shown to induce several genes that mediate angiogenesis. For instance, FVIIa induces IL-8 expression in macrophage and fibroblasts (Camerer, E., Gjemes, E., Wiiger, M., Pringel S. and Prydz, H. (2000) J. Biol. Chem. 275, 6580-6585.). IL-8 is a chemoattractant for endothelial cells and monocyte, and promotes angiogenesis in vivo (Radcliffe, R., and Nemerson, Y. (1976) J. Biol. Chem. 251, 4797-4802; Szekanecz, Z., Shah, M. R., Harlow, L. A., Pearce, W. H., Koch, A. E. (1994) Pathology 62, 134-139). FVIIa also induces the expression of the CCN gene family members CYR61 and connective tissue growth factor (CTGF) (Camerer, E., Gjernes, E., Wiiger, M., Pringel S. and Prydz, H. (2000) J. Biol. Chem. 275, 6580-6585). The cystein rich protein CYR61 acts as a ligand for the integrin alpha v beta 3 (Leu, Shr-Jeng., Lam, S. C, Lai, L. F. (2002) J. Boil. Chem. 277, 46248-46255). CYR61 stimulates endothelial cell growth and migration, and in vivo angiogenesis (Kireeva, M, L., Mo, F. E., Yang, G. P., Lau, L. F. 9 1996). Mol Cell Biol 16, 1326-1334; Babic, A. M., Kireeva, K. L., Kolesnikova, T. V., and Lau, L. F. (1998) Proc. Natl. Acad. Sci. U.S.A. 95, 6355-6360). CYR 61 gene knock out mice show gross vascular defects (Mo, F.-E., Mutean, A. G., Chen, C.-C, Stolz, D. B., Watkins, S C, and Lau, L. F. (2002) Mol. Cell. Biol. 22, 8709-8720). Other FVIIa induced angiogenic genes include VEGF and FGF-5 (Ollivier, V., Chabbat. J., Herbert. J. M., Hakim, J., and de Prost. D. (2000). Thromb. Vase. Biol. 20, 1374-1381; Camerer, E., Gjemes, E., Wiiger, M., Pringel S. and Prydz, H. (2000) J. Biol. Chem. 275, 6580-6585). Although FVIIa may induce angiognesis, TF-VIIa activation in vivo is usually associated with the induction of coagulation and platelet activation. Since the LP FVIIa is devoid of the coagulation activity but induces proangiogenesis genes, this molecule may be uniquely suited for angiogenic therapy. Additionally, an LP can be used to modulate hemostatic or thrombolytic activity. For example, increasing hemostatic or thrombolytic activity can treat or prevent a blood coagulation condition such as e.g., afibrinogenemia, a factor deficiency, a blood platelet disease (e.g. thrombocytopenia), or a wound resulting from e.g., trauma, surgery, early wound repair as well as hemostasis, profuse bleeding, and patients with impaired liver function, etc. Other potential uses are for patients with liver disease, anticoagulation- induced bleeding, surgery, thrombocytopenia, thrombasthenia, von Willebrand disease, and other bleeding disorders. Alternatively, a composition of the invention can be used to decrease hemostatic or thrombolytic activity or to inhibit or dissolve a clotting condition. Such compositions can be important in a treatment or prevention of a heart condition, e.g., an attack infarction, stroke, or mycardial scarring. An LP may also be useful in ameliorating, treating, preventing, modulating and/or diagnosing an autoimmune disease, disorder, syndrome, and/or condition such as results, e.g., from the inappropriate recognition by a cell of the immune system of the self as a foreign material. Such an inappropriate recognition results in an immune response leading to detrimental effect destruction on the host, e.g., on a host cell, tissue, protein, or moiety, e.g., a carbohydrate side chain. Therefore, administration of an LP which inhibits a detrimental immune response, particularly, e.g., a proliferation, differentiation, or chemotaxis of a T-cell, may be effective in detecting, diagnosing, ameliorating, or preventing such an autoimmune disease, disorder, syndrome, and/or condition. Examples of autoimmune conditions that can be affected by the present invention include, e.g., without limit Addison's Disease syndrome hemolytic anemia, anti-phospholipid syndrome, rheumatoid arthritis, dermatitis, allergic encephalomyelitis, glomerulonephritis, Goodpasture's Syndrome, Graves' Disease syndrome, Multiple Sclerosis, Myasthenia Gravis, Neuritis, Ophthalmia, BuUous Pemphigoid, Pemphigus, Polyendocrinopathies, Purpura, Reiter's Disease syndrome, Stiff-Man Syndrome, Autoimmune Thyroiditis, Systemic Lupus Erythematosus, Autoimmune Pulmonary Inflammation, Guillain-BarreSyndrome, insulin dependent diabetes mellitis, and autoimmune inflammatory eye disease. Similarly, allergic reactions and conditions, such as asthma (e.g., allergic asthma) or other respiratory problems, may also be ameliorated, treated, modulated or prevented, and/or diagnosed by an LP polynucleotide or polypeptide (or fragment thereof), or an agonist or antagonist thereto. Moreover, such inventive compositions can be used to effect, e.g., anaphylaxis, hypersensitivity to an antigenic molecule, or blood group incompatibility. An LP may also be used to modulate, ameliorate, treat, prevent, and/or diagnose organ rejection or graft- versus-host disease (GVHD). Generally speaking, organ rejection occurs by a host's, immune-cell destruction of a transplanted tissue or cell. A similarly destructive immune response is involved in GVHD, however, in this case, transplanted foreign immune cells destroy host tissues and/or cells. Administration of a composition of the invention, which ameliorates or modulates such a deleterious immune response (e.g., a deleterious proliferation, differentiation, or chemotaxis of a T cell), can be effective in modulating, ameliorating, diagnosing, and/or preventing organ rejection or GVHD. Similarly, an LP may also be used to detect, treat, modulate, ameliorate, prevent, and/or diagnose an inflammation, e.g., by inhibiting the proliferation and/or differentiation of a cell involved in an inflammatory response, or an inflammatory condition (either chronic or acute), including, e.g., without limitation, chronic prostatitis, granulomatous prostatitis and malacoplakia, an inflammation associated with an infection (such as, e.g., septic shock, sepsis, or systemic inflammatory response syndrome (SIRS)), ischemia-reperfusion injury, endotoxin lethality, arthritis, complement-mediated hyperacute rejection, nephritis, cytokine or chemokine induced lung injury, inflammatory bowel disease syndrome, Crohn's disease syndrome, or a condition resulting from an over production of a cytokine(s) (e.g., TNF or IL-1 .)
Anti-Hemopoietic Activity The naturally occurring balance between endogenous stimulators and inhibitors of angiogenesis is one in which inhibitory influences typically predominate (see, e.g.,
Rastinejad, et al., Cell 56345-355 (1989)). When neovascularization occurs under normal physiological conditions, such as wound healing, organ regeneration, embryonic development, and female reproductive processes, angiogenesis is stringently regulated, and delimited spatially and temporally. In pathological angiogenesis such as, e.g., during solid tumor formation, these regulatory controls fail and unregulated angiogenesis can become pathologic by sustaining progression of many neoplastic and non-neoplastic diseases. A number of serious diseases are dominated by abnormal neovascularization (including, e.g., solid tumor growth and metastases, arthritis, some types of eye conditions, and psoriasis; see, e.g., reviews by Moses, et al., Biotech. 9630-634 (1991); Folkman, et al., N. Engl. J. Med., 333: 1757-1763 (1995); Auerbach, et al., J. Microvasc. Res. 29:401-4 11 (1985); Folkman, "Advances in Cancer Research", eds. Klein and Weinhouse, Academic Press, New York, pp. 175-203 (1985); Patz, Am. J. Opthalmol. 94:7 15-743 (1982); and Folkman, et al., Science 221:7 19-725 (1983). In a number of pathological conditions, angiogenesis contributes to a disease- state, e.g., for example, significant data have accumulated suggesting that solid tumor formation is dependent on angiogenesis (see, e.g., Folkman and Klagsbrun, Science 235:442-447 (1987)). In another embodiment of the invention, administration of an LP provides for the treatment, amelioration, modulation, diagnosis, and/or inhibition of a disease, disorder, syndrome, and/or condition associated with neovascularization. Malignant and metastatic conditions that can be effected in a desired fashion using an LP include, e.g., without limitation, a malignancy, solid tumor, and a cancer as described herein or as otherwise known in the art (for a review of such disorders, syndromes, etc. see, e.g., Fishman, et al., Medicine, 2d Ed., J. B. Lippincott Co., Philadelphia (1985)). Thus, the present invention provides a method of ameliorating, modulating, treating, preventing, and/or diagnosing an angiogenesis-related disease and/or disorder, comprising administering to a subject in need thereof a beneficially effective amount of an LP. For example, cancers that may be so affected using a composition of the invention includes, e.g., without limit a solid tumor, including e.g., 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 metastases; melanomas; glioblastoma; Kaposi's sarcoma; leiomyosarcoma; non-small cell lung cancer; colorectal cancer; advanced malignancies; and blood bom tumors such as e.g., leukemia.
XI. Kits This invention also contemplates use of LP proteins, fragments thereof, peptides, and their fusion products in a variety of diagnostic kits and methods for detecting the presence of LP protein or a binding partner. Typically, the kit will have a compartment containing either a defined LP protein peptide or gene segment or a reagent, which recognizes one or the other, e.g., binding partner fragments or antibodies. A kit for determining the binding affinity of a test compound to a LP protein would typically comprise a test compound; a labeled compound, e.g., a binding agent or antibody having known binding affinity for the LP protein; a source of LP protein (naturally occurring or recombinant); and a means for separating bound from free labeled compound, such as a solid phase for immobilizing the LP protein. Once compounds are screened, those having suitable binding affinity to the LP protein are evaluated in suitable biological assays, as are well known in the art, to determine whether they act as agonists or antagonists to the binding partner. The availability of recombinant LP protein or polypeptides also provides well-defined standards for calibrating such assays. A preferred kit for determining the concentration of, e.g., a LP protein in a sample would typically comprise a labeled compound, e.g., binding partner or antibody, having known binding affinity for the LP protein, a source of LP protein (naturally occurring or recombinant), and a means for separating the bound from free labeled compound, for example, a solid phase for immobilizing the LP protein. Compartments containing reagents, and instructions, will normally be provided. Antibodies, including antigen binding fragments, specific for an LP protein or fragments thereof are useful in diagnostic applications to detect the presence of elevated levels of LP protein and or its fragments. Such diagnostic assays can employ lysates, live cells, fixed cells, immunofluorescence, cell cultures, body fluids, and further can involve the detection of antigens related to the protein in serum, or the like. Diagnostic assays may be homogeneous (without a separation step between free reagent and antigen-LP or - WDS protein complex) or heterogeneous (with a separation step). Various commercial assays exist, such as radioimmunoassay (RIA), enzyme-linked immunosorbent assay (ELISA), enzyme immunoassay (EIA), enzyme-multiplied immunoassay technique
