NZ278504A - Human vascular ibp-like growth factor polypeptide (vigf) - Google Patents

Human vascular ibp-like growth factor polypeptide (vigf)

Info

Publication number
NZ278504A
NZ278504A NZ278504A NZ27850494A NZ278504A NZ 278504 A NZ278504 A NZ 278504A NZ 278504 A NZ278504 A NZ 278504A NZ 27850494 A NZ27850494 A NZ 27850494A NZ 278504 A NZ278504 A NZ 278504A
Authority
NZ
New Zealand
Prior art keywords
polypeptide
polynucleotide
vigf
isolated
accompanying drawings
Prior art date
Application number
NZ278504A
Inventor
Gregg A Hastings
Craig A Rosen
Original Assignee
Human Genome Sciences Inc
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Human Genome Sciences Inc filed Critical Human Genome Sciences Inc
Priority to NZ278504A priority Critical patent/NZ278504A/en
Publication of NZ278504A publication Critical patent/NZ278504A/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/475Growth factors; Growth regulators
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P3/00Drugs for disorders of the metabolism
    • A61P3/06Antihyperlipidemics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • 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
    • C12N2799/00Uses of viruses
    • C12N2799/02Uses of viruses as vector
    • C12N2799/021Uses of viruses as vector for the expression of a heterologous nucleic acid
    • C12N2799/026Uses of viruses as vector for the expression of a heterologous nucleic acid where the vector is derived from a baculovirus

Landscapes

  • Health & Medical Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • General Health & Medical Sciences (AREA)
  • Animal Behavior & Ethology (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Public Health (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Veterinary Medicine (AREA)
  • General Chemical & Material Sciences (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Engineering & Computer Science (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Zoology (AREA)
  • Genetics & Genomics (AREA)
  • Biophysics (AREA)
  • Biochemistry (AREA)
  • Gastroenterology & Hepatology (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Toxicology (AREA)
  • Molecular Biology (AREA)
  • Diabetes (AREA)
  • Hematology (AREA)
  • Obesity (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)
  • Peptides Or Proteins (AREA)

Description

New Zealand No International No 278504 TO BE ENTERED AFTER ACCEPTANCE AND PUBLICATION Priority dates 09 12 1994 Complete Specification Filed 09 12 1994 Classification (6) C12N15/18, C07K14/65, A61K38/30, C12Q1/68 Publication date 28 October 1999 Journal No 1445 NEW ZEALAND PATENTS ACT 1953 COMPLETE SPECIFICATION Title of Invention Human vascular ibp-like growth factor Name, address and nationality of applicant(s) as in international application form HUMAN GENOME SCIENCES, INC , a Delaware corporation of 9410 Key West Avenue, Rockville, Maryland 20850-3338, United States of America HUMAN VASCULAR IBP-LIKE GROWTH FACTOR, This invention relates to newly identified polynucleotides, polypeptides encoded by such polynucleotides, the use of such polynucleotides and polypeptides, as well as the production of such polynucleotides and polypeptides The invention also relates to inhibiting the action of such polypeptides The polypeptide of the present invention is related to a family of growth regulators comprising cef 10/cyr 61, connective tissue growth factor (CTGF), and nov, as well as the insulin-like growth factor binding protein (IBP) family which modulates the activity of insulin-like growth factor (IGF) The mRNA corresponding to the polypeptide of this invention is highly expressed m vascular cell-types, thus, this polypeptide is hereinafter referred to as human vascular IBP-like growth factor or "VIGF" Growth factors and other mitogens, including transforming oncogenes, are capable of rapidly inducing a complex set of genes to be expressed by certain cells (Lau, L F. and Nathans, D., Molecular Aspects of Cellular Regulation. 6.165-202 (1991) These genes, which have been named immediate early or early response genes, are transcriptionally activated within minutes after contact with a growth factor or mitogen, independent of de novo protein 08 36 10 synthesis. A group of these immediate early geneB encodes secreted, extracellular proteins which are needed for coordination of complex biological processes such as differentiation and proliferation, regeneration and wound healing (Rvseck, R P et al, Cell Growth Differ . 2 235-233 (1991).
Highly related proteins which belong to this group include cef 10 from chicken, which was detected after induction by the viral oncogene pp60,"t (Simmons, D L. et al, PNAS. USA, 86 117B-1182 (1989) A closely related protein, cyr 61, is rapidly activated by serum or platelet-derived growth factor (PDGF) (O'Brien, TP et al, Mol. Cell Biol , 10.3569-3577 (1990) The overall amin6 acid identity between cef 10 and cyr 61 is as high as 63% A third member is human connective tissue growth factor (CTGF) (Bradham, D M. et al , J Cell Biol . 114:1285-1294 (1991) CTGF is a cysteine-rich peptide which is secreted by human vascular endothelial cells in high levels after activation with transforming growth factor beta (TGF-3) CTGF exhibits PDGF— like biological and immunological activities and competes with PDGF for a particular cell surface receptor A fourth member of the immediate-early proteins is fzsp-12, which has been shown to be induced by serum and has been mapped to a region of the murine genome (Ryseck, R.P et al , Cell Growth Differ . 2 235-233 (1991) Yet another member of this family is the chicken gene, nov, normally arrested in adult kidney cells, which was found to be overexpressed in myeloblastosis-associated virus type 1 induced nephroblastomas Further, expression of an ammo-terminal-truncated nov product in chicken embryo fibroblasts was sufficient to induce transformation (Joliot, V et al , Mol Cell Biol . 12 10-21 (1992) The expression of these immediate early genes act as "third messengers" m the cascade of events triggered by growth factors. It is also thought that they are needed to 08 36 10 integrate and coordinate complex biological processes, such as differentiation and wound healing m which cell proliferation is a common event This emerging family of growth regulators is called the CCN family for CTGF; cef 10/cyr 61, and nov The VIGF polypeptide of the present invention is thought to be a member of this family of growth regulators The VIGF polypeptide also contains a stretch of cysteines which is highly homologous to insulin-like growth factor (IGF) -binding protein.
At least two different binding proteins have been identified in adult human serum, namely, IGF-bmdmg protein 5 3 and IGF-bmdmg protein 1 The IGF-bmdmg proteins have both stimulatory and inhibitory effects on IGF. Clemmons, et al, J. Clin Invest , 77:1548 (1986) showed increased binding to fibroblast and smooth muscle cell surface receptors of IGF m complex with its binding protein The inhibitory effects of IGF-bmdmg protein on various IGF actions in vitro, have been shown and they include stimulation of glucose transport by adipocytes, sulfate incorporation by chondrocytes and thymidine incorporation m fibroblast (Zapf, et al., J. Clin. Invest., 63-1077 (1979)). In addition, inhibitory effects of IGF-bmdmg proteins on growth factor mediated mitogen activity m normal cells has been shown In accordance with one aspect of the present invention, there is provided a novel mature polypeptide which is VIGF, as well as biologically active and diagnostically or therapeutically useful fragments, analogs and derivatives thereof.
This invention provides for an isolated polynucleotide selected from the group consisting of (a) a polynucleotide encoding tne VIGF polypeptide having the deduced amino acid sequence of Figure 1 or a fragment, analog or derivative of said polypeptide, 3 (b) a polynucleotide encoding the VIGF polypeptide having the ammo acid sequence encoded by the cDNA contained in ATCC Deposit No 75874 or a fragment, analog or derivative of said polypeptide.
Also provided are isolated nucleic acid molecules encoding human VIGF, including mRNAs, DNAs, cDNAs, genomic DNAs as well as analogs and biologically active and diagnostically or therapeutically useful fragments and derivatives thereof Described but not claimed is a process for producing such a polypeptide by recombinant techniques comprising culturing recombinant prokaryotic and/or eukaryotic host cells, containing a human VIGF nucleic acid sequence, under conditions promoting expression of said protein and subsequent recovery of said protein Accordingly, a further aspect of the invention provides a vector containing a polynucleotide of the invention A yet further aspect of the present invention provides a host cell transfected with the vector of tne invention A still further aspect of the present invention provides an isolated DNA molecule hybndizable to a polynucleotide of the invention and encoding a polypeptide having VIGF activity Another aspect of the present invention provides a pharmaceutical composition comprising a polynucleotide of the invention and a pharmaceutically acceptable carrier, excipient or diluent The reader's attention is directed to our related New Zealand Patent Specification No. 337632 which describes and claims a process of utilizing such polypeptide, or polynucleotide encoding such polypeptide for therapeutic purposes, for example, to treat inuscle wasting diseases, osteoporosis, to aid m implant fixation, to stimulate wound healing or tissue regeneration, to promote angiogenesis and to proliferate vascular smooth muscle and endothelial cell production A further aspect of the present invention provides antibodies against polypeptides of the invention Other antagonists to such polypeptides are also described but not claimed, which may be used to inhibit the action of such polypeptides, for example, to limit the production of excess connective tissue during wound healing or pulmonary fibrosis Further, nucleic acid probes are described but not claimed which comprise nucleic acid molecules of sufficient length to specifically hybridize to VIGF sequences Also described are diagnostic assays for detecting diseases related to the under-expression and over-expression of the VIGF polypeptide and mutations m the nucleic acid sequences encoding such polypeptide Accordingly, a further aspect of the present invention provides a process for diagnosing a disease or susceptibility to a disease related to a mutation m VIGF nucleic acid sequence comprising isolating a nucleic acid sequence encoding VIGF from a sample derived from a host and determining a mutation m said VIGF nucleic acid sequence with reference to a polynucleotide of the invention A yet further aspect of the invention provides a diagnostic process comprising analysing for the presence of a polynucleotide of the invention m a sample derived from a host (followed by page 4a) 4 Another aspect of the present invention provides an isolated polypeptide selected from the group consisting of (a) a polypeptide comprising amino acids 22-184 of SEQ ID NO 2, (b) a polypeptide comprising ammo acids 1-184 of SEQ ID NO 2, (c) a polypeptide comprising ammo acids 51-54 of SEQ ID NO 2, (d) a polypeptide comprising ammo acids 76-90 of SEQ ID NO 2, (e) a polypeptide fragment of SEQ ID NO 2, wherein said fragment has the ability to stimulate the proliferation of endothelial cells m the presence of comitogen Con A, (f) a polypeptide variant of (a), (b), (c), (d) or (e) resulting from conservative substitutions, and (g) an antigenic polypeptide fragment of the polypeptide of (a) , (b) , (c), (d) , (e) or (f) Further aspects of the invention provide pharmaceutical compositions, methods of diagnosis and methods of identifying binding partners, all as set forth m the accompanying claim set These and other aspects of the present invention should be apparent to those skilled m the art from the teachings herein (followed by page 5] lATY n 7 " 4a - / 0n°'^ " 8 Sep 1999 .?FrF,,/F, PCT/US94/143S8 The following drawings are illustrative of embodiments of the invention and are not meant to limit the scope of the invention as encompassed by the claimB.