(EMIT), substrate-labeled fluorescent immunoassay (SLFIA), and the like. For example, unlabeled antibodies are employed by using a second antibody which is labeled and which recognizes an antibody to a LP protein or to a particular fragment thereof. Similar assays are also extensively discussed in the literature (see, e.g., Harlow and Lane (1988) Antibodies: A Laboratory Manual. CSH Press, NY; Chan (ed.) (1987) Immunoassay: A Practical Guide Academic Press, Orlando, FL; Price and Newman (eds.) (1991) Principles and Practice of Immunoassay Stockton Press, NY; and Ngo (ed.) (1988) Nonisotopic Immunoassay Plenum Press, NY). Anti-idiotypic antibodies may have similar use to diagnose the presence of antibodies against an LP protein or polypeptide, as such may be diagnostic of various abnormal states, conditions, disorders, or syndromes. For example, overproduction of LP protein may result in production of various immunological or other medical reactions which may be diagnostic of abnormal physiological states, e.g., in cell growth, activation, or differentiation. Frequently, the reagents for diagnostic assays are supplied in kits, to optimize the sensitivity of the assay. For the instant invention, depending upon the nature of the assay, the protocol, and the label, either labeled or unlabeled antibody or binding partner, or labeled LP protein is provided. This is usually in conjunction with other additives, such as buffers, stabilizers, materials necessary for signal production such as substrates for enzymes, and the like. Preferably, the kit contains instructions for proper use and disposal of the contents after use. Typically, the kit has compartments for each useful reagent. Desirably, the reagents are provided as a dry lyophilized powder, where the reagents may be reconstituted in an aqueous medium providing appropriate concenfrations of reagents for performing the assay. Many of the aforementioned constituents of the drug screening and the diagnostic assays may be used without modification, or may be modified in a variety of ways. For example, labeling may be achieved by covalently or non-covalently joining a moiety that directly or indirectly provides a detectable signal. In any of these assays, the protein, test compound, LP protein or polypeptide (or antibodies thereto) are labeled either directly or indirectly. Possibilities for direct labeling include label groups such as, e.g., without limitation, radiolabels (e.g., ^^1); enzymes (U.S. Pat. No. 3,645,090) such as peroxidase and alkaline phosphatase; and fluorescent labels (U.S. Pat. No. 3,940,475) that are capable of monitoring the change in fluorescence intensity, wavelength shift, or fluorescence polarization. Possibilities for indirect labeling include biotinylation of one constituent followed by binding to avidin coupled to an art known label such as one of the above. There are also numerous methods of separating the bound from the free protein, or alternatively bound from free test compound. An LP protein is immobilized on various matrices followed by washing. Suitable matrices include plastic such as an ELISA plate, filters, and beads. Methods of immobilizing the LP protein to a matrix include, without limitation, direct adhesion to plastic, use of a capture antibody, chemical coupling, and biotin-avidin. The last step in this approach involves the precipitation of protein binding partner or antigen/antibody complex by any of several methods including those utilizing, e.g., an organic solvent such as polyethylene glycol or a salt such as ammonium sulfate. Other suitable separation techniques include, without limitation, a fluorescein antibody magnetizable particle method described in Rattle, et al. (1984) Clin. Chem. 30:1457- 1461, and the double antibody magnetic particle separation as described in U.S. Pat. No. 4,659,678. Methods for linking proteins or their fragments to the various labels have been extensively reported in the literature and do not require detailed discussion here. Many of the techniques involve the use of activated carboxyl groups either through the use of carbodiimide or active esters to form peptide bonds, the formation of thioethers by reaction of a mercapto group with an activated halogen such as chloroacetyl, or an activated olefin such as maleimide, for linkage, or the like. Fusion proteins will also find use in these applications. Another diagnostic aspect of this invention involves use of oligonucleotide or polynucleotide sequences taken from the sequence of a LP protein. These sequences are used as probes for detecting levels of the LP protein message in samples from natural sources, or patients suspected of having an abnormal condition, e.g., cancer or developmental problem. The preparation of both RNA and DNA nucleotide sequences, the labeling of the sequences, and the preferred size of the sequences has received ample description and discussion in the literature. Normally an oligonucleotide probe should have at least about 14 nucleotides, usually at least about 18 nucleotides, and the polynucleotide probes may be up to several kilobases. Various labels may be employed, most commonly radionuclides, particularly 32p. However, other techniques may also be employed, such as using biotin-modifϊed nucleotides for introduction into a polynucleotide. The biotin then serves as the site for binding to avidin or antibodies, which may be labeled with a variety of labels, such as radionuclides, fluorophores, enzymes, or the like. Alternatively, antibodies may be employed which can recognize specific duplexes, including DNA duplexes, RNA duplexes, DNA-RNA hybrid duplexes, or DNA-protein duplexes. The antibodies in turn may be labeled and the assay carried out where the duplex is bound to a surface, so that upon the formation of duplex on the surface, the presence of antibody bound to the duplex is detected. The use of probes to the novel anti-sense RNA may be carried out using many conventional techniques such as nucleic acid hybridization, plus and minus screening, recombinational probing, hybrid released translation (HRT), and hybrid arrested translation (HART). This also includes amplification techniques such as polymerase chain reaction (PCR). Diagnostic kits, which also test for the qualitative or quantitative presence of other markers, are also contemplated. Diagnosis or prognosis may depend on the combination of multiple indications used as markers. Thus, kits may test for combinations of markers. See, e.g., Viallet, et al. (1989) Progress in Growth Factor Res. 1:89-97. In specific embodiments, a kit may include, e.g., a recombinantly produced or chemically synthesized polypeptide antigen. The polypeptide antigen of the kit may also be attached to a solid support. In a more specific embodiment the detecting means of the above-described kit includes, e.g., a solid support to which said polypeptide antigen is attached. Such a kit may also include, e.g., a non-attached reporter-labeled anti-human antibody. In this embodiment, binding of the antibody to the polypeptide antigen is detected by binding of the reporter-labeled antibody. In an additional embodiment, the invention includes, e.g., a diagnostic kit for use in screening a biological sample, e.g., such as serum, containing an antigen of a polypeptide (or fragment thereof) of the invention. The diagnostic kit can include, e.g., a substantially isolated antibody specifically and/or selectively immunoreactive with a polypeptide or polynucleotide antigen, and, a 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, e.g., a second, labeled monoclonal antibody. Alternatively, or in addition, the detecting means may include, e.g., a labeled, competing antigen. In one diagnostic configuration, test serum is reacted with a solid phase reagent having a surface-bound antigen obtained by an art known method or as described herein. After binding with specific antigen antibody to the reagent and removing unbound serum components, e.g., 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 that is detected by incubating the solid phase in the presence of a suitable fluorometric, luminescent or calorimetric substrate (Sigma, St. Louis, MO). The solid surface reagent in the above assay is prepared by art known techniques for attaching proteinaceous material to a solid support, such as, e.g., polymeric beads, dip sticks, 96-well plate, or filter material. Methods for attachment generally include, e.g., non-specific adsorption of a protein or polypeptide (or fragment thereof) to a solid support or covalent attachment of a polypeptide, protein (or fragment thereof), typically, e.g., through a free amine group, to a chemically reactive group, such as, e.g., an activated carboxyl, hydroxyl, or aldehyde group on the solid support. Alternatively, streptavidin coated plates are used in conjunction with biotinylated antigen(s).
Other Preferred Embodiments Other preferred embodiments of the claimed invention include an isolated or recombinant nucleic acid molecule comprising a polynucleotide sequence that is at least 95% identical to a polynucleotide sequence of at least about: 10, 12, 14, 16, 18, 20, 22, 24, 26, 28, 30, 32, 34, 36, 38, 40, 42, 44, 46, 48, 50, 52, 54, 56, 58, 60, 62, 64, 66, 68, 70, 72, 74, 76, 78, 80, 82, 84, 86, 88, 90, 92, 94, 96, 98, 100, 110, 120, 130, 140, or 150 contiguous nucleotides of a sequence of Table 1. Other preferred embodiments of the claimed invention include an isolated or recombinant nucleic acid molecule comprising a polynucleotide sequence that is at least 95% identical to a polynucleotide sequence of at least about: 10, 12, 14, 16, 18, 20, 22, 24, 26, 28, 30, 32, 34, 36, 38, 40, 42, 44, 46, 48, 50, 52, 54, 56, 58, 60, 62, 64, 66, 68, 70, 72, 74, 76, 78, 80, 82, 84, 86, 88, 90, 92, 94, 96, 98, 100, 110, 120, 130, 140, or 150 contiguous nucleotides of a mature coding portion of a sequence of Table 1. Also preferred is a nucleic acid molecule wherein said sequence of contiguous nucleotides is include, e.g. in the nucleotide sequence of SEQ ID NO:X in the range of positions beginning with the nucleotide at about the position of the 5' nucleotide and ending with the nucleotide at about the position of the 3' nucleotide of a sequence of Table 1. Also preferred is a nucleic acid molecule wherein said sequence of contiguous nucleotides is included, e.g., in the nucleotide sequence of SEQ ID NO:X in the range of positions beginning with the nucleotide at about the position of the 5' nucleotide of the Start Codon and ending with the nucleotide at about the position of the 3' nucleotide of a sequence of Table 1. Similarly preferred is a nucleic acid molecule comprising polynucleotide sequence of SEQ ID NO:X in the range of positions beginning with the nucleotide at about the position of the 5' nucleotide of a correspondingly encoded First Amino Acid of a Signal Peptide and ending with the nucleotide at about the position of the 3' nucleotide of a sequence of Table 1. Also preferred is an isolated or recombinant nucleic acid molecule comprising a polynucleotide sequence that is at least 95% identical to a polynucleotide sequence of at least about: 10, 12, 14, 16, 18, 20, 22, 24, 26, 28, 30, 32, 34, 36, 38, 40, 42, 44, 46, 48, 50, 52, 54, 56, 58, 60, 62, 64, 66, 68, 70, 72, 74, 76, 78, 80, 82, 84, 86, 88, 90, 92, 94, 96, 98, 100, 110, 120, 130, 140, or 150 contiguous nucleotides in at least one polynucleotide sequence fragment of SEQ ID NO:X. More preferably said polynucleotide sequence that is at least 95% identical to one, exhibits 95% sequence identity to at least: 2, 3, 4, 5, 6, 7, 8, 9, 10, or more polynucleotide fragments 22, 24, 26, 28, 30, 32, 34, 36, 38, 40, 42, 44, 46, 48, 50, 52, 54, 56, 58, 60, 62, 64, 66, 68, 70, 72, 74, 76, 78, 80, 82, 84, 86, 88, 90, 92, 94, 96, 98, 100, 110, 120, 130, 140, or 150 contiguous nucleotides in length of the mature coding portion of a sequence of Table 1., wherein any one such fragment is at least 21 contiguous nucleotides in length. Further preferred is an isolated or recombinant nucleic acid molecule comprising a polynucleotide sequence that is at least 95% identical to a polynucleotide sequence of at least about: 200, 250, 300, 350, 400, 450, or 500 contiguous nucleotides of the mature coding portion of a sequence of Table 1. Also preferred is an isolated or recombinant nucleic acid molecule comprising a polynucleotide sequence that is at least 95% identical to a sequence of at least about: 10, 12, 14, 16, 18, 20, 22, 24, 26, 28, 30, 32, 34, 36, 38, 40, 42, 44, 46, 48, 50, 52, 54, 56, 58, 60, 62, 64, 66, 68, 70, 72, 74, 76, 78, 80, 82, 84, 86, 88, 90, 92, 94, 96, 98, 100, 110, 120, 130, 140, or 150 contiguous nucleotides in at least one nucleotide sequence fragment of SEQ ID NO:X, wherein the length of at least one such fragment is about 200, 250, 300, 350, 400, 450, or 500 contiguous nucleotides of a sequence of Table 1. Another preferred embodiment is an isolated or recombinant nucleic acid molecule comprising a polynucleotide sequence that is at least 95% identical to a sequence of Table 1 beginning with the nucleotide at about the position of the 5' Nucleotide of the First Amino Acid of the Signal Peptide and ending with the nucleotide at about the position of the 3' Nucleotide of a sequence of Table 1. A further preferred embodiment is an isolated or recombinant nucleic acid molecule comprising a polynucleotide sequence, which is at least 95% identical to the complete mature coding portion of a sequence of Table 1. Also preferred is an isolated or recombinant nucleic acid molecule comprising polynucleotide sequence that hybridizes under stringent hybridization conditions to a mature coding portion of a polynucleotide of the invention (or fragment thereof), wherein the nucleic acid molecule that hybridizes does not hybridize under stringent hybridization conditions to a nucleic acid molecule having a nucleotide sequence consisting of only A residues or of only T residues. Cardiovascular Conditions A composition of the invention may be used to, modulate, ameliorate, effect, treat, prevent, and/or diagnose a cardiovascular disease, disorder, syndrome, and/or condition. As described herein, including, e.g., without limitation, cardiovascular abnormalities, such as arterio-arterial fistula, arteriovenous fistula, cerebral arteriovenous malformations, congenital heart defects, pulmonary atresia, and Scimitar Syndrome peripheral artery disease, syndrome, such as limb ischemia. Additional cardiovascular disorders encompass, e.g., congenital heart defects which include, e.g., aortic coarctation, car triafriatum, 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 e.g., aortopulmonary septal defect, endocardial cushion defects, Lutembacher's Syndrome, trilogy of Fallot, and ventricular heart septal defects. Further cardiovascular conditions include, e.g., heart disease syndrome, such as, e.g., arrhythmias, carcinoid heart disease syndrome, high cardiac output, low cardiac output, cardiac tamponade, endocarditis (including bacterial endocarditis), heart aneurysm, cardiac arrest, congestive heart failure, congestive cardiomyopathy, paroxysmal dyspnea, cardiac edema, heart hypertrophy, congestive cardiomyopathy, left ventricular hypertrophy, right ventricular hypertrophy, post-infarction heart rupture, ventricular septal rupture, heart valve disease, myocardial disease, myocardial ischemia, pericardial effusion, pericarditis (including constrictive and tuberculous pericarditis), pneumopericardium, post-pericardiotomy syndrome, pulmonary heart disease syndrome, rheumatic heart disease syndrome, ventricular dysfunction, hyperemia, cardiovascular pregnancy complications, Scimitar Syndrome, cardiovascular syphilis, and cardiovascular tuberculosis. Further cardiovascular disorders include, e.g., arrhythmias including, e.g., sinus arrhythmia, atrial fibrillation, afrial flutter, bradycardia, extra systole, Adams-Stokes Syndrome, bundle-branch block, sinoafrial block, long QT syndrome, parasystole, Lown- Ganong-Levine Syndrome, Mahaim-type pre-excitation syndrome, Wolff-Parkinson- White syndrome, sick sinus syndrome, and ventricular fibrillation tachycardias. Tachycardias encompassed with the cardiovascular condition described herein include, e.g., paroxysmal tachycardia, supravenfricular tachycardia, accelerated idioventricular rhythm, atrioventricular nodal re-entry tachycardia, ectopic atrial tachycardia, ectopic junctional tachycardia, sinoafrial nodal re-entry tachycardia, sinus tachycardia, Torsades de Pointes Syndrome, and ventricular tachycardia. Additional cardiovascular disorders include, e.g., heart valve disease such as, e.g., aortic valve insufficiency, aortic valve stenosis, heart 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. Myocardial conditions associated with cardiovascular disease include, e.g., myocardial diseases such as, e.g., alcoholic cardiomyopathy, congestive cardiomyopathy, hypertrophic cardiomyopathy, aortic subvalvular stenosis, pulmonary subvalvular stenosis, restrictive cardiomyopathy, Chagas cardiomyopathy, endocardial fibroelastosis, endomyocardial fibrosis, Kearns Syndrome, myocardial reperfusion injury, and myocarditis. Cardiovascular conditions include, e.g., myocardial ischemias such as, e.g., coronary disease syndrome, such as e.g., angina pectoris, coronary aneurysm, coronary arteriosclerosis, coronary thrombosis, coronary vasispasm, myocardial infarction, and myocardial stunning. Cardiovascular diseases also encompassed herein include, e.g., vascular diseases such as e.g., aneurysms, angiodysplasia, angiomatosis, bacillary angiomatosis, Hippel- Lindau Disease syndrome, Klippel-Trenaunay- Weber Syndrome, Sturge- Weber Syndrome, angioneurotic edema, aortic disease, Takayasu's Arteritis, aortitis, Leriche's Syndrome, arterial occlusive disease, arteritis, enarteritis, polyarteritis nodosa, cerebrovascular disease, diabetic angiopathies, diabetic retinopathy, embolism, thrombosis, erytbromeialgia, hemorrhoids, hepatic veno-occlusive disease syndrome, hypertension, hypotension, ischemia, peripheral vascular diseases, phlebitis, pulmonary veno-occlusive disease syndrome, Raynaud's disease syndrome, CREST syndrome, retinal vein occlusion, Scimitar syndrome, superior vena cava syndrome, telangiectasia, ataxia telangiectasia, hereditary hemorrhagic telangiectasia, varicocele, varicose veins, varicose ulcer, vasculitis, and venous insufficiency. Cardiovascular conditions further include, e.g., aneurysms such as, e.g., dissecting aneurysms, false aneurysms, infected aneurysms, ruptured aneurysms, aortic aneurysms, cerebral aneurysms, coronary aneurysms, heart aneurysms, and iliac aneurysms. Arterial occlusive cardiovascular conditions include, e.g., arteriosclerosis, intermittent claudication, carotid stenosis, fibromuscular dysplasias, mesenteric vascular occlusion, Moyamoya disease syndrome, renal artery obstruction, retinal artery occlusion, and thromboangiitis obliterans. Cerebrovascular cardiovascular conditions include, e.g., carotid artery disease, cerebral amyloid angiopathy, cerebral aneurysm, cerebral anoxia, cerebral arteriosclerosis, cerebral arteriovenous malformation, cerebral artery disease, cerebral embolism and thrombosis, carotid artery thrombosis, sinus thrombosis, Wallenberg's syndrome, cerebral hemorrhage, epidural hematoma, subdural hematoma, subarachnoid hemorrhage, cerebral infarction, cerebral ischemia (including transient cerebral ischemia), subclavian steal syndrome, perivenfricular leukomalacia, vascular headache, cluster headache, migraine, and vertebrobasilar insufficiency., Embolic cardiovascular conditions include, e.g., air embolisms, amniotic fluid embolisms, cholesterol embolisms, blue toe syndrome, fat embolisms, pulmonary embolisms, and thromboembolisms. Thrombotic cardiovascular conditions include, e.g., coronary thrombosis, hepatic vein thrombosis, retinal vein occlusion, carotid artery thrombosis, sinus thrombosis, Wallenberg's syndrome, and thrombophlebitis. Ischemic conditions include, e.g., cerebral ischemia, ischemic colitis, compartment syndromes, anterior compartment syndrome, myocardial ischemia, reperfusion injuries, and peripheral limb ischemia. Vasculitic conditions include, e.g., 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. A binding composition can be beneficial in ameliorating critical limb ischemia and coronary disease. A binding composition may be administered using any art known method, described herein A binding composition may administered as part of a therapeutic composition or formulation, as described in detail herein. Methods of delivering a binding composition are also described herein.
Hemopoietic Activity The naturally occurring balance between endogenous stimulators and inhibitors of angiogenesis is one in which inhibitory influences typically predominate (see, e.g.,