Figure 1 shows the cDNA and corresponding deduced amino acid sequence of the VIGF polypeptide The initial 21 amino acids represent the putative leader sequence such that the mature polypeptide comprises 163 ammo acids. The standard one letter abbreviations for amino acids are uBed Sequencing was performed using a 373 Automated DNA sequencer (Applied Biosystems, Inc ) Seqeuncing accuracy is predicted to be greater than 97% accurate Figure 2 shows the amino acid sequence homology between VIGF and other proteins which are members of the CCN family.
Figure 3 shows an SDS-polyacrylamide gel which displays the results of VIGF bacterial purification and electrophoresis Figure 4 shows a gel which displays the results of a Northern Blot analysis performed on VIGF Figure 5 shows a gel which displays the results of a cell-type analysis of VIGF gene expression in the various tissues displayed Lane 1 is umbilical vein endothelial cells, Lane 2 is aortic smooth muscle cells and Lane 3 is dermal foreskin fibroblast cells Figure 5A shows the results after a two hour exposure and Figure 5B shows the results after a thirty-six hour exposure In accordance with an aspect of the present invention, there is provided an isolated nucleic acid (polynucleotide) which encodes for the mature polypeptide having the deduced amino acid sequence of Figure 1 or for the mature polypeptide encoded by the cDNA of the clone deposited as ATCC Deposit No 75874 on August 25, 1994 A polynucleotide encoding a polypeptide of the present invention may be obtained from human umbilical vein and aortic endothelial cells, aortic smooth muscle cells, and pulmonary artery The polynucleotide of this invention was —5— 08 36 10 discovered in a cDNA library derived from human umbilical vein endothelial cellB It is structurally related to the IBP and CCN families It contains an open reading frame encoding a protein of 184 amino acid residues of which approximately the first 21 amino acids residues are the putative leader sequence such that the mature protein comprises 163 amino acids The designation of VIGF as a hybrid member of both the CCN growth factor and IBP families was based primarily through conservation of amino acid sequences Similarity of VIGF to the CCN family 16 inferred because of the 40-45% similarity over the entire polypeptide, 12 of a total of IB VIGF cysteines are conserved, and 94% identity with the IBP signature (GCGCCXXCAXXXXXXC) which is perfectly conserved in every member of the CCN family The VIGF polypeptide also has significant similarity to the IBP family In two adjacent regions, ammo acids 30-44 (IBP signature) and 55-69, there is at least 80% identity to the IBP family. These regions are contained within the putative IGF binding domain of the IBPs The human tissue and cell-type specific expression has been determined by Northern blot analysis The 2 3-2 4 kb VIGF mRNA is localized m the adult lung and kidney as shown using the procedure of Example 4 VIGF gene expression was undetectable m heart, brain, placenta, liver, skeletal muscle, and pancreas Cultured human umbilical vein endothelial and aortic smooth muscle cells are cell-types which express VIGF mRKA at a high level while dermal foreskin fibroblasts show a very low level Together, these results indicate that VIGF is primarily of vascular origin.
The polynucleotide of the present invention may be in the form of RNA or m the form of DNA, which DNA includes cDNA, genomic DNA, and synthetic DNA. The DNA may be double-stranded or single-stranded, and if single stranded may be the coding strand or non-coding (anti-sense) strand. The 08 36 10 coding Bequence which encodes the mature polypeptide may be identical to the coding sequence shown in Figure 1 or that of the deposited clone or may be a different coding sequence which coding sequence, as a result of the redundancy or degeneracy of the genetic code, encodes the same mature polypeptide as the DNA of Figure 1 or the deposited cDNA The polynucleotide which encodes for the mature polypeptide of Figure 1 or for the mature polypeptide encoded by the deposited cDNA may include only the coding sequence for the mature polypeptide, the coding sequence for the mature polypeptide and additional coding sequence such as a leader or secretory sequence or a proprotem sequence; the coding sequence for the mature polypeptide (and optionally additional coding sequence) and non-coding sequence, such as introns or non-coding sequence 5' and/or 3' of the coding sequence for the mature polypeptide Thus, the term "polynucleotide encoding a polypeptide" encompasses a polynucleotide which includes only coding sequence for the polypeptide as well as a polynucleotide which includes additional coding and/or non-coding sequence The present invention further relates to variants of the hereinabove described polynucleotides which encode for fragments, analogs and derivatives of the polypeptide having the deduced amino acid sequence of Figure 1 or the polypeptide encoded by the cDNA of the deposited clone The variant of the polynucleotide may be a naturally occurring allelic variant of the polynucleotide or a non-naturally occurring variant of the polynucleotide.
Thus, the present invention includes polynucleotides encoding the same mature polypeptide as shown in Figure 1 or the same mature polypeptide encoded by the cDNA of the deposited clone as well as variants of such polynucleotides which variants encode for a fragment, derivative or analog of the polypeptide of Figure 1 or the polypeptide encoded by the cDNA of the deposited clone. Such nucleotide variants 08 36 10 PCIYUS94/143U8 include deletion variants, substitution variants and addition or insertion variants As hereinabove indicated, the polynucleotide may have a coding sequence which as a naturally occurring allelic variant of the coding sequence shown in Figure 1 or of the coding sequence of the deposited clone As known in the art, an allelic variant is an alternate form of a polynucleotide sequence which may have a substitution, deletion or addition of one or more nucleotides, which does not substantially alter the function of the encoded polypeptide The present invent.on also includes polynucleotides, wherein the coding sequence for the mature polypeptide may be fused in the same reading frame to a polynucleotide sequence which aids m expression and secretion of a polypeptide from a host cell, for example, a leader sequence which functions as a secretory sequence for controlling transport of a polypeptide from the cell The polypeptide having a leader sequence is a preprotein and may have the leader sequence cleaved by the host cell to form the mature form of the polypeptide. The polynucleotides may also encode for a proprotem which is the mature protein plus additional 5' a. 10 acid residues A mature protein having a prosequence is a proprotein and is an inactive form of the protein Once the prosequence is cleaved an active mature protein remains Thus, for example, the polynucleotide of the present invention may encode for a mature protein, or for a protein having a prosequence or for a protein having both a prosequence and a presequence (leader sequence) The polynucleotides of the present invention may also have the coding sequence fused in frame to a marker sequence which allows for purification of the polypeptide of the present invention The marker sequence may be a hexa-histidme tag supplied by a pQE-9 vector to provide for purification of the mature polypeptide fused to the marker in the case of a bacterial host, or, for example, the marker 08 36 10 Boquence may be a hemagglutinin (HA) tag when a mammalian host, e.g. COS-7 cells, is used The HA tag corresponds to an epitope derived from the influenza hemagglutinin protein (Wilson, I., et al , Cell, 37-767 (1984)).
The present invention further relates to polynucleotides which hybridize to the heremabove-described sequences if there as at least 50% and preferably 70% identity between the sequences The present invention particularly relates to polynucleotides which hybridize under stringent conditions to the heremabove-descnbed polynucleotides As herein used, the term "stringent conditions" means hybridization will occur only if there is at least 95% and preferably at least 97% identity between the sequences The polynucleotides which hybridize to the hereinabove described polynucleotides in a preferred embodiment encode polypeptides which retain substantially the same biological function or activity as the mature polypeptide encoded by the cDNA of Figure 1 or the deposited cDNA The deposit(s) referred to herein will be maintained under the terms of the Budapest Treaty on the International Recognition of the Deposit of Micro-organisms for purposes of Patent Procedure These deposits are provided merely as convenience to those of skill in the art and are not an admission that a deposit is required under 35 U.S.C §112 The sequence of the polynucleotides contained in the deposited materials, as well as the amino acid sequence of the polypeptides encoded thereby, are incorporated herein by reference and are controlling in the event of any conflict with any description of sequences herein A license may be required to make, use or sell the deposited materials, and no such license is hereby granted The present invention further relates to a VIGF polypeptide which has the deduced amino acid sequence of Figure 1 or which has the ammo acid sequence encoded by the 08 36 10 deposited cDNA, as well as fragments, analogs and derivatives of such polypeptide The terms "fragment," "derivative" and "analog" when referring to the polypeptide of Figure 1 or that encoded by the deposited cDNA, means a polypeptide which retains essentially the same biological function or activity as such polypeptide Thus, an analog includes a proprotein which can be activated by cleavage of the proprotein portion to produce an active mature polypeptide.