Rastinejad, et al., Cell 56345-355 (1989)). When neovascularization occurs under normal physiological conditions, such as wound healing, organ regeneration, embryonic development, and female reproductive processes, angiogenesis is stringently regulated, and delimited spatially and temporally. In pathological angiogenesis such as, e.g., during solid tumor formation, these regulatory controls fail and unregulated angiogenesis can become pathologic by sustaining progression of many neoplastic and non-neoplastic diseases. A number of serious diseases are dominated by abnormal neovascularization (including, e.g., solid tumor growth and metastases, arthritis, some types of eye conditions, and psoriasis; see, e.g., reviews by Moses, et al., Biotech. 9630-634 (1991); Folkman, et al., N. Engl. J. Med., 333: 1757-1763 (1995); Auerbach, et al., J. Microvasc. Res. 29:401-4 11 (1985); Folkman, "Advances in Cancer Research," eds. Klein and Weinhouse, Academic Press, New York, pp. 175-203 (1985); Patz, Am. J. Opthalmol. 94:7 15-743 (1982); and Folkman, et al., Science 221:7 19-725 (1983). Moreover, TGF beta 1 and its receptors ALK-5 and ALK-1 , are implied in the vascular maturation phase of angiogenesis (see, e.g., Bull Acad Natl Med. 2000;184 (3):537-44). In a number of pathological conditions, angiogenesis contributes to a disease-state, e.g., for example, significant data have accumulated suggesting that solid tumor formation is dependent on angiogenesis (see, e.g., Folkman and Klagsbrun, Science 235:442-447 (1987)). In another embodiment of the invention, administration of a composition of the invention provides for the treatment, amelioration, modulation, diagnosis, and/or inhibition of a disease, disorder, syndrome, and/or condition associated with neovascularization. Malignant and metastatic conditions that can be effected in a desired fashion using a composition of the invention includes, e.g., without limitation, a malignancy, solid tumor, and a cancer as described herein or as otherwise known in the art (for a review of such disorders, syndromes, etc. see, e.g., Fishman, et al., Medicine, 2d Ed., J. B. Lippincott Co., Philadelphia (1985)). Thus, the present invention provides a method of ameliorating, modulating, treating, preventing, and/or diagnosing an angiogenesis-related disease and/or disorder, comprising administering to a subject in need thereof a beneficially effective amount of a composition of the invention. For example, cancers that may be so affected using a composition of the invention includes, e.g., without limit a solid tumor, including e.g., 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 metastases; melanomas; glioblastoma; Kaposi's sarcoma; leiomyosarcoma; non-small cell lung cancer; colorectal cancer; advanced malignancies; and blood born tumors such as e.g., leukemia. Moreover, a composition may be delivered topically, to treat or prevent cancers such as, e.g., skin cancer, head and neck tumors, breast tumors, and Kaposi's sarcoma. Within yet another aspect, a composition of the invention may be utilized to treat superficial forms of bladder cancer by, e.g., intravesical administration into the tumor, or near the tumor site; via injection or a catheter. Of course, the appropriate mode of administration will vary according to the cancer to be treated. Other modes of delivery are discussed herein. A composition of the invention may also be useful in modulating, ameliorating, treating, preventing, and/or diagnosing another disease, disorder, syndrome, and/or condition, besides a cell proliferative condition (e.g., a cancer) that is assisted by abnormal angiogenic activity. Such close group conditions include, e.g., without limitation, benign tumors, e.g., such as hemangiomas, acoustic neuromas, neurofibromas, trachomas, and pyogenic granulomas; atherosclerotic plaques; ocular angiogenic diseases, e.g., diabetic retinopathy, retinopathy of prematurity, macular degeneration, cornea graft rejection, neovascular glaucoma, retrolental fibroplasia, rubeosis, retinoblastoma, uvietis and Pterygia (abnormal blood vessel growth) of the eye; rheumatoid arthritis; psoriasis; delayed wound healing; endomefriosis; 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. As noted above, the present invention also provides methods for ameliorating, treating, preventing, and/or diagnosing neovascular diseases of the eye, including e.g., corneal graft neovascularization, neovascular glaucoma, proliferative diabetic retinopathy, retrolental fibroplasia and macular degeneration. Moreover, ocular diseases, disorders, syndromes, and/or conditions associated with neovascularization that can be modulated ameliorated, treated, prevented, and/or diagnosed with a composition of the invention include, e.g., without limit; neovascular glaucoma, diabetic retinopathy, retinoblastoma, retrolental fibroplasia, uveitis, retinopathy of premature macular degeneration, corneal graft neovascularization, as well as other inflammatory eye diseases, ocular tumors, and diseases associated with choroidal or iris neovascularization (see, e.g., reviews by Waltman, et al., (1978) Am. J. Ophthal. 8.51704-710 and Gartner, et al., (1978) Sun. Ophthd. 22:291-3 12). Thus, within one aspect of the present invention methods are provided for treating or preventing neovascular diseases of the eye such as comeal neovascularization (including comeal graft neovascularization), comprising administering to a patient a therapeutically effective amount of a composition of the invention to the cornea, such that the formation of blood vessels is inhibited or delayed. Briefly, the cornea is a tissue that normally lacks blood vessels. In certain pathological conditions however, capillaries may extend into the cornea from the pericorneal vascular plexus of the limbus. When the cornea becomes vascularized, it also becomes clouded, resulting in a decline in the patient's visual acuity. Visual loss may become complete if the cornea completely opacifies. A wide variety of diseases, disorders, syndromes, and/or conditions can result in comeal neovascularization, including e.g., comeal infections (e.g., trachoma, herpes simplex keratitis, leishmaniasis and onchocerciasis), immunological processes (e.g., graft rejection and Stevens- Johnson's syndrome), alkali bums, trauma, inflammation (of any cause), toxic and nutritional deficiency states, and as a complication of using contact lenses. Within another aspect, methods are provided for treating or preventing neovascular glaucoma, comprising administering to a patient a therapeutically effective amount of a composition of the invention to the eye, such that the formation of blood vessels is inhibited. In one embodiment, the composition may be administered topically to the eye to treat or prevent early forms of neovascular glaucoma. Within other embodiments, the composition may be implanted by injection into the region of the anterior chamber angle. Within other embodiments, the composition may also be placed in any location such that the composition is continuously released into the aqueous humor. Within another aspect, methods are provided for treating or preventing proliferative diabetic retinopathy, comprising administering to a patient a therapeutically effective amount of a composition of the invention to the eyes, such that the formation of blood vessels is inhibited. Additional, diseases, disorders, syndromes, and/or conditions that can be modulated, ameliorated, treated, prevented, and/or diagnosed with a composition of the invention include, e.g., without limitation, hemangioma, arthritis, psoriasis, angiofibroma, atherosclerotic plaques, delayed wound healing, granulations, hemophilic joints, hyperfrophic scars, nonunion fractures, Osier- Weber syndrome, pyogenic granuloma, scleroderma, trachoma, and vascular adhesions. Moreover, diseases, disorders, states, syndromes, and/or conditions that can be modulated, ameliorated, treated, prevented, and/or diagnosed with a composition of the invention include, e.g., without limitation, solid tumors, blood bom tumors such as leukemias, tumor metastasis, Kaposi's sarcoma, benign tumors (e.g., hemangiomas), acoustic neuromas, neurofibromas, trachomas, and pyogenic granulomas, rheumatoid arthritis, psoriasis, ocular angiogenic diseases, e.g., diabetic retinopathy, retinopathy of prematurity, macular degeneration, comeal graft rejection, neovascular glaucoma, retrolental fibroplasia, rubeosis, retinoblastoma, and uvietis, delayed wound healing, endometriosis, vasculogenesis, granulations, hyperfrophic scars (keloids), nonunion fractures, scleroderma, trachoma, vascular adhesions, myocardial angiogenesis, coronary collaterals, cerebral collaterals, arteriovenous malformations, ischemic limb angiogenesis, Osler- Webber Syndrome, plaque neovascularization, telangiectasia, hemophiliac joints, angiofibroma fibromuscular dysplasia, wound granulation, Crohn's disease, syndrome, atherosclerosis, birth-control inhibition of vascularization necessary for embryo implantation during the control of menstruation, and diseases that have angiogenesis as a pathologic consequence such as, e.g., cat scratch disease (Rochele minalia quintosa), ulcers (Helicobacter pylori), Bartonellosis and bacillary angiomatosis. Diseases at the Cellular Level Diseases associated with increased cell survival or the inhibition of apoptosis that could be modulated, ameliorated, freated, prevented, and/or diagnosed by a composition of the invention include, e.g., cancers (such as, e.g., follicular lymphomas, carcinomas with p53 mutations, and hormone-dependent tumors, including, e.g., but without limit, colon cancer, cardiac tumors, pancreatic cancer, melanoma, retinoblastoma, glioblastoma, lung cancer, intestinal cancer, testicular cancer, stomach cancer, neuroblastoma, myxoma, myoma, lymphoma, endothelioma, osteoblastoma, osteoclastoma, osteosarcoma, chondrosarcoma, adenoma, breast cancer, prostate cancer, Kaposi's sarcoma and ovarian cancer); autoimmune conditions (such as, e.g., multiple sclerosis, Sjogren's syndrome, Hashimoto's thyroiditis, biliary cirrhosis, Behcet's disease syndrome, Crohn's disease syndrome, polymyositis, systemic lupus erythematosus, immune-related glomerulonephritis, and rheumatoid arthritis); viral infections (such as, e.g., herpes viruses, pox viruses, and adeno viruses); inflammation; graft v. host disease syndrome, acute graft rejection, and chronic graft rejection. In preferred embodiments, a composition of the invention is used to inhibit growth, progression, and/or metastases of cancers such as, in particular, those listed herein. Additional diseases, states, syndromes, or conditions associated with increased cell survival that could be modulated, ameliorated, freated, prevented, or diagnosed by a composition of the invention include, e.g., without limitation, progression, and/or metastases of malignancies and related disorders such as leukemia including acute leukemias (such as, e.g., acute lymphocytic leukemia, acute myelocytic leukemia, including myeloblastic, promyelocytic, myelomonocytic, monocytic, and erythroleukemia) and