The polypeptide of the present invention may be a recombinant polypeptide, a natural polypeptide or a synthetic polypeptide, preferably a recombinant polypeptide The fragment, derivative or analog of the polypeptide of Figure 1 or that encoded by the deposited cDNA may be (i) one in which one or more of the amino acid residues are substituted with a conserved or non-conserved amino acid residue (preferably a conserved amino acid residue) and such substituted ammo acid residue may or may not be one encoded by the genetic code, or (n) one in which one or more of the ammo acid residues includes a substituent group, or (in) one in which the mature polypeptide is fused with another compound, such as a compound to increase the half-life of the polypeptide (for example, polyethylene glycol), or (iv) one m which the additional amino acids are fused to the mature polypeptide, such as a leader or secretory sequence or a sequence which is employed for purification of the mature polypeptide or a proprotein sequence Such fragments, derivatives and analogs are deemed to be within the scope of those skilled in the art from the teachings herein.
The polypeptides and polynucleotides of the present invention are preferably provided m an isolated form, and preferably are purified to homogeneity The term "isolated" means that the material is removed from its original environment (e.g., the natural environment if it is naturally occurring). For example, a naturally- 08 36 10 FCT/DS94/143S8 occurring polynucleotide or polypeptide present in a living animal is not isolated, but the same polynucleotide or polypeptide, separated from some or all of the coexisting materials in the natural system, is isolated. Such polynucleotides could be part of a vector and/or such polynucleotides or polypeptides could be part of a composition, and still be isolated in that such vector or composition is not part of its natural environment.
The present invention also relates to vectors which include polynucleotides of the present invention, host cells which are genetically engineered with vectors of the invention and the production of polypeptides of the invention by recombinant techniques.
Host cells are genetically engineered (transduced or transformed or transfected) with the vectors of this invention which may be, for example, a cloning vector or an expression vector. The vector may be, for example, in the form of a pla6mid, a viral particle, a phage, etc. The engineered host cells can be cultured in conventional nutrient media modified as appropriate for activating promoters, selecting transformants or amplifying the VIGF genes. The culture conditions, such as temperature, pH and the like, are those previously used with the host cell selected for expression, and will be apparent to the ordinarily skilled artisan.
The polynucleotides of the present invention may be employed for producing polypeptides by recombinant techniques Thus, for example, the polynucleotide may be included in any one of a variety of expression vectors ior expressing a polypeptide Such vectors include chromosomal, nonchromosomal and synthetic DNA sequences, e.g., derivatives of SV40; bacterial plasmids, phage DNA; baculovirus; yeast plasmids, vectors derived from combinations of plasmids and phage DNA, viral DNA such as vaccinia, adenovirus, fowl pox virus, and pseudorabies. 08 36 10 FCT/US94/14388 However, any other vector may be used as long as it ib replicable and viable in the host.
The appropriate DNA sequence may be inserted into the vector by a variety of procedures. In general, the DNA sequence is inserted into an appropriate restriction endonucleaee site(s) by procedures known in the art. Such procedures and others are deemed to be within the scope of those skilled in the art The DNA sequence in the expression vector is operatively linked to an appropriate expression control sequence(s) (promoter) to direct mRNA synthesis As representative examples of such promoters, there may be mentioned LTR or SV40 promoter, the E coll. lac or trp■ the phage lambda PL promoter and other promoters known to control expression of genes m prokaryotic or eukaryotic cells or their viruses The expression vector also contains a ribosome binding site for translation initiation and a transcription terminator The vector may also include appropriate sequences for amplifying expression.
In addition, the expression vectors preferably contain one or more selectable marker genes to provide a phenotypic trait for selection of transformed host cells such as dihydrofolate reductase or neomycin resistance for eukaryotic cell culture, or such as tetracycline or ampicillin resistance in E coll The vector containing the appropriate DNA sequence as hereinabove described, as well as an appropriate promoter or control sequence, may be employed to transform an appropriate host to permit the host to express the protein As representative examples of appropriate hosts, there may be mentioned bacterial cells, such as E coll. Streptomvces. Salmonella tvphimurium. fungal cells, such as yeast; insect cells such as Drosonhila S2 and Sf9. animal cells such as CHO, COS or Bowes melanoma, adenoviruses, plant cells, etc. The selection of an appropriate host is deemed 08 36 10 FCT/US94/143S8 to be within the scope of those skilled m the art from the teachings herein.
More particularly, the present invention also includes recombinant constructB comprising one or more of the sequences as broadly described above. The constructs comprise a vectorf Buch as a plasmid or viral vector, into which a sequence of the invention has been inserted, in a forward or reverse orientation In a preferred aspect of this embodiment, the construct further comprises regulatory sequences, including, for example, a promoter, operably linked to the sequence. Large numbers of suitable vectors and promoters are known to those of skill in the art, and are commercially available. The following vectors are provided by way of example. Bacterial- pQE70, pQE60, pQE-9 (Qiagen), pBS, pDIO, phagescnpt, psiX174, pbluescript SK, pBSKS, pNHBA, pNH16a, pNH18A, pNH46A (Stratagene), pTRC99a, pKK223-3, pKK233-3, pDR540, pRIT5 (Pharmacia) Eukaryotic. pWLNEO, pSV2CAT, pOGM, pXTl, pSG (Stratagene) pSVK3, pBPV, pMSG, pSVL (Pharmacia) However, any other plasmid or vector may be used as long as they are replicable and viable in the host Promoter regions can be selected from any desired gene using CAT (chloramphenicol transferase) vectors or other vectors with selectable markers Two appropriate vectors are PKK2 32-8 and PCM7 Particular named bacterial promoters include lacl, lacZ, T3, T7, gpt, lambda P„, P, and trp Eukaryotic promoters include CMV immediate early, HSV thymidine kinase, early and late SV40, LTRs from retrovirus, and mouse metallothionem-I. Selection of the appropriate vector and promoter is well within the level of ordinary skill m the art In a further embodiment, the present invention relates to host cells containing the above-described constructs The host cell can be a higher eukaryotic cell, such as a mammalifn cell, or a lower eukaryotic cell, such as a yeast 08 36 10 PCMJS94/14388 cell, or the host cell can be a prokaryotic cell, such as a bacterial cell Introduction of the construct into the hoBt cell can be effected by calcium phosphate transfection, DEAE-Dextran mediated transfection, or electroporation (Davis, L., Dibner, M , Battey, I., Basic Methods m Molecular Biology, (1986)) .
The constructs in host cells can be used in a conventional manner to produce tne gene product encoded by the recombinant sequence Alternatively, the polypeptides of the invention can be synthetically produced by conventional peptide synthesizers.
Mature proteins can be expressed m mammalian cells, yeast, bacteria, or other cells under the control of appropriate promoters Cell-free translation systems can also be employed to produce such proteins using RNAs derived from the DNA constructs of the present invention Appropriate cloning and expression vectors for use with prokaryotic and eukaryotic hosts are described by Sambrook, et al., Molecular Cloning A Laboratory Manual, Second Edition, Cold Spring Harbor, N Y., (19B9), the disclosure of which is hereby incorporated by reference Transcription of the DNA encoding the polypeptides of the present invention by higher eukaryotes is increased by inserting an enhancer sequence into the vector Enhancers are cis-acting elements of DNA, usually about from 10 to 300 bp that act on a promoter to increase its transcription Examples including the SV40 enhancer on the late side of the replication origin bp 100 to 27 0, a cytomegalovirus early promoter enhancer, the polyoma enhancer on the late side of the replication origin, and adenovirus enhancers Generally, recombinant expression vectors will include origins of replication and selectable markers permitting transformation of the host cell, e g., the ampicillin resistance gene of E. coll and S cerevisiae TRP1 gene, and a promoter derived from a highly-expressed gene to direct 08 36 10 transcription of a downstream structural sequence. Such promoters can be derived from operons encoding glycolytic erzymes such as 3-phosphoglycerate kinase (PGK), a-factor, acid phosphatase, or heat shock proteins, among others The heterologous structural sequence is assembled in appropriate phase with translation initiation and termination sequences, and preferably, a leader sequence capable of directing secretion of translated protein into the penplasmic space or extracellular medium Optionally, the heterologous sequence can encode a fusion protein including an N-terminal identification peptide imparting desired characteristics, e.g., stabilization or simplified purification of expressed recombinant product Useful expression vectors for bacterial use are constructed by inserting a structural DNA sequence encoding a desired protein together with suitable translation initiation and termination signals in operable reading phase with a functional promoter The vector will comprise one or more phenotypic selectable markers and an origin of replication to ensure maintenance of the vector and to, if desirable, provide amplification within the host Suitable prokaryotic hosts for transformation include E co11. Bacillus subtilis. Salmonella typhimunum and various species within the genera Pseudomonas, Streptomyces, and Staphylococcus, although others may also be employed as a matter of choice.
As a representative but nonlimiting example, useful expression vectors for bacterial use can comprise a selectable marker and bacterial origin of replication derived from commercially available plasmids comprising genetic elements of the well known cloning vector pBR322 (ATCC 37017) Such commercial vectors include, for example, pKK223-3 (Pharmacia Fine Chemicals, Uppsala, Sweden) and GEM1 (Promega Biotec, Madison, WI, USA) These pBR322 "backbone" -15 — 08 36 10 PCT/U S!>4/14388 sections are combined with an appropriate promoter and the structural sequence to be expressed Following transformation of a suitable host strain and growth of the host strain to an appropriate cell density, the selected promoter is induced by appropriate means (e g., temperature shift or chemical induction) and cells are cultured for an additional period.
Cells are typically harvested by centrifugation, disrupted by physical or chemical means, and the resulting crude extract retained for further purification Microbial cells employed m expression of protemB can be disrupted by any convenient method, including freeze-thaw cycling, sonication, mechanical disruption, or use of cell lysmg agents, such methods are well know to those skilled in the art Various mammalian cell culture systems can also be employed to express recombinant protein Examples of mammalian expression systems include the COS-7 lines of monkey kidney fibroblasts, described by Gluzman, Cell, 23 175 (1981), and other cell lines capable of expressing a compatible vector, for example, the C127, 3T3, CHO, HeLa and BHK cell lines Mammalian expression vectors will comprise an origin of replication, a suitable promoter and enhancer, and also any necessary ribosome binding sites, polyadenylation site, splice donor and acceptor sites, transcriptional termination sequences, and 5' flanking nontranscribed sequences. DNA sequences derived from the SV40 splice, and polyadenylation sites may be UBed to provide the required nontranscribed genetic elements The VIGF polypeptides can be recovered and purified from recombinant cell cultures by methods including ammonium sulfate or ethanol precipitation, acid extraction, anion or cation excnange chromatography, phosphocellulose chromatography, hydrophobic interaction chromatography, affinity chromatography, hydroxylapatite chromatography and 08 36 10 lectin chromatography. Protein refolding steps can be used, as neceBsary, in completing configuration of the mature protein. Finally, high performance liquid chromatography (HPLC) can be employed for final purification steps.
The polypeptides of the present invention may be a naturally purified product, or a product of chemical synthetic procedures, or produced by recombinant techniques from a prokaryotic or eukaryotic host (for example, by bacterial, yeast, higher plant, insect and mammalian cells in culture) Depending upon the host employed in a recombinant production procedure, the polypeptides of the present invention may be glycosylated or may be non-glycosylated. Polypeptides of the invention may also include an initial methionine amino acid residue This VIGF polypeptide of the present invention may be employed m wound-healing and associated therapies concerned with re-growth of tissue, such as connective tissue, skin, bone, cartilage, muscle, lung or kidney VIGF polypeptide may also be employed to enhance the growth of vascular smooth muscle and endothelial cells leading to the stimulation of angiogenesis The VIGF— mediated increase m angiogenesis would be beneficial to ischemic tissues and to collateral coronary development in the heart subsequent to coronary stenosis VIGF polypeptide may also be employed during implant fixation to stimulate the growth of cells around the implant and therefore, facilitate it6 attachment to its intended site VIGF polypeptide may also be employed to increase IGF stability in tissues or in serum It may also increase binding to the IGF receptor Since IGF has been shown in vitro to enhance human marrow erythroid and granulocytic progenitor cell growth, VIGF polypeptide may also be employed to stimulate erythropoiesiB or granulopoiesis. 08 36 10 PCT/VS94114388 In accordance with yet a further aspect of the present invention, there is provided a process for utilizing such polypeptides, or polynucleotides encoding such polypeptides, as a research reagent for in vitro purposes related to scientific research, synthesis of DNA and manufacture of DNA vectors, for the purpose of developing therapeutics and diagnostics for the treatment of human diBeaBe Fragments of the full length VIGF gene may be used as a hybridization probe for a cDNA library to isolate the full length VIGF gene and to isolate other genes which have a high sequence similarity to the VIGF gene or similar biological activity Probes of this type can be, for example, between 20 and 2000 base pairs. Preferably, however, the probes have between 30 and 50 bases The probe may also be used to identify a cDNA clone corresponding to a full length transcript and a genomic clone or clones that contain the complete VIGF gene including regulatory and promotor regions, exons, and mtrons An example of a screen comprises isolating the coding region of the VIGF gene by using the known DNA sequence to synthesize an oligonucleotide probe Labeled oligonucleotides having a sequence complementary to that of the gene of the present invention are used to screen a library of human cDNA, genomic DNA or mRNA to determine which members of the library the probe hybridizes to This invention provides a method for identification of the receptor for VIGF The gene encoding the receptor can be identified by numerous methods known to those of skill in the art, for example, ligand panning and FACS sorting (Coligan, et al , Current Protocols in Immun , 1(2), Chapter 5, (1991)). Preferably, expression cloning is employed wherein polyadenylated RNA is prepared from a cell responsive to VIGF, and a cDNA library created from this RNA is divided into pools and used to transfect COS cells or other cells that are not responsive to VIGF. Transfected cells which are grown on glass slides are exposed to labeled VIGF. VIGF can 08 36 10 PCT/US94/1438S be labeled by a variety of means including lodination or inclusion of a recognition site for a site-specific protein kinaBe. Following fixation and incubation, the slides are subjected to autoradiographic analysis Positive pools are identified and sub-pools are prepared and retransfected using an iterative Bub-pooling and rescreening process, eventually yielding a single clone that encodes the putative receptor As an alternative approach for receptor identification, labeled VIGF can be photoaffmity linked with cell membrane or extract preparations that express the receptor molecule Cross-linked material is resolved by PAGE and exposed to X-ray film. The labeled complex containing the VIGF-receptor can be excised, resolved into peptide fragments, and subjected to protein microsequencing. The amino acid sequence obtained from microsequencing would be used to design a set of degenerate oligonucleotide probes to screen a cDNA library to identify the gene encoding the putative receptor.
This invention is also related to a method of screening compounds to identify those which mimic VIGF (agonists) or prevent the effect of VIGF An example of such a method takes advantage of the ability of VIGF to stimulate the proliferation of endothelial cells m the presence of the comitogen Con A Human umbilical vein endothelial cells are obtained and cultured in 9 6-well flat-bottomed culture plates (Costar, Cambridge, MA) and supplemented with a reaction mixture appropriate for facilitating proliferation of the cells, the mixture containing Con-A (Calbiochem, La Jolla, CA) Con-A and the compound to be screened are added and after incubation at 37°C, cultures are pulsed with J[H]thymidine and harvested onto glass fiber filters (PhD, Cambridge Technology, Watertown, MA) Mean J[H]-thymidine incorporation (cpm) of triplicate cultures is determined using a liquid scintillation counter (Beckman Instruments, 08 36 10 Irvine, CA) . Significant 3[H]-thymidine incorporation indicates stimulation of endothelial cell proliferation.
To assay for antagonists, the assay described above is performed, however, in this assay VIGF is added along with the compound to be screened and the ability of the compound to inhibit J[H]-thymidine incorporation m the presence of VIGF, indicates that the compund is an antagonist to VIGF. Alternatively, VIGF antagonists may be detected by combining VIGF and a potential antagonist with membrane-bound VIGF receptors or recombinant receptors under appropriate conditions for a competitive inhibition assay VIGF can be labeled, such as by radioactivity, such that the number of VIGF molecules bound to the receptor can determine the effectiveness of the potential antagonist Also, a mammalian cell or membrane preparation expressing the VIGF receptor would be incubated with labeled VIGF m the presence of the compound The ability of the compound to enhance or block this interaction could then be measured. Alternatively, VIGF, labelled IGF and a potential compound could be incubated under conditions where VIGF would naturally bind to IGF The extent of this interaction could be measured to determine if the compound is an effective antagonist or agonist Examples of potential VIGF antagonists include an antibody, or m some cases, an oligonucleotide, which binds to the polypeptide Alternatively, a potential antagonist may be a closely related protein, for example, a mutated form of VIGF, which recognizes the VIGF receptor but imparts no effect, thereby competitively inhibiting the action of VIGF.
Another potential VIGF antagonist is an antisense construct prepared using antisense technology Antisense technology can be used to control gene expression through triple-helix formation or antisense DNA or RNA, both of which methods are based on binding of a polynucleotide to DNA or RNA For example, the 5' coding portion of the 08 36 10 PCT/US94/I4388 polynucleotide sequence, which encodes for the mature polypeptides of the present invention, is used to deaign an antisense RNA oligonucleotide of from about 10 to 40 base pairs in length. A DNA oligonucleotide is designed to be complementary to a region of the gene involved in transcription (triple helix -see Lee et al , Nucl Acids Res., 6.3073 (1979), Cooney et al, Science, 241:456 (1988), and Dervan et al., Science, 251 1360 (1991)), thereby preventing transcription and the production of VIGF The antisense RNA oligonucleotide hybridises to the mRNA m vivo and blocks translation of the mRNA molecule into the VIGF (antisense - Okano, J Neurochem, , 56:560 (1991), Oligodeoxynucleotides as Antisense Inhibitors of Gene Expression, CRC Press, Boca Raton, FL (1988)) The oligonucleotides described above can also be delivered to cells such that the antisense RNA or DNA may be expressed in vivo to inhibit production of VIGF.
Potential VIGF antagonists include small molecules which bind to the active site, the receptor binding site, IGF or other grovrth factor binding site of the polypeptide thereby blocking the normal biological activity of VIGF Examples of small molecules include but are not limited to small peptides or peptide-like molecules The antagonists may be employed to inhibit tumor neovascularization and the neointimal proliferation of smooth muscle cells prevalent in atherosclerosis and restenosis subsequent to balloon angioplasty The antagonists may also be employed to inhibit the over production of scar tissue seen m a keloid which forms after surgery, fibrosis after myocardial infarction, or fibrotic lesions associated with pulmonary fibrosis The antagonists may be employed in a composition with a pharmaceutically acceptable carrier, e.g , as hereinafter described.