chronic leukemias (e.g., chronic myelocytic, chronic 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, syndrome, and solid tumors including, e.g., without limitation, sarcomas and carcinomas (such as, e.g., 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, hepatoma, bile duct carcinoma, choriocarcinoma, seminoma, embryonal carcinoma, Wilm's tumor, cervical cancer, testicular tumor, lung carcinoma, small cell lung carcinoma, bladder carcinoma, epithelial carcinoma, glioma, astrocytoma, medulloblastoma, craniopharyngioma, ependymoma, pinealoma, hemangioblastoma, acoustic neuroma, oligodendroglioma, menangioma, melanoma, neuroblastoma, and retinoblastoma). Diseases associated with increased apoptosis that could be modulated, ameliorated, treated, prevented, and/or diagnosed by a composition of the invention include, e.g., AIDS, conditions (such as, e.g., Alzheimer's disease syndrome, Parkinson's disease syndrome, Amyotrophic lateral sclerosis, Retinitis pigmentosa, Cerebellar degeneration and brain tumor, or prion associated disease); autoimmune conditions (such as, e.g., multiple sclerosis, Sjogren's syndrome, Hashimoto's thyroiditis, biliary cirrhosis, Behcet's disease syndrome, Crohn's disease syndrome, polymyositis, systemic lupus erythematosus, immune-related glomerulonephritis, and rheumatoid arthritis); myelodysplastic syndromes (such as aplastic anemia), graft v. host disease syndrome; ischemic injury (such as that caused by myocardial infarction, stroke and reperfusion injury); liver injury (such as, e.g., hepatitis related liver injury, ischemia reperfusion injury, cholestosis (bile duct injury), and liver cancer); toxin-induced liver disease (such as, e.g., that caused by alcohol), septic shock, cachexia, and anorexia. Moreover, a composition of the invention could be used to prevent and heal damage to the lungs due to various pathological states, such as, e.g., stimulating proliferation and differentiation to promote repair of alveoli and bronchiolar epithelium. For example, emphysema, inhalation injuries, that (e.g., from smoke inhalation) and burns, which cause necrosis of the bronchiolar epithelium and alveoli could be effectively ameliorated, treated, prevented, and/or diagnosed using a polynucleotide or polypeptide of the invention (or fragment thereof), or an agonist or antagonist thereto. Also, a composition of the invention could be used to stimulate the proliferation of and differentiation of type II pneumocytes, to help treat or prevent hyaline membrane diseases, such as e.g., infant respiratory distress syndrome and bronchopulmonary displasia, (in premature infants). A composition of the invention could stimulate the' proliferation and/or differentiation of a hepatocyte and, thus, could be used to alleviate or treat a liver condition such as e.g., fulminant liver failure (caused, e.g., by cirrhosis), liver damage caused by viral hepatitis and toxic substances (e.g., acetaminophen, carbon tetrachloride, and other known hepatotoxins).
Kits This invention also contemplates use of binding compositions in a variety of diagnostic kits and methods for detecting the presence of an LP composition of the invention. Typically, ti e kit will have a compartment containing either a defined an LP composition of the invention or a binding composition, which recognizes one or the other, e.g., binding partner fragments or antibodies. A preferred kit for determining the concentration of an LP composition of the invention in a sample would typically comprise a labeled compound, e.g., binding composition or antibody, having known binding affinity for the an LP composition of the invention protein, a source of an LP composition of the invention(naturally occurring or recombinant), and a means for separating the bound from free labeled compound, for example, a solid phase for immobilizing the an LP composition of the invention protein. Compartments containing reagents, and instructions, will normally be provided. Antibodies, including antigen binding fragments, specific for a an LP composition of the invention or fragments thereof are useful in diagnostic applications to detect the presence of elevated levels of an LP composition of the invention and/or its fragments. Such diagnostic assays can employ lysates, live cells, fixed cells, immunofluorescence, cell cultures, body fluids, and further can involve the detection of antigens related to the protein in serum, or the like. Diagnostic assays may be homogeneous (without a separation step between free reagent and antigen-an LP composition of the invention or - WDS protein complex) or heterogeneous (with a separation step). Various commercial assays exist, such as radioimmunoassay (RIA), enzyme-linked immunosorbent assay (ELISA), enzyme immunoassay (EIA), enzyme-multiplied immunoassay technique (EMIT), substrate-labeled fluorescent immunoassay (SLFIA), and the like. For example, unlabeled antibodies are employed by using a second antibody which is labeled and which recognizes an antibody to an LP composition of the invention or to a particular fragment thereof. Similar assays are also extensively discussed in the literature (see, e.g., Harlow and Lane (1988) Antibodies: A Laboratory Manual, CSH Press, NY; Chan (ed.) (1987) Immunoassay: A Practical Guide Academic Press, Orlando, FL; Price and Newman (eds.) (1991) Principles and Practice of Immunoassay Stockton Press, NY; and Ngo (ed.) (1988) Nonisotopic immunoassay Plenum Press, NY). Anti-idiotypic antibodies may have similar use to diagnose the presence of antibodies against an an LP composition of the invention or polypeptide, as such may be diagnostic of various abnormal states, conditions, disorders, or syndromes. For example, overproduction of an LP composition of the invention may result in production of various immunological or other physiological reactions which may be diagnostic of abnormal physiological states, e.g., in cell growth, activation, or differentiation. Frequently, the reagents for diagnostic assays are supplied in kits, to optimize the sensitivity of the assay. For the instant invention, depending upon the nature of the assay, the protocol, and the label, either labeled or unlabeled antibody or binding composition, or labeled an LP composition of the invention is provided. This is usually in conjunction with other additives, such as buffers, stabilizers, materials necessary for signal production such as substrates for enzymes, and the like. Preferably, the kit contains instructions for proper use and disposal of the contents after use. Typically, the kit has compartments for each useful reagent. Desirably, the reagents are provided as a dry lyophilized powder, where the reagents may be reconstituted in an aqueous medium providing appropriate concentrations of reagents for performing the assay. Many of the aforementioned constituents of the drug screening and the diagnostic assays may be used without modification, or may be modified in a variety of ways. For example, labeling may be achieved by covalently or non-covalently joining a moiety that directly or indirectly provides a detectable signal. In any of these assays, the protein, test compound, an LP composition of the invention or polypeptide (or antibodies thereto) are labeled either directly or indirectly. Possibilities for direct labeling include label groups such as, e.g., without limitation, radiolabels (e.g., 125j). enzymes (U.S. Pat. No. 3,645,090) such as peroxidase and alkaline phosphatase; and fluorescent labels (U.S. Pat. No. 3,940,475) that are capable of monitoring the change in fluorescence intensity, wavelength shift, or fluorescence polarization. Possibilities for indirect labeling include biotinylation of one constituent followed by binding to avidin coupled to an art known label such as one of the above. There are also numerous methods of separating the bound from the free protein, or alternatively bound from free test compound. An LP composition of the invention is immobilized on various matrices followed by washing. Suitable matrices include plastic such as an ELISA plate, filters, and beads. Methods of immobilizing the an LP composition of the invention to a matrix include, without limitation, direct adhesion to plastic, use of a capture antibody, chemical coupling, and biotin-avidin. The last step in this approach involves the precipitation of protein/binding composition or antigen/antibody complex by any of several methods including those utilizing, e.g., an organic solvent such as polyethylene glycol or a salt such as ammonium sulfate. Other suitable separation techniques include, without limitation, a fluorescein antibody magnetizable particle method described in Rattle, et al. (1984) Clin. Chem. 30:1457- 1461, and the double antibody magnetic particle separation as described in U.S. Pat. No. 4,659,678. Methods for linking proteins or their fragments to the various labels have been extensively reported in the literature and do not require detailed discussion here. Many of the techniques involve the use of activated carboxyl groups either through the use of carbodiimide or active esters to form peptide bonds, the formation of thioethers by reaction of a mercapto group with an activated halogen such as chloroacetyl, or an activated olefin such as maleimide, for linkage, or the like. Fusion proteins will also find use in these applications. Diagnostic kits, which also test for the qualitative or quantitative presence of other markers, are also contemplated. Diagnosis or prognosis may depend on the combination of multiple indications used as markers. Thus, kits may test for combinations of markers. See, e.g., Viallet, et al. (1989) Progress in Growth Factor Res. 1:89-97. In specific embodiments, a kit may include, e.g., a recombinantly produced or chemically synthesized polypeptide antigen. The polypeptide antigen of the kit may also be attached to a solid support. In a more specific embodiment the detecting means of the above-described kit includes, e.g., a solid support to which said polypeptide antigen is attached. Such a kit may also include, e.g., a non-attached reporter-labeled anti-human antibody. In this embodiment, binding of the antibody to the polypeptide antigen is detected by binding of the reporter-labeled antibody. In an additional embodiment, the invention includes, e.g., a diagnostic kit for use in screening a biological sample, e.g., such as serum, containing an antigen of a polypeptide (or fragment thereof) of the invention. The diagnostic kit can include, e.g., a substantially isolated antibody specifically and/or selectively immunoreactive with a polypeptide or polynucleotide antigen, and, a 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, e.g., a second, labeled monoclonal antibody. Alternatively, or in addition, the detecting means may include, e.g., a labeled, competing antigen. In