The VIGF polypeptides and antagonist or agonists of the present invention may be employed in combination with a 08 36 10 suitable pharmaceutical carrier. Such compositions comprise a therapeutically effective amount of the polypeptide, and a pharmaceutical^ acceptable carrier or excipient Such a carrier includes but is not limited to saline, buffered saline, dextrose, water, glycerol, ethanol, and combinations thereof. The formulation should suit the mode of administration The invention also provides a pharmaceutical pack or kit comprising one or more containers filled with one or more of the ingredients of the pharmaceutical compositions of the invention Associated with such container(e) can be a notice m the form prescribed by a governmental agency regulating the manufacture, use or sale of pharmaceuticals or biological products, which notice reflects approval by the agency of manufacture, use or sale for human administration In addition, the pharmaceutical compositions may be employed m conjunction with other therapeutic compounds The pharmaceutical compositions may be administered in a convenient manner such as by rhe oral, topical, intravenous, intraperitoneal, intramuscular, subcutaneous, intranasal or intradermal routes The pharmaceutical compositions are administered in an amount which is effective for treating and/or prophylaxis of the specific indication In general, they are administered m an amount of at least about 10 ^g/kg body weight and in most cases they will be administered m an amount not in excess of about 8 mg/Kg body weight per day In most cases, the dosage is from about 10 jig/kg to about 1 mg/kg body weight daily, taking into account the routes of administration, symptoms, etc VIGF in combination with other growth factors including but not limited to, PDGF, IGF, FGF, EGF or TGF-/J may accelerate physiological responses as seen m wound healing The VIGF polypeptide and agonists and antagonists which are polypeptides, may also be employed in accordance with the 08 36 10 present invention by expression of such polypeptides m vivo, which is often referred to as "gene therapy." Thus, tor example, cells from a patient may be engineered with a polynucleotide (DNA or RNA) encoding a polypeptide ex vivo, with the engineered cells then being provided to a patient to be treated with the polypeptide Such methods are well—known in the art For example, cells may be engineered by procedures known in the art by use of a retroviral particle containing RNA encoding a polypeptide of the present invention.
Similarly, cells may be engineered in vivo for expression of a polypeptide in vivo by, for example, procedures known in the art As known in, the art, a producer cell for producing a retroviral particle containing RNA encoding the polypeptide of the present invention may be administered to a patient for engineering cells in vivo and expression of the polypeptide in vivo. These and other methods for administering a polypeptide of the present invention by such method should be apparent to those skilled in the art from the teachings of the present invention For example, the expression vehicle for engineering cells may be other than a retrovirus, for example, an adenovirus which may be used to engineer cells in vivo after combination with a suitable delivery vehicle.
This invention is also related to the use of the VIGF gene as a diagnostic Detection of a mutated form of VIGF will allow a diagnosis of a disease or a susceptibility to a disease, such as a tumor, since mutations m VIGF may cause tumors Individuals carrying mutations in the human VIGF gene may be detected at the DNA level by a variety of techniques Nucleic acids for diagnosis may be obtained from a patient's cells, 6uch as from blood, urine, saliva, tissue biopsy and autopsy material The genomic DNA may be used directly for detection or may be amplified enzymatically by using PGR 08 36 10 WOW/17931 (Saiki et al. , Nature, 324 * 163-166 (1986)) prior to analysis RNA or cDNA may also be used for the same purpose As an example, PCR primers complementary to the nucleic acid encoding VIGF can be UBed to identify and analyze VIGF mutations For example, deletions and insertions can be detected by a change m size of the amplified product in comparison to the normal genotype Point mutations can be identified by hybridizing amplified DNA to radiolabeled VIGF RNA or alternatively, radiolabeled VIGF antisense DNA sequences Perfectly matched sequences can be distinguished from mismatched duplexes by KNase A digestion or by differences in melting temperatures Genetic testing based on DNA sequence differences may be achieved by detection of alteration in electrophoretic mobility of DNA fragments in gels with or without denaturing agents Small seguence deletions and insertions can be visualized by high resolution gel electrophoresis DNA fragments of different sequences may be distinguished on denaturing formamidme gradient gels in which the mobilities of different DNA fragments are retarded in the gel at different positions according to their specific melting or partial melting temperatures (see, e g , Myers et al. , Science, 230 1242 (19B5)).
Sequence changes at specific locations may also be revealed by nuclease protection assays, such as RNase and SI protection or the chemical cleavage method (e.g , Cotton et al , PNAS, USA, 85.4397-4401 (19B5)) Thus, the detection of a specific DNA sequence may be achieved by methods such as hybridization, RNase protection, chemical cleavage, direct DNA sequencing or the use of restriction enzymes, (e.g , Restriction Fragment Length Polymorphisms (RFLP)) and Southern blotting of genomic DNA In addition to more conventional gel-electrophoresis and DNA sequencing, mutations can also be detected by in situ analysis 08 36 10 VIGF protein expression may be linked to vascular disease or neovascularization associated with tumor formation. VIGF has a signal peptide and the mRKA is highly expressed in endothelial cells and to a lesser extent in smooth muscle cells which indicates that the protein ib present in serum Accordingly, an anti-VIGF antibody could be used to diagnose vascular disease or neovascularization associated with tumor formation since an altered level of this polypeptide may be indicative of such disorders A competition assay may be employed wherein antibodies specific to VIGF is attached to a solid support »*"nd labeled VIGF and a sample derived from the host are pas6ed over the solid support and the amount of label detected attached to the solid support- can be correlated to a quantity of VIGF m the sample The sequences of the present invention are also valuable for chromosome identification The sequence is specifically targeted to and can hybridize with a particular location on an individual human chromosome Moreover, there is a current need for identifying particular sites on t.he chromosome. Few chromosome marking reagents based on actual sequence data (repeat polymorphisms) are presently available for marking chromosomal location. The mapping of DNAs to chromosomes according to the present invention zb an important first step in correlating those sequences with genes associated with disease Briefly, sequences can be mapped to chromosomes by preparing PCR primers (preferably 15-25 bp) from the cDNA. Computer analysis of the 3' untranslated region is used to rapidly select primers that do not span more than one exon in the genomic DNA, thus complicating the amplification process. These primers are then used for PCR screening of somatic cell hybrids containing individual human chromosomes. Only those hybrids containing the human gene corresponding to the primer will yield an amplified fragment 08 36 10 PCR mapping of somatic cell hybrids is a rapid procedure for assigning a particular DNA to a particular chromosome UBing the preBent invention with the same oligonucleotide primers, sublocalization can be achieved with panels of fragments from specific chromosomes or pools of large genomic clones in an analogous manner Other mapping strategies that can similarly be used to map to its chromosome include in situ hybridization, prescreening with labeled flow-sorted chromosomes and preselection by hybridization to construct chromosome specific-cDNA libraries Fluorescence xn situ hybridization (FISH) of a cDNA clone to a metaphase chromosomal spread can be used to provide a precise chromosomal location in one step. This technique can be used with cDNA as short as 500 or 600 bases, however, clones larger than 2,000 bp have a higher likelihood of binding to a unique chromosomal location with sufficient signal intensity for simple detection FISH requires use of the clones from which the EST was derived, and the longer the better. For example, 2,000 bp is good, 4,000 is better, and more than 4,000 is probably not necessary to get good results a reasonable percentage of the time. For a review of this technique, Bee Verma et al , Human Chromosomes, a Manual of Basic Techniques, Pergamon Press, New York (19 88) Once a sequence has been mapped to a precise chromosomal location, the physical position of the sequence on the chromosome can be correlated with genetic map data Such data are found, for example, in V McKusick, Mendelian Inheritance in Man (available on line through Johns Hopkins University Welch Medical Library) The relationship between genes and diseases that have been mapped to the same chromosomal region are then identified through linkage analysis (coinhentance of physically adjacent genes) Next, it is necessary to determine the differences in the cDNA or genomic sequence between affected and unaffected individuals If a mutation is observed in some or all of the 08 36 10 affected individuals but not. in any normal individuals, then the mutation is likely to be the causative agent of the disease.
With current resolution of phyeical mapping and genetic mapping techniques, a cDNA precisely localized to a chromosomal region associated with the disease could be one of between 50 and 500 potential causative genee (This assumes 1 megabase mapping resolution and one gene per 20 kb) The polypeptides, their fragments or other derivatives, or analogs thereof, or cells expressing them can be used as an immunogen to produce antibodies thereto. These antibodies can be, for example, polyclonal or monoclonal antibodies. The present invention also includes chimeric, single chain, and humanized antibodies, as well as Fab fragments, or the product of an Fab expression library Various procedures known in the art may be used for the production of such antibodies and fragments Antibodies generated against the polypeptides corresponding to a sequence of the present invention can be obtained by direct injection of the polypeptides into an animal or by administering the polypeptides to an animal, preferably a nonhuman The antibody so obtained will then bind the polypeptides itself In this manner, even a sequence encoding only a fragment of the polypeptides can be used to generate antibodies binding the whole native polypeptides Such antibodies can then be used to isolate the polypeptide from tissue expressing that polypeptide.
For preparation of monoclonal antibodies, any technique which provides antibodies produced by continuous cell line cultures can be used Examples include the hybridomo technique (Kohler and Milstem, 1975, Nature, 256:495-497 ), the trioma technique, the human B-cell hybridoma technique (Kozbor et al., 1983, Immunology Today 4 72), and the EBV-hybndoma technique to produce human monoclonal antibodies 08 36 10 (Cole, et al., 1985, in Monoclonal Antibodies and Cancer Therapy, Alan R. Lisb, Inc , pp 77-96), Techniques described for the production of single chain antibodies (U.S. Patent 4,946,770) can be adapted to produce single chain antibodies to immunogenic polypeptide products of this invention. Also, transgenic mice may be used to express humanized antibodies to immunogenic polypeptide products of this invention.
The present invention will be further described with reference to the following examples, however, it is to be understood that the present invention is not limited to such examples All parts or amounts, unless otherwise specified, are by weight In order to facilitate understanding of the following examples certain frequently occurring methods and/or terms will be described.