one diagnostic configuration, test serum is reacted with a solid phase reagent having a surface-bound antigen obtained by an art known method or as described herein. After binding with specific antigen antibody to the reagent and removing unbound serum components, e.g., 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 that is detected by incubating the solid phase in the presence of a suitable fluorometric, luminescent or calorimetric substrate (Sigma, St. Louis, MO). The solid surface reagent in the above assay is prepared by art known techniques for attaching proteinaceous material to a solid support, such as, e.g., polymeric beads, dip sticks, 96-well plate, or filter material. Methods for attachment generally include, e.g., non-specific adsorption of a protein or polypeptide (or fragment thereof) to a solid support or covalent attachment of a polypeptide, protein (or fragment thereof), typically, e.g., through a free amine group, to a chemically reactive group, such as, e.g., an activated carboxyl, hydroxyl, or aldehyde group on the solid support. Alternatively, streptavidin coated plates are used in conjunction with biotinylated antigen(s). The broad scope of this invention is best understood with reference to the following examples, which are not intended to limit the invention to specific embodiments. EXAMPLES General Methods Many of the standard methods described herein are described or referenced, e.g., in Maniatis, et al. (Cur. ed..) Molecular Cloning, A Laboratory Manual Cold Spring Harbor Laboratory, Cold Spring Harbor Press, NY; Sambrook, et al.; Ausubel, et al, Biology Greene Publishing Associates, Brooklyn, NY; or Ausubel, et al. (1987 and Supplements) Current Protocols in Molecular Biology Wiley/Greene, NY; Innis, et al. (eds.) (1990) PCR Protocols: A Guide to Methods and Applications Academic Press, NY. Methods for protein purification include such methods as ammonium sulfate precipitation, column chromatography, electrophoresis, centrifugation, crystallization, and others. See, e.g., Ausubel, et al. (1987 and periodic supplements); Deutscher (1990) "Guide to Protein Purification," Methods in Enzymology vol. 182, and other volumes in this series; Coligan, et al. (1995 and supplements) Current Protocols in Protein Science John Wiley and Sons, New York, NY; P. Matsudaira (ed.) (1993) A Practical Guide to Protein and Peptide Purification for Microsequencing, Academic Press, San Diego, CA; and manufacturer's literature on use of protein purification products, e.g., Pharmacia, Piscataway, NJ, or Bio-Rad, Richmond, CA. Combination with recombinant techniques allows fusion to appropriate segments (epitope tags), e.g., to a FLAG sequence or an equivalent which can be fused, e.g., via a protease-removable sequence. See, e.g., Hochuli (1989) Chemische Industrie 12:69-70; Hochuli (1990) "Purification of Recombinant Proteins with Metal Chelate Absorbent" in Setlow (ed.) Genetic Engineering. Principle and Methods 12:87-98, Plenum Press, NY; and Crowe, et al. (1992) QIAexpress: The High Level Expression and Protein Purification System QUIAGEN, Inc., Chatsworth, CA. Standard immunological techniques are described, e.g., in Hertzenberg, et al. (eds. 1996) Weir's Hanbook of Experimental Immunology vols. 1-4, Blackwell Science; Coligan (1991) Current Protocols in Immunology Wiley/Greene, NY; and Methods in Enzymology volumes. 70, 73, 74, 84, 92, 93, 108, 116, 121, 132, 150, 162, and 163. Assays for neural cell biological activities are described, e.g., in Wouterlood (ed. 1995) Neuroscience Protocols modules 10, Elsevier; Methods in Neurosciences Academic Press; and Neuromethods Humana Press, Totowa, NJ. Methodology of developmental systems is described, e.g., in Meisami (ed.) Handbook of Human Growth and Developmental Biology CRC Press; and Chrispeels (ed.) Molecular Techniques and Approaches in Developmental Biology Interscience. FACS analyses are described in Melamed, et al. (1990) Flow Cytometry and Sorting Wiley-Liss, Inc., New York, NY; Shapiro (1988) Practical Flow Cytometry Liss, New York, NY; and Robinson, et al. (1993) Handbook of Flow Cytometry Methods Wiley-Liss, New York, NY.
Example 1:
Cell Culture: THP-1 cells were obtained from ATCC. Cells were cultured in RPMI-1640 medium supplemented with 0.05 mM b-mercaptoethanol and 10% FBS. For all experiments, cells were plated at a density of 500,000 cells/ml in 24 well plates.
Cytokine measurements: THP-1 cells were treated with 10 nM PMA for 24 hours. Appropriate agents were added to the cultures and then incubated for an additional 24 hours. Culture supernatants were collected for measurement of IL-6 and IL-8 using ELISA kits from R&D systems. To determine the concentration of other cytokines, supernatants were analyzed with the LINCO 10-plex cytokine kit, using luminex technology.
Factor Xa assay: Factor Xa assay was performed using soluble tissue factor or PMA treated THP-1 cells as a source of TF. For assays using THP-1 cells, the cells were first freated with 10 nM PMA for 24 hours before the addition of FVIIa inhibitors. The cells were scraped in assay buffer containing 0.03M Tris and 0.15M NaCL, pH 7.4. Equal volume of FX and FVIIa (Enzyme Research Labs) cocktail prepared fresh in assay buffer containing 0.1% HSA, was added to the cell suspension to achieve a final concentration of 60nM of FX and FVIIA. The plate was incubated at 37°C for 15 minutes before addition of FXa substrate MW 2222 prepared in buffer containing 0.06M Tris, 0.3M NaCl, 0.024M EDTA, pH 7.4. Optical density was determined at 405 nm using a Gilford spectrophotometer. Mono S anion exchange FPLC chromatography: FPLC column was equilibrated with 0.05 M Tris-HCL buffer, pH 7.4. FVIIa (25 ul) was applied to the column and then eluted with a linear gradient of 0. IM to IM NaCl prepared in the column buffer. The grdient volume was 15 ml. The effluent was monitored for absorption at 280 nm using an online detector. One ml fractions were collected.
Amino acids sequencing: Protein from SDS-gels were extracted in water and subjected to amino acid sequencing using automatic amino acid sequencer.
Cloning and expression of FVII and truncated FVII: Genes encoding FVII and truncated FVII were amplified by utilizing the polymerase chain reaction (PCR) from FVII cDNA obtained from Origene (Cat # 006137). The oligonucleotide primers,5'- gaggcgcgccgccaccatggccaacgcgttcctggag-3' (HFP-1), 5'- atgcggccgctgggctagggaaatggggc-3' (HFP-2), 5'-gaggctagccgccaccatggtctcccaggccctcagg- 3' (HFP-3), 5'-gaaatccagaacagcttcgtcctctccg-3' (HFP-4) were designed based on the published human cDNA sequences (NCBI accession #s NM_019616.1 and JO 2933.1). It should be noted that an additional sequence corresponding to Ascl, Nhel , Xmnl and Notl were added at the 5 '-end of these primers for cloning purposes. Two restriction fragments Nhel/Xmnl fragment corresponding to amino acid residues -1 to 40 and Xmnl /Notl fragment corresponding to amino acid residues 40 to residue 406 were generated. The sequences were assembled into pCDNA 3.1 (hygro) vector between Nhel and Notl sites to form an expression vector, pRB12-137 that contained a full length cDNA encoding signal peptide, and FVII mature protein. The vector pRB12-137A was then converted into vector pRB12-142A containing cDNA sequence that encoded the truncated form of F VII plus 38 amino acid residues of signal peptide by using site-directed loop out mutagenesis with oligonuceloti.de primers, 5'-gcaccggcgccggcgcctgttctggatttc-3' (HFP-5) and 5'-gaaatccagaacaggcgccggcgccggtgc-3' (HFP-6). After confirming the sequences, both expression vectors were stably transfected into HEK-293 cells by using FuGene 6 transfection reagent (Roche, Cat # 1814443). Transfected cells were selected for hygromycin B resistance (125 ugm mL), individual colonies isolated and screened for high level expression in serum free medium. Western blot analysis: FVII, FVIIa purchased from either ERL or American Diagnostic were subjected to non-reducing SDS-PAGE. The proteins were transferred to PVDF membrane and immunoblotted using FVIIa antibody (American Diagnostica). PVDF membranes were developed using ECL detection system (Amersham).
Results:
Induction of cytokines in human macrophage THP-1 cells by FVIIa: Figure 1 A shows that FVIIa isolated from human plasma produced a concentration-dependent stimulation of IL-6 production in THP-1 cells. The pattern of cytokine induction by FVIIa was selective. IL-6 and IL-8 were the major cytokines produced in response to FVIIa. A slight increase in IL1 D, IL-10 and TNF D over background was also observed. The levels of other cytokines were not detectable (Figure IB), induction of cytokines by FVIIa was completely dependent on the expression of TF in THP-1 cells. Figure IC inset shows that THP-1 cells normally do not express detectable levels of TF. Stimulation with PMA induced TF expression in a time-dependent manner. Parallel measurements of IL-6 expression in these cultures showed that addition of FVIIa or PMA alone did not induce IL-6 expression. FVIIa only induced IL-6 expression in cells expressing TF following the treatment with PMA. These data show that FVIIa induces IL-6 and IL-8 expression in THP-1 cells in a TF-dependent manner.
Cytokine induction by FVIIa does not require thrombin or factor Xa: To examine if IL-6 production in response to FVIIa involves thrombin, we determined the effect of thrombin inhibitor hirudin on IL-6 production. Addition of hirudin did not affect IL-6 production in either the control or FVIIa treated cells (Figure 2). Similarly, addition of factor Xa to control or the FVIIa freated cells did not enhance IL-6 expression. In this study, a slight decrease in IL-6 production was observed in FXa treated cultures. These data suggest that neither thrombin nor FXa were required for IL-6 induction by FVIIa.
Relationship between coagulation and cytokine inducing activity of FVIIa: During the studies reported herein, we found that FX activation activity (conversion of factor X to activated factor Xa by TF-VIIa complex) and IL-6 induction by various preparations of FVIIa did not go hand in hand. Figure 3 A shows the factor X to factor Xa converting activity of three different preparations of FVIIa. Preparation 1, a recombinant FVIIa produced using mammalian cell expression system showed the expected factor X activating activity. Preparation 2, a freshly purified FVIIa from human plasma was slightly more active than the recombinant preparation. Preparation 3 was also plasma derived FVIIa but had been stored at -20C for several months. The stored material demonstrated low activity for FX to FXa conversion (figure 3A). IL-6 inducing activity of these preparations is shown in figure 3B. Preparations 1 and 2 that exhibited good factor X activation showed little or no induction of IL-6 whereas preparation 3 that exhibited very low FX activation showed the highest induction of IL-6. These results indicated that factor X activation and cytokine induction by FVIIa were dissociable activities.