"Plasmids" are designated by a lower case p preceded and/or followed by capital letters and/or numbers The starting plasmids herein are either commercially available, publicly available on an unrestricted basis, or can be constructed from available plasmids in accord with published procedures. In addition, equivalent plasmids to those described are known in the art and will be apparent to the ordinarily skilled artisan- "Digestion" of DNA refers to catalytic cleavage of the DNA with a restriction enzyme that acts only at certain sequences in the DNA. The various restriction enzymes used herein are commercially available and their reaction conditions, cofactors and other requirements were used as would be known to the ordinarily skilled artisan. For analytical purposes, typically 1 fjg of plasmid or DNA fragment is used with about 2 units of enzyme in about 20 y1 of buffer solution For the purpose of isolating DNA fragments for plasmid construction, typically 5 to 50 pq of DNA are digested with 20 to 250 units of enzyme in a larger 08 36 10 volume. Appropriate buffers and substrate amounts for particular restriction enzymes are specified by the manufacturer. Incubation times of about 1 hour at 37*C are ordinarily used, but may vary in accordance with the supplier's instructions. After digestion the reaction is electrophoresed directly on a polyacrylamide gel to isolate the desired fragment.
Size separation of the cleaved fragments is performed using 8 percent polyacrylamide gal described by Goeddel, D et al , Nucleic Acids Res., 8:4057 (1980).
"Oligonucleotides" refers to either a single stranded polydeoxynucleotide or two complementary polydeoxynucleotide strands which may be chemically synthesized Such synthetic oligonucleotides have no 5' phosphate and thus will not ligate to another oligonucleotide without adding a phosphate with an ATP in the presence of a kinase A synthetic oligonucleotide will ligate to a fragment that has not been dephosphorylated.
"Ligation" refers to the process of forming phosphodiester bonds between two double stranded nucleic acid fragments (Maniatis, T., et al., Id., p 146) Unless otherwise provided, ligation may be accomplished using known buffers and conditions with 10 units of T4 DNA ligaBe ("ligase") per 0 5 jjg of approximately equimolar amounts of the DNA fragments to be ligated Unless otherwise stated, transformation was performed as described in the method of Graham, F and Van der Eb, A., Virology, 52 456-457 (1973) Example 1 Bacterial Expression and Purification of VIGF The DNA sequence encoding VIGF, ATCC # 75874, was initially amplified using PCR oligonucleotide primers corresponding to the 5' sequences of the processed VIGF protein (minus the signal peptide sequence) and the vector sequences 3' to the VIGF gene. Additional nucleotides 08 36 10 corresponding to VIGF were added to the 5' and 3' sequences respectively. The 5' oligonucleotide primer has the sequence 5' CGCAAQCTTAAATAATTATGCGGTGGACTGC 3' contains a Hind III restriction enzyme site (in bold) followed by 21 nucleotides of VIGF coding sequence starting from the presumed terminal ammo acid of the processed protein codon (underlined). The 3' oligonucleotide primer 5' CGCTCTftGATpAGCGTGGATTTAACCA 3' contains an Xba I restriction Bite (in bold) followed by the reverse complement of nucleotides corresponding to the carboxy-terminal 5 amino acids and the trantlational stop codon (underlined). The restriction enzyme sites correspond to the restriction enzyme sites on the bacterial expression vector pQE-9 (Qiagen, Inc Chatsworth, CA,). pQE-9 encodes antibiotic resistance (Amp'), a bacterial origin of replication (on), an IPTG-regulatable promoter operator (P/0), a ribosome binding site (RBS), a 6-His tag and restriction enzyme sites The VIGF PCR product and pQE-9 were then digested with Hind III and Xba I and ligated together with T4 DNA ligase The desired recombinants would contain the VIGF coding sequence inserted downstream from the pQE-9 encoded histidine tag and the ribosome binding site. The ligation mixture was then used to transform E. coli strain M15[pREP4] (Qiagen, Inc.) by the procedure described in Sambrook, J et al , Molecular Cloning A Laboratory Manual, Cold Spring Laboratory Press, (1989) M15[pREP4] contains multiple copies of the plasmid pREP4, which expresses the lacl repressor and also confers kanamycin resistance (Kanr) Transformants were identified by their ability to grow on LB plates and ampicillin/kanamycm resistant colonies were selected Plasmid DNA was isolated and confirmed by restriction analysis Clones containing the desired constructs were grown overnight (0/N) in liquid culture in LB media supplemented with both Amp (100 ug/ml) and Kan (25 ug/ml) The 0/N culture was used to inoculate a large culture at a ratio of 1 100 to 1 250 The cells were 08 36 10 PCT/U S94/14388 grown to an optical density 600 (O.D.600) of between 0.4 and 0 6 IPTG ("Iaopropy1—B-D-thiogalacto pyranoside") was then added to a final concentration of 1 mM. IPTG induces by inactivating the lacl repressor, clearing the P/O leading to increased gene expression. Cells were grown an extra 3 to 4 hours such that there is an exponential growth culture present Cells were then harvested by centrifugation The VIGF/6-Histidine-containing M15 [pREP4 ] cells were lyaed in 6M GnHCl,50 mM NaP04 at pH B.O. The lysate was loaded on a Nickel-Chelate column and the flow-through collected. The column was washed with 6M GnHCl, 5 0 mM NaPO, at pH B.O, 6 0 and 5.0 The VIGF fusion protein (>90% pure) was eluted at pH 2 0 Samples from the pre-column lysate (Figure 3, lane 2), column flow through (lane 3), pH 5.0 wash (lane 4), and pH 2.0 eluate (lane 5) were precipitated with sodium deoxycholate and trichloroacetic acid. For the purpose of renaturation, the pH 2.0 eluate was adjusted to 3 molar guanidme HC1, lOOmM sodium phosphate, 10 mmolar glutathione (reduced) and 2 mmolar glutathione (oxidized) After incubation in this solution for 12 hours the protein was dialyzed to 10 mmolar sodium phosphate. To run the gel, the pellets were resuspended in SDS/NaOH and SDS-PAGE loading buffer, heat denatured, then electrophoresed on a 15% denaturing polyacrylamide gel The Gibco BRL low range molecular weight standard was also electrophoresed (lane 1) The proteins were visualized with Coomassie Brilliant Blue R-250 stain Figure 3 shows an SDS-polyacrylamide gel which displays the results of VIGF purification.
Example 2 Cloning and expression of VIGF using the baculovirus expression system The DNA sequence encoding the full length VIGF protein, ATCC # 75B74, is digested with the restriction enzymes PvuII and Xbal. The 639 nucleotide PvuII, Xbal fragment contains 08 36 10 PCTAJS94/14388 the entire VIGF coding region plus 11 and 77 nucleotides of 5* and 3' untranslated DNA, respectively This fragment, designated F2, is isolated from a 1% agarose gel using a commercially available kit ("Geneclean", iJlO 101 Inc., La Jolla, Ca ) The vector pA2 ib used for the expression of the VIGF protein using the baculovirus expression syBtem (for review see Summers, M D and Smith, G.E. 1987, A manual of methods for baculovirus vectors and insect cell culture procedures, Texas Agricultural Experimental Station Bulletin No. 1555) This expression vector contains the strong polyhedrin promoter of the Autographa califormca nuclear polyhidrosis virus (AcMNPV) followed by the recognition sites for the restriction endonucleases Smal and Xbal. The polyadenylation site of the simian virus (SV)40 is used for efficient polyadenylation For an easy selection of recombinant viruses the beta-galactosidase gene from E.coli is inserted in the same orientation as the polyhedrin promoter followed by the polyadenylation signal of the polyhedrin gene. The polyhedrin sequences are flanked at both sides by viral sequences for the celx-mediated homologous recombination of cotransfected wild-type viral DNA Many other baculovirus vectors could be used in place of pA2 such as, pRGl, pAc373, pVL941 and pAcIMl (Luckow, V A and Summers, M D , Virology, 170 31-39) The plasmid is digested with the restriction enzymes Smal and Xbal and then dephosphorylated using calf intestinal phosphatase by procedures known in the art The DNA is then isolated from a 1% agarose gel using the commercially available kit ("Geneclean" BIO 101 Inc , La Jolla, Ca. ) This vector DNA is designated V2.
Fragment F2 and the dephosphorylated plasmid V2 are ligated with T4 DNA ligase E coli strain XL1 Blue (Stratagene Cloning Systems, 11011 North Torrey Pines Road La Jolla, Ca 92037) are then transformed and bacteria * 08 36 10 WOW17931 identified that contained the plasmid (pBac VIGF) with the VIGF cDNA using the enayraes BamHI and Xbal Thts sequence of the cloned fragment is confirmed by DNA sequencing /ig of the plasmid pBac VIGF is cotranBfected with 1.0 /ig of a commercially available linearized baculovirus ("BaculoGoldT" baculovirus DNA", Pharmingen, San Diego, CA ) using the lipofection method (Feigner et al Proc Natl. Acad Sci USA, 84 7413-7417 (1987)) 1/ag of BaculoGold™" virus DNA and 5 fig of the plasmid pBac VIGF are mixed ixi a sterile well of a microtiter plate containing 50 fil of serum free Grace'b medium (Life Technologies Inc , Gaithersburg, MD) Afterwards 10 /il Lipofectm plus 90 pi Grace's medium are added, mixed and incubated for 15 minutes at room temperature Then, the transfection mixture is added dropwise to the Sf9 insect cells (ATCC CRL 1711) seeded in a 35 mm tissue culture plate with lml Grace' medium without serum The plate is reeked back and forth to mix the newly added solution The plate is then incubated for 5 hours at 27°C. After 5 hours the transfection solution is removed from the plate and 1 ml of Grace's insect medium supplemented with 10\ fetal calf serum is added The plate is put back into an incubator and cultivation continued at 27°C for four days After four days the supernatant is collected and a plaque assay performed similar as described by Summers and Smith (supra). As a modification an agarose gel with "Blue Gal" (Life Technologies Inc , Gaithersburg) is used which allows an easy isolation of blue stained plaques (A detailed description of a "plaque assay" can also be found in the user's guide for insect cell culture and baculovirology distributed by Life Technologies Inc , Gaithersburg, page 9-10) .
Four days after the serial dilution, the viruses are added to the cells and blue stained plaques are picked with the tip of an Eppendorf pipette The agar containing the 08 36 10 recombinant viruses is then resuspended in eui Eppendorf tube containing 200 /xl of Grace's medium The agar ib removed by a brief centnfugation and the supernatant containing the recombinant baculoviruBes is used to infect Sf9 cells seeded in 35 mm dishes. Four days later the supernatants of these culture dishes are harvested and then stored at 4°C.
Sf9 cells are grown in Grace's medium supplemented with 10% heat-inactivated FBS The cells are infected with the recombinant baculovirus V-VIGP at a multiplicity of infection (MOI) of 2 Six hours later the medium is removed and replaced with SF900 II medium minus methionine and cysteine (Life Technologies Inc , Gaithersburg) 42 hours later 5 /iCi of 33S-methionine and 5 /iCi 3iS cysteine' (AmerBhara) are added The cells are further incubated for IS hours before they are harvested by centnfugation and the labelled protemB visualized by SDS-PAGE and autoradiography Example 3 Expression of Recombinant VIGF m CHO cellB The vector pN346 is used for the expression of the VIGF protein. Plasmid pN346 is a derivative of the plasmid pSV2-dhfr [ATCC Accession No 37146] Both plasmids contain the mouse dhfr gene under control of the SV40 early promoter Chinese hamster ovary or other cells lacking dihydrofolate activity that are transfected with these plasmids can be selected by growing the cells in a selective medium (alpha minus MBM, Lift Technologies) supplemented with the chemotherapeutic agent methotrexate The amplication of the DHFR genes in cells resistant to methotrexate (MTX) has been well documented (see, e g , Alt, F W , Kellems, R M , Bertino, J R , and Schimke, R T , 1978, J Biol Chem 253 :1357-1370, Hamlin, J L and Ma, C 1990, Biochem et Biophys Acta, 1097 107-143, Page, M J and Sydenham, M.A 1991, Biotechnology Vol. 9 64-68) Cells grown m increasing concentrations of MTX develop resistance to the drug by 08 36 10 V/O 96/17931 PCT/US94/143JW overproducing the target enzyme, DHFR, as a result of amplification of the DHFR gene If a second gene is linked co the dhf 1 gene it is usually co-amplified and overexpressed Subsequently, when the methotrexate is withdrawn, cell lines contain the amplified gene integrated into the chromosome(s).
Plasmid pN346 contains for the expression of the gene of interest a strong promoter of the long terminal repeat (LTR) of the Rouse Sarcoma Virus (Cullen, et al , Molecular and Cellular Biology, March 1985, 438-447) plus a fragment isolated from the enhancer of the immediate early gene of human cytomegalovirus (CMV) (Boshart et al , Cell 41:521-530, 1985) Downstream of the promoter are the following single restriction enzyme cleavage sites that allow the integration of the genes BamHI, Pvull, and Nrul. Behind these cloning sites the plasmid contains translational stop codons m all three reading frames followed by the 3' mtron and the polyadenylation site of the rat preproinsulm gene Other high efficient promoters can also be used for the expression, eg., the human B-actin promoter, the SV40 early or late promoters or the long terminal repeacs from ocher retroviruses, e g , HIV and HTLVI For the polyadenylation of the mRNA other signals, e g , from the human growth hormone or globin genes can be used as well Stable cell lines carrying a gene of interest integrated into the chromosome can also be selected upon co-tranBfection with a selectable marker such as gpt, G418 or hygromycin It is advantageous to use more than one selectable marker in the beginning, e g G418 plus methotrexate The plasmid pN346 is digested with the restriction enzyme BamHI and then dephosphorylated using calf intestinal phosphatase by procedures known in the art The vector is then isolated from a 1% agarose gel 08 36 10 The DNA sequence encoding the full length VIGP protein, ATCC #75874, is amplified using PCR oligonucleotide primers corresponding to the 5' and 3' sequences of the gene The 5' primer has the sequ&nce 5' CGCAQATCTCCGCCACCATGAA GAGCGTCTTGCTGCTG 3' and contains a Bglll restriction enzyme site (in bold) followed by B nucleotides resembling an efficient signal for the initiation of translation m eukaryotic cells (Kozak, M-, J Mol Biol , 196 947-950, (19B7) ) The remaining nucleotides correspond to the amino terminal 7 attuno acids including the translational initiation codon (underlined) The 3' primer has the sequence 5' CGCAGA.TCTAGCCTTCTCTCASAAATCA.CA 3' and contains a Bglll restriction site (m bold) sn<3. 21 nucleotides that are the reverse complement of 3' untranslated DNA starting 7 nucleotides downstream from the translational stop codon The PCR product is digested with Bglll and purified on a l% agarose gel using a commercially available kit ("Geneclean," BIO 101 Inc , La Jolla, Ca ) This fragment is then ligated to BamHI digested, phosphatased pN346 plasmid with T4 DNA ligase XllBlue (Stratagene) E coll are transformed and plated on LB, 50 ug/ml ampicillm plates Colonies bearing the desired recombinant m the proper orientation are screened for by PCR with a 5* primer which corresponds to the Rous sarcoma virus promoter anQ a 3 • primer which corresponds to the reverse complement of VIGP codons 73-79 The sequence of the cloned fragment is confirmed by DNA sequencing Transfection of CH0-4hfr-cslla Chinese hamster ovary cells lacking an active DHFR enzyme are used for transfection. 5 jug of the expression plasmid pN346VIGF are cotransf ected with 0.5 fig of the plasmid pSVneo using the lipofectm method (Feigner et al , Bupra) The plasmid pSV2-neo contains a dominant selectable marker, the gene neo from Tn.5 encoding an enzyme that confers resistance to a group of antibiotics including G418 The cells are seeded m alpha minus MEM supplemented with l mg/ml 08 36 10 G41B After 2 days, the cells are trypsmized and seeded an hybridoma cloning plates (Greiuer, Germany) and cultivated from 10-14 days. After thiB period, single clones are trypsmized and then seeded in 6-well petn dishes using different concentrations of methotrexate (25, 50 run, 100 nni, 200 nm, 400 nm) . Clones growing at the highest concentrations of methotrexate are then transferred to new 6-well plates containing even higher concentrations of methotrexate (500 nM, 1 /iM, 2 /iM, 5 /iM) The same procedure is repeated until clones grew at a concentration of 100 /iM The expression of the desired gene product is analyzed by Western blot analysis and SDS-PAGE.