Identification of a LPFVII: To investigate the characteristics of the FVIIa preparations that may explain their different biological activities, we analyzed these preparations by SDS-gel electrophoresis and western blotting. Figure 4A shows that the recombinant FVIIa mainly contained a 52 kD protein band corresponding to FVIIa. Preparations 2 contained FVIIa as a major protein band but also contained a lower molecular weight band corresponding to 46 kD. The major protein in preparation 3 was the 46 kD band. Both bands cross-reacted with FVIIa antibodies in the western blots. The two major bands corresponding 52 kD and 46 kD were eluted from the gel and subjected to amino acid sequencing. The 52 kD protein band exhibited two N-terminals corresponding to the two chains of FVIIa. The N-terminus (ANAFLEELRPGS--) corresponded to the N- terminal sequence of the light chain of FVIIa. The second N-terminal (IVGGKVCPK — ) matched with the N-terminal sequence of the protease domain. The 46 kD protein band also contained two peptides. One N-terminal matched with the N-terminus of the protease domain (IVGGKVCPK—) whereas the second N-terminal (LFWISYSDG— ) overlapped with a sequence within FVIIa starting at residue Leu39. These results showed that the 46 kD protein (LPFVII) was generated as a result of truncation of the first 38 amino acid residues of the native FVIIa (figure 4D). The specific activity for IL-8 production for preparation 3, which consisted of > 90% truncated form, was >40 fold greater (see figure 3B) than the recombinant FVIIa which did not contain the truncated FVIIa. These data show that the truncated molecule was significantly more potent than the native FVIIa.
Activities of native and N-terminal truncated FVIIa: Preparation 2 was fractionated using a FPLC monoS anion exchange column chromatography. The FPLC fractions were analyzed for the presence of FVIIa by western blotting. The factor X converting activity and IL-8 inducing activity of each fraction was also determined. Figure 5 A shows that the 46 kD protein band was present in the early fractions (5-9) whereas fractions 10-17 were enriched in the 52 kD FVIIa band. The 46 kD band corresponding to the N-terminal truncated FVIIa stimulated IL-8 expression but demonstrated no factor X activation activity. Factor X activation activity was co-eluted with the protein corresponding to the 52 kD FVIIa. These data further support that the N-terminal truncated FVIIa lacked FX activation activity but stimulated IL-8 expression. To further demonstrate the coagulation and cytokine inducing activities of FVIIa and N-terminal truncated FVIIa, we cloned the genes encoding the two proteins. It should be noted that the proteins are expressed as FVII and truncated-FVII. The expressed FVII can be activated to FVIIa by a single cleavage by FXa at amino acid residues Rl 52-1153 (Radcliffe, R., andNemerson, Y. (1976) J. Biol. Chem. 251, 4797- 4802). We analyzed the recombinant proteins for FX activation and IL-8 inducing activity before after activation with FXa. As shown in figure 6, the recombinant truncated FVII did not produce FX activation. Also, the truncated FVII or the native FVII did not stimulate IL-8 expression unless activated by treatment with FXa. Following treatment with Xa, FVII was activated to FVIIa and effectively stimulated IL-8 expression. The truncated FVII also induced IL-8 expression after activation with FXa. These data show that the recombinant truncated FVIIa lacking factor X activation activity stimulated IL-8 expression. These data also shows that conversion of FVII to FVIIa or truncated FVIIa was required for induction of IL-8.
Role of protease activity in IL-8 induction: To examine the role of protease activity of FVIIa, we determined the effect active site inhibitors of FVIIa on IL-8 expression.
Treatment with chloromethylketone, an active site inhibitor that covalently cross links with the active site "serine", produced a concentration-dependent inhibition of IL-8 expression, resulting in >80% inhibition at 3uM (figure 7). We also tested the effect of reversible active site inhibitors (2081054 and 2094607) on IL-8 expression. To confirm that the compounds inhibited TF-FVIIa protease activity under the conditions used for the determination of their effect on IL-8 expression, THP-1 cells expressing TF were treated with the indicated concentrations of compounds and TF-VIIa protease activity was then determined. Figure 7 shows that 2081054 and 2094607 produced >95% inhibition of protease activity of TF-FVIIa complex in THP-1 cells. The reversible inhibitors only produces <20% inhibition of IL-8 expression which was not concentration dependent. These results suggest that IL-8 expression in response to FVIIa may not require its protease activity.
Discussion: The experiments described herein have shown that in human macrophage THP-1 cells, FVIIa induced the expression of a select set of cytokine genes. Among the various cytokines assayed, IL-6 and IL-8 were the major activities induced by FVIIa. Cytokine induction by FVIIa required the expression of TF. Following treatment with PMA, the induction of IL-6 by FVIIa coincided with the expression of TF activity in the cells. Other studies have shown that expression of VEGF gene by FVIIa required thrombin or FXa (OUivier, et al, 2000 Thromb. Vase. Biol. 20, 1374-1381). Applicant's data show that in THP-1 cells neither thrombin nor FXa was necessary for cytokine induction by FVIIa. Measurement of FX to FXa converting activity and cytokine inducing activity of various FVIIa preparations showed that the coagulation activity was dissociable from the cytokine inducing activity of FVIIa. By these experiments Applicants have identified a N-terminal truncated form of
FVIIa (LPFVII) that was highly potent inducer of cytokine expression in THP-1 cells. The truncated FVIIa was produced by removal of the first 38 amino acid residues from the protein by proteolytic cleavage. The "Gla" domain resides in the N-terminus of FVIIa, removal of the first 38 amino acid residues rendered the molecule devoid of FX binding ability. This result was consistent with Applicant' s demonstration of a lack of FX to FXa converting activity of LPFVII. A lack of coagulation activity and maintenance of cytokine inducing activity in LP FVIIa was further confirmed by cloning and expression of LP FVIIa in mammalian cells. Recombinant proteins were expressed and secreted as native FVII and a truncated FVII (LPFVII). As has been shown before, FVIIa can be generated from FVII by a single proteolytic cleavage by FXa or FXIIa at residues Rl 52-1153 (Radcliffe, R., and Nemerson, Y. (1976) J. Biol. Chem. 251, 4797- 4802). Assessment of the activities of the recombinant FVII and truncated FVII before and after FXa treatment demonstrated that the truncated FVIIa lacks FX activation activity but unexpectedly maintained stimulation of IL-8 expression in THP-1 cells. These data clearly demonstrate unexpectedly that the coagulation activity and the cytokine inducing activity of TF- Vila are dissociable. Not being bound by theory, our investigation of the role of proteolytic active site of FVIIa in cytokine expression showed that treatment with chloromethylketone, an irreversible active site inhibitor, reduced the cytokine inducing activity of FVIIa. However, a reversible inhibitor that effectively inhibited the proteolytic activity of FVIIa, did not inhibit IL-8 expression in THP-1 cells. The reason for the differences in the activity of the two different types of FVIIa inhibitors is not understood at this time. Our data show that inactivation of FVIIa required stochiometricallly 50 molar excess of chloromethylketone and an extended incubation period. It is possible that inhibition by chloromethylketone is not solely due to a selective cross-linking with the active site "serine". Chloromethylketone could potentially react with other serine residues in the protein, rendering a confirmationally different FVIIa. Thus, a lack of inhibition of IL-8 expression by the reversible active site inhibitors may indicate that the active site may not be essential for cellular signaling by VIIa-TF. Alternatively, the proteolytic activity may not be required for signaling but participates in the generation of the truncated FVIIa from FVIIa. The conversion of FVIIa to truncated FVIIa may occur following binding to TF on cell surface. The truncated FVIIa may then acts as a signaling ligand. Our findings may have important implications in the role of FVIIa and the truncated FVIIa in inflammation and angiogenesis. FVIIa-TF pathway has been shown to induce several genes that mediate angiogenesis. For example, FVIIa induces IL-8 expression in macrophage and fibroblasts (Camerer, et al., 2000 J. Biol. Chem. 275, 6580-6585). IL-8 is a chemoatfractant for endothelial cells and monocyte, and promotes angiogenesis in vivo (Radcliffe, R., and Nemerson, Y. (1976) J. Biol. Chem. 251, 4797- 4802; Szekanecz, et al., 1994 Pathology 62, 134-139). FVIIa also induces the expression of the CCN gene family members CYR61 and connective tissue growth factor (CTGF) (Camerer, et al., 2000 J. Biol. Chem. 275, 6580-6585). The cystein rich protein CYR61 acts as a ligand for the integrin alpha v beta 3 (22). CYR61 stimulates endothelial cell growth and migration, and in vivo angiogenesis (Kireeva, et al., 1996 Mol Cell Biol 16, 1326-1334; Babic, et al. 1998 Proc. Natl. Acad. Sci. U.S.A. 95, 6355-6360). CYR 61 gene knock out mice show gross vascular defects (Mo, et al., 2002 Mol. Cell. Biol. 22, 8709-8720). Other FVIIa induced angiogenic genes include VEGF and FGF-5 (OUivier, et al., 2000 Thromb. Vase. Biol. 20, 1374-1381; Camerer, et al., 2000 J. Biol. Chem. 275, 6580-6585). Although FVIIa may induce angiognesis, TF-VIIa activation in vivo is usually associated with the induction of coagulation and platelet activation. Since the truncated FVIIa is devoid of the coagulation activity but induces proangiogenesis genes, this molecule may be uniquely suited for angiogenic therapy. Tissue factor is expressed at high level in atherosclerotic plaque (Wilcox, et al., 1989 Proc. Natl. Acad. Sci. U.S.A. 86, 2839-2843; Marmur, et al., 1996 Circ. 94, 1226- 1232; and Mallat, et al., 2000 Circ. 101, 841-843) and following vascular injury (Jang,et al., 1995 Circ. 92, 3041-3050). TF-VIIa pathway has been implicated in the pathogenesis of vascular inflammation and plaque rupture. Generation of truncated FVIIa in the atherosclerotic plaque may provide a potent stimulus for the induction and maintenance of vascular inflammation.
SEQUENCE LISTING
SEQ ID NO: 1 is primate LP FVIIa nucleic acid sequence. SEQ ID NO: 2 is primate LP FVIIa amino acid sequence.