Example 4 Tissue Localization of VIGF Gene Expression bv Northern Blot Analyt^g A multiple tissue Northern blot (Clontech Laboratories, Inc , 4030 Fabian Way; Palo Alto, California 94303) containing 2 ug of human adult brain, heart, placenta, lung, liver skeletal muscle, kidney, and pancreas poly A+ mRNA per lane is prehybndized m Church buffer (Church, G M & Gilbert, W , Proc Natl Acad Sci USA 81, 1991-1995 (1984)) at 60°C for one hour The DNA sequence coding for VIGF, ATCC# 75874, is amplified from the full length cDNA cloned in pBluescript SK(-) using the M13 Forward (5' GGGTTTTCCCAGTCACGAC 3') and Reverse (5' ATGCTTCCGGCTCGTATG 3') primers Twenty-five nanograms of PCR product is random primer radiolabeled (Prime-It II, Stratagene Cloning Systems, 11011 Nortb Toxrey Pines Rd , La Jolla, California 92037) with 3JP-dCTP The heat denatured VIGF probe is added directly to the prehybridization buffer and incubated 16 hr at 60°C Two ten minute washes are performed in 0 2X SSC, 0 1% SDS at 60oC. Autoradiography is performed at -80°C. 08 36 10 WO 96/17931 PCT/US94/14388 A 2 3 k±> transcript is Been in lung and kidney after a four day exposure (Figure 4) Example 5 Cell-Type Analysis of VIGF Gene Expression by Northern Blot Analysis Human umbilical vein endothelial, aortic smooth muscle, dermal foreskin fibroblast cells (Clonetics, 9520 Chesapeake Drive, Suite #201, San Diego, California 92123) were grown to 75-90% confluency Total RNA is extracted with RNAzol (Biotecx Laboratories, Inc , 6023 South Loop East Houston, Texas 77033) A 1 2% agarose formaldehyde gel is prepared and run with 20 ug of total RNA per lane and an RNA ladder size marker (Life Technologies, Inc , 8400 Helgerraan Ct , P O Box 6009 Gaithersburg, Maryland 20884) according to Sambrook et al (1989) The IUJA is transferred overnight to Hybond N+ (Amersham Corp., 263 6 South Clearbrook Drive; Arlington Heights, Illinois 60005) and bound to the membrane with a Stratalinker uv Crosslinker (Stratagene Cloning Systems, La Jolla, California) The blot is prehybridized in Church buffer (Church, G M & Gilbert, W., PNAS, USA 81 1991-1995 (1984)) at 60°C for one hour The DNA sequence encoding VIGF, ATCC # 75874, is amplified from the full length cDNA cloned in pBluescnpt SK(-) using the M13 Forward (5' GGGTTTTCCCAGTCACGAC 3') and Reverse (5' ATGCTTCCGGCTCGTATG 3') primers Twenty-five nanograms of PCR product is random primer radiolabeled (Prime-It II, Stratagene) with "P-dCTP The heat denatured VIGF probe is added directly to the prehybridization buffer and incubated 16 hr at 60°C Two ten minute washes were performed in 0 2X SSC, 0 1% SDS at 60°C. Autoradiography is performed at -80°C A 2.3-2 4 kb transcript is seen m umbilical vein endothelial (lane 1) and aortic Bmooth muscle cells (lane 2) after a two hour exposure (Figure 5A) and also in dermal 08 36 10 WOW/17931 PCT/DS94/143S8 foreskin fibroblast (lane 3) cells after a 36 hour exposure (Figure 5B) 08 36 10 PCI/US94/14388 Numerous modifications and variations of the present invention are possible m light of the above teachings and, therefore, within the scope of the appended claims, the invention may be practiced otherwise than as particularly described 08 36 10 SEQUENCE LISTING (1) GENERAL INFORMATION (X) APPLICANT: HASTINGS, ET AL (ii) TITLE OF INVENTION Human Vascular IBP-Like Growth Factor (ill) NUMBER OF SEQUENCES 2 (iv) CORRESPONDENCE ADDRESS (A) ADDRESSEE- CARELLA, BYRNE, BAIN, GILFILLAN, CBCCHI, STEWART & OLSTEIN (B) STREET 6 BECKER FARM ROAD (C) CITY- ROSKLAUD (D) STATE NEW JERSEY (B) COUNTRY USA (F) ZIP 07068 (v) COMPUTER READABLE FORM (A) MEDIUM TYPE 3.5 INCH DISKETTE (B) COMPUTER IBM PS/2 (C) OPERATING SYSTEM MS-DOS (D) SOFTWARE WORD PERFECT 5 1 (vi) CURRENT APPLICATION DATA (A) APPLICATION NUMBER (B) FILING DATE Concurrently (C) CLASSIFICATION (Vll) PRIOR APPLICATION DATA (A) APPLICATION NUMBER (B) FILING DATE 08 36 10 (viii) ATTORNEY/AGENT INFORMATION (A) NAME. FERRARC, GREGORY D (B) REGISTRATION NUMBER 36,134 (C) REFERENCE/DOCKET NUMBBR 325BOO-219 (ax) TELECOMMUNICATION INFORMATION (A) TELEPHONE 201-994-1700 (B) TELEFAX 201-994-1744 (2) INFORMATION FOR SEQ ID NO 1 (i) SEQUENCE CHARACTERISTICS (A) LENGTH 1271 BASE PAIRS (B) TYPE NUCLEIC ACID (C) STRANDEDNESS SINGLE (D) TOPOLOGY LINEAR (11) MOLECULE TYPE cDNA (xi) SEQUENCE DESCRIPTION SBQ ID NO 1 CTGCTTCCCA CCAGCAAAGA CCACGACTGG AGAGCCGAGC CGGAGCAGCT a 1 CO AAGAGCGTCT TGCTGCTGAC CACGCTCCTC GTGCCTGCAC ACCTGGTGGC CGCCTGGAGC 12 0 AATAATTATG CGGTGGACTG CCCTCAACAC TGTGACAGCA GTGAGTGCAA AAGCAGCCCG 180 CGCTGCAAGA GGACAGTGCT CGACGACTGT GGCTGCTGCC GAGTGTGCGC TGCAGGGCGG 24 0 GGAGAAACTT GCTACCGCAC AGTCTCAGGC ATGGATGGCA TGAAGTGTC-G CCCGGGGCTG 300 AGGTGTCAGC CTTCTAATGG GGAGGATCCT TTTGGTGAAG AGTTTGGTAT CTGCAAAGAC 360 TGTCCCTACG GCACCTTCGG GATGGATTGC AGAGAGACCT GCAACTGCCA GTCAGGCATC 420 TGTGACAGGG GGACGGGAAA ATGCCTGAAA TTCCCCTTCT TCCAATATTC AGTAACCAAG 480 TCTTCCAACA GATTTGTTTC TCTCACGGAG CATGACATGG CATCTGGAGA TGGCAATATT 540 GTGAGAGAAG AAGTTGTGAA AGAGAATGCT GCCGGGTCTC CCGTAATGAG GAAATGGTTA 600 AATCCACGCT GATCCCGGCT GTGATTTCTG AGAGAAGGCT CTATTTTCGT GAYTGTTCAA 660 CACACAGCCA ACATTTTAGG AACTTTCTAG ATTATAGCAT AAGGACATGT AAUTITIGAA 720 GACCAAATGT GATGCATGQ1 GGATCCAGAA AACAAAAAGT AGGATACTTA CAATCCATAA 780 CATCCATATQ ACTGAACACT TGTATGTGTT TGTTAAATAT TCGAATGCAT GTAGATTTGT 840 TAAATGTGTG TGTATAGTAA CACTGAAGAA CTAAAAATGC AATTTAGGTA ATCTTACkTG 900 u 1 AACCAAAGAG GGAGCTAGGC AAAGCTGAAG ACCGCAGTGA GTCAAATTAG 960 TTCTITGACT TTGATGTACA TTAATGTTCC GATATGGAAT GAAGACTTAA GAGCAGGAGA 1020 08 36 10 AGATGGGGAG GGGGTGGGAG TGGGAAATAA AATATTTAGC CCTTCCTTGG TAGGTAGCTT 1080 CTCTAGAATT TAATTRTGCT TTTTTTTTTT TTTTTGGGC? TTGGGAAAAG TCAAAATAAA 114 0 ACAACCAGAA AACCCCTGAA GGAAGTAAGA TGTTTGAAGC TTATGGAAAT TTGAGTAACA 1200 AACAGCTTTG ANCTGAGAGC AATTYCAAAA GGCTGCTGAT GTAGCCCCCG GGTTNCCTNT 12S0 (2) INFORMATION FOR SEQ ID NO 2 (I) SEQUENCE CHARACTERISTICS (A) LENGTH 184 AMINO ACIDS (B) TYPE AMINO ACID (C) STRANDEDNESS (D) TOPOLOGY LINEAR (II) MOLECULE TYPE PROTEIN (Xi) SEQUENCE DESCRIPTION- SEQ ID NO 2 Met Lys Ser Val Leu Leu Leu Thr Thr Leu Leu Val Pro Ala His 2 7 12 Leu Val Ala Ala Trp Ser Asn Asn Tyr Ala Val Asp Cys Pro Gin 17 22 26 Has Cys Asp Ser Ser Glu Cys Lys Ser Ser Pro Arg Cys Lys Arg 31 36 41 Thr Val Leu Asp Asp Cys Gly Cys Cys Arg Val Cys Ala Ala Gly 46 51 56 Arg Gly Glu Thr Cys Tyr Arg Thr Val Ser Gly Met Asp Gly Met 61 66 71 Lys Cys Gly Pro Gly Leu Arg Cys Gin Pro Ser Asn Gly Glu Asp 76 81 86 Pro Phe Gly Glu Glu Phe Gly lie Cys -Lys Asp Cys Pro Tyr Gly 91 96 101 Thr Phe Gly Met Asp Cys Arg Glu Thr Cys Asn Cys Gin Ser Gly 106 111 116 lie Cys Asp Arg Gly Thx Gly Lys Cys Leu Lys Phe Pro Phe Phe 121 126 131 Gin Tyr Ser Val Thr Lys Ser Ser Asn Arg Phe Val Ser Leu Thr 136 141 1J6 Glu His Asp Met Ala Ser Gly Asp Gly Asn lie Val Arg Glu Glu 151 156 161 Val Val Lys Glu Asn £JLa 2U-a Glv Ser Pro Val Met Arg Lys Trp 166 171 176 Leu Asn Pro Arg NTCTOAAGGA C 1271 181 43 - j ~8s£Pi3S9 Pc

Claims (69)

WHAT IS CLAIMED IS
1. An isolated polynucleotide selected from the group consisting of (a) a polynucleotide encoding the VIGF polypeptide having the deduced amino acid sequence of Figure 1 or a fragment, analog or derivative of said polypeptide, (b) a polynucleotide encode ng the VIGF polypeptide having the amino acid sequence encoded by the cDNA contained in ATCC Deposit No 7587 4 or a fragment, analog or derivative of said polypeptide
2 The polynucleotide of claim 1, wherein the polynucleotide is DNA
3 The polynucleotide of claim 1, wherein the polynucleotide is RNA
4. The polynucleotide of claim 2, wherein the polynucleotide is genomic DNA
5. The polynucleotide of any one of the preceding claims, wherein the polynucleotide encodes VIGF having the deduced amino acid sequence of Figure 1.
6. The polynucleotide of any one of claims 1 to 4, wherein the polynucleotide encodes the VIGF polypeptide encoded by the cDNA of ATCC Deposit No 75874
7 The polynucleotide of any one of the preceding claims having the coding sequence of VIGF as shown m Figure 1
8 A polynucleotide encoding a polypeptide selected from the group consisting of (a) a polypeptide comprising amino acids 22-184 of SEQ ID NO 2, lhlltL^ru^r 44 I QFnz 071't j " 8 SEP tfog SI}/ ir (b) a polypeptide comprising amino acids 1-184 of SEQ ID NO 2, (c) a polypeptide comprising amino acids 51-64 of SEQ ID NO 2, (d) a polypeptide comprising amino acids 76-90 of SEQ ID NO-2, (e) a polypeptide fragment of SEQ ID NO.2, wherein said fragment has the ability to stimulate the proliferation of endothelial cells in the presence of comitogen Con A, (f) a polypeptide variant of (a), (b), (c), (d) or (e) resulting from conservative substitutions, and (g) an antigenic polypeptide fragment of the polypeptide of (a), (o), (c), (d), (e) or (f).
9. A vector containing a polynucleotide of any one of the previous claims
10