Claims

WHAT IS CLAIMED IS:
1. An isolated and/or recombinant polynucleotide comprising sequence encoding an antigenic polypeptide comprising at least: 17, 20, 25, 30, 35, 40, 45, 50, 55, 60, 65, 70, 75, 80, 85, 90, 95, 100, 110, 120, 130, 140, 150, 160, 170, 180, 190, 200, 210, 220, 230, 240, 250, 260, 270, 280, 290, 300, 310, 320, 330, 340, 350, 360, 370, 380, 390, 400, or 405 contiguous amino acids from a mature portion of an LP of Table 1.
2. The polynucleotide of Claim 1, encoding: a) a full length polypeptide of Table 1; b) a mature polypeptide of Table 1; c) an antigenic fragment at least: 12 17, 20, 25, 30, 35, 40, 45, 50, 55, 60, 65, 70, 75, 80, 85, 90, 95, 100, 110, 120, 130, 140, 150, 160, 170, 180, 190, 200, 210, 220, 230, 240, 250, 260, 270, 280, 290, 300, 310, 320, 330, 340, 350, 360, 370, 380, 390, 400, or 405 contiguous amino acid residues in length of Table 1; d) at least two fragments of an LP sequence of Table 1, wherein said fragments do not overlap; e) a plurality of fragments of an LP sequence of Table 1, wherein said fragments do not overlap; or f) a mature polypeptide of Table 1 having less than five amino acid substitutions.
3. The polynucleotide of Claim 1, which hybridizes at 55°C, less than 500 mM salt, to: a) the coding portion of SEQ ID NO: 1 ; or b) the coding portion of a nucleic acid of Table 1.
4. The polynucleotide of Claim 3, wherein said temperature is at least 65° C, and said salt is less than 300 mM.
5. The polypeptide of Claim 3, comprising at least 35, 40, 45, 50, 55, 60, 65, 70, 75, 80, 85, 90, 95, 100, 110, 120, 130, 140, 150, 160, 170, 180, 190, 200, 210, 220, 230, 240, 250, 260, 270, 280, 290, 300, 310, 320, 330, 340, 350, 360, 370, 380, 390, 400, 410, 420, 430, 440, 450, 460, 470, 480, 490, 500, 510, 520, 530, 540, 550, 560, 570, 580, 590, 600, 610, 620, 630, 640, 650,660, 670, 680, 690, 700, 710, 720, 730, 740, 750, 760, 770, 780, 790, 800, 810, 820, 830, 840, 850, 860, 870, 880, 890, 900, 910, 920, 930, 940, 950, 960, 970, 980, 990,1000, 1010, 1020, 1030, 1040, 1050, 1060, 1070, 1080, 1090, 1100, 1110, 1120, 1130, 1140, 1150, 1160, 1170, 1180, 1190, 1200, 1210, 1220, 1230, 1240 or 1250 contiguous nucleotides to a nucleotide sequence of an LP of Table 1 or: a) the coding portion of SEQ ID NO: 1.
6. An expression vector comprising a polynucleotide of Claim 5, wherein said temperature is at least 65° C, and said salt is less than 300 mM.
7. The expression vector of Claim 6, which further comprises a plurality of nucleotide segments with identity to a coding portion of an LP nucleic acid sequence of Table 1.
8. A host cell containing the expression vector of Claim 6, including an eukaryotic cell.
9. A method of making an antigenic polypeptide comprising expressing a recombinant polynucleotide of Claim 1.
10. A method for detecting a polynucleotide of Claim 1 , comprising contacting said polynucleotide with a probe that hybridizes, under stringent conditions, to at least 25 contiguous nucleotides of: a) the coding portion of a LP nucleic acid of Table 1 or b) the mature coding portion of SEQ ID NO: 1; to form a duplex, wherein detection of said duplex indicates the presence of said polynucleotide.
11. A kit for the detection of a polynucleotide of Claim 1 , comprising a probe that detectably hybridizes, under sfringent hybridization conditions, to at least 34 contiguous nucleotides of a polynucleotide of Claim 1.
12. The kit of claim 11 , wherein said probe is detectably labeled.
13. A binding compound comprising an antibody binding site which specifically and/or selectively binds to at least an 8, 9, 10, 11, 12, 13, 14, 15, 16, or 17 contiguous amino acid segment of: a) primate LP of Table 1.
14. The binding compound of Claim 13 , wherein: a) said antibody binding site is: i) specifically and/or selectively immunoreactive with a polypeptide of Table 1, or ii) raised against a purified or recombinantly produced human LP protein selected from :LPFVII, or Table 1; or iii) in a monoclonal antibody, Fab, or F(ab)2; b) said binding compound is: i) an antibody molecule; ii) a polyclonal antiserum; iii) detectably labeled; iv) sterile; or v) in a buffered composition; or c) said binding compound is produced by: i) immunizing an animal with an antigenic fragment of an LP of Table 1 or Table 2; ii) humanizing an antibody; iii) genetic engineering; iv) via a phage display system; or v) in an artificial system.
15. A method using the binding compound of Claim 13, comprising contacting said binding compound with a biological sample comprising an antigen, thereby forming an LP binding compound:antigen complex.
16. The method of Claim 15, wherein said biological sample is human, and wherein said binding compound is an antibody.
17. A detection kit comprising said binding compound of Claim 14, and: a) instructional material for the use of said binding compound for said detection; or b) a compartment providing segregation of said binding compound.
18. A substantially pure or isolated antigenic polypeptide, which binds to said binding composition of Claim 13, and further comprises at least: 25, 30, 35, 40, 45, 50, 55, 60, 65, 70, 75, 80, 85, 90, 95, 100, 110, 120, 130, 140, 150, 160, 170, 180, 190, 200, 210, 220, 230, 240, 250, 260, 270, 280, 290, 300, 310, 320, 330, 340, 350, 360, 370, 380, 390, 400, or 405 contiguous amino acids from a: a) primate LP FVIIa (SEQ ID NO: 2) or b) primate LP of Table 1.
19. The polypeptide of Claim 18, which: a) comprises at least a fragment of at least 25, 30, 35, 40, 45, 50, 55, 60, 65, 70, 75, 80, 85, 90, 95, 100, 110, 120, 130, 140, 150, 160, 170, 180, 190, 200, 210, 220, 230, 240, 250, 260, 270, 280, 290, 300, 310, 320, 330, 340, 350, 360, 370, 380, 390, 400, or 405 contiguous amino acids from a primate LP protein selected from:LPFVII, or an LP of Table 1; b) is a soluble polypeptide; c) is detectably labeled; d) is in a sterile composition; e) is in a buffered composition; f) is recombinantly produced, or g) has a naturally occurring polypeptide sequence.
20. A method of screening a sample for a binding partner for a polypeptide encoded by a nucleic acid sequence of Claim 1 comprising screening in said sample for a specif :ιύc biinn/dtiinngcr
Figure imgf000158_0001
s σaαiirdl r pvrtγoγti-£e*ϊinn
PCT/US2004/019099 2003-08-05 2004-07-22 Lp mammalian proteins; related reagents WO2005044849A2 (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US49249903P 2003-08-05 2003-08-05
US60/492,499 2003-08-05

Publications (2)

Publication Number Publication Date
WO2005044849A2 true WO2005044849A2 (en) 2005-05-19
WO2005044849A3 WO2005044849A3 (en) 2005-08-04

Family

ID=34572730

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2004/019099 WO2005044849A2 (en) 2003-08-05 2004-07-22 Lp mammalian proteins; related reagents

Country Status (1)

Country Link
WO (1) WO2005044849A2 (en)

Cited By (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US8158584B2 (en) 2008-05-02 2012-04-17 Acceleron Pharma, Inc. Pharmaceutical preparations comprising an ALK1-Fc fusion protein
US8455428B2 (en) 2006-11-02 2013-06-04 Acceleron Pharma, Inc. ALK1 receptor and ligand antagonist and uses thereof
US8642031B2 (en) 2006-11-02 2014-02-04 Acceleron Pharma, Inc. Antagonists of BMP9, BMP10, ALK1 and other ALK1 ligands, and uses thereof
US10059756B2 (en) 2006-11-02 2018-08-28 Acceleron Pharma Inc. Compositions comprising ALK1-ECD protein

Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP0200421A2 (en) * 1985-04-17 1986-11-05 Zymogenetics, Inc. Expression of factor VII activity in mammalian cells

Patent Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP0200421A2 (en) * 1985-04-17 1986-11-05 Zymogenetics, Inc. Expression of factor VII activity in mammalian cells

Non-Patent Citations (2)

* Cited by examiner, † Cited by third party
Title
NICOLAISEN E M: "GENERATION OF GLA-DOMAINLESS FVIIA BY CATHEPSIN G-MEDIATED CLEAVAGE" FEBS LETTERS, ELSEVIER SCIENCE PUBLISHERS, AMSTERDAM, NL, vol. 306, no. 2 / 3, 20 July 1992 (1992-07-20), pages 157-160, XP000286918 ISSN: 0014-5793 *
PERSSON E ET PETERSON L C: "Structurally and functionally distinct Ca2+ binding sites in the gamma-carboxyglutamic acid-containing domain of factor VIIa" EUROPEAN JOURNAL OF BIOCHEMISTRY, BERLIN, DE, vol. 234, no. 1, 15 November 1995 (1995-11-15), pages 293-300, XP002101115 ISSN: 0014-2956 *

Cited By (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US8455428B2 (en) 2006-11-02 2013-06-04 Acceleron Pharma, Inc. ALK1 receptor and ligand antagonist and uses thereof
US8642031B2 (en) 2006-11-02 2014-02-04 Acceleron Pharma, Inc. Antagonists of BMP9, BMP10, ALK1 and other ALK1 ligands, and uses thereof
US9452197B2 (en) 2006-11-02 2016-09-27 Acceleron Pharma, Inc. Antagonists of BMP9, BMP10, ALK1 and other ALK1 ligands, and uses thereof
US10059756B2 (en) 2006-11-02 2018-08-28 Acceleron Pharma Inc. Compositions comprising ALK1-ECD protein
US8158584B2 (en) 2008-05-02 2012-04-17 Acceleron Pharma, Inc. Pharmaceutical preparations comprising an ALK1-Fc fusion protein

Also Published As

Publication number Publication date
WO2005044849A3 (en) 2005-08-04

Similar Documents

Publication Publication Date Title
US20080146505A1 (en) 47 Human Secreted Proteins
EP1233983A2 (en) Kunitz-type protease inhibitor polynucleotides, polypeptides, and antibodies
CA2398255A1 (en) Bcl-2-like polynucleotides, polypeptides, and antibodies
CA2368068A1 (en) 27 human secreted proteins
WO2001024815A1 (en) Plasminogen-like polynucleotides, polypeptides, and antibodies
CA2373638A1 (en) Adam polynucleotides and polypeptides
WO2001012786A1 (en) Retinoid receptor interacting polynucleotides, polypeptides, and antibodies
US20070190612A1 (en) 31 Human Secreted Proteins
CA2381451A1 (en) Attractin-like polynucleotides, polypeptides, and antibodies
WO2001008635A2 (en) Calcium channel transport polynucleotides, polypeptides, and antibodies
CA2392740A1 (en) Il-6 like polynucleotides, polypeptides, and antibodies
CA2373759A1 (en) Serine proteases
CA2365917A1 (en) Bone morphogenic proteins
WO2005044849A2 (en) Lp mammalian proteins; related reagents
WO2000061614A2 (en) Uncoupling proteins
US20030050466A1 (en) TM4SF polynucleotides, polypeptides, and antibodies
CA2392693A1 (en) Four disulfide core domain-containing (fdcd) polynucleotides, polypeptides, and antibodies
CA2384083A1 (en) Human neuropeptide receptor
EP1248801A1 (en) Human polynucleotides, polypeptides, and antibodies
EP1235837A1 (en) Kringle domain-containing polynucleotides, polypeptides, and antibodies
WO2001037857A1 (en) Major intrinsic protein (mip)-like polynucleotides, polypeptides, and antibodies
WO2001018049A1 (en) Steroid hormone receptor polynucleotides, polypeptides, and antibodies
CA2370523A1 (en) 49 human secreted proteins
CA2363716A1 (en) 49 human secreted proteins

Legal Events

Date Code Title Description
AK Designated states

Kind code of ref document: A2

Designated state(s): AE AG AL AM AT AU AZ BA BB BG BR BW BY BZ CA CH CN CO CR CU CZ DE DK DM DZ EC EE EG ES FI GB GD GE GH GM HR HU ID IL IN IS JP KE KG KP KR KZ LC LK LR LS LT LU LV MA MD MG MK MN MW MX MZ NA NI NO NZ OM PG PH PL PT RO RU SC SD SE SG SK SL SY TJ TM TN TR TT TZ UA UG US UZ VC VN YU ZA ZM ZW

AL Designated countries for regional patents

Kind code of ref document: A2

Designated state(s): BW GH GM KE LS MW MZ NA SD SL SZ TZ UG ZM ZW AM AZ BY KG KZ MD RU TJ TM AT BE BG CH CY CZ DE DK EE ES FI FR GB GR HU IE IT LU MC NL PL PT RO SE SI SK TR BF BJ CF CG CI CM GA GN GQ GW ML MR NE SN TD TG

121 Ep: the epo has been informed by wipo that ep was designated in this application
122 Ep: pct application non-entry in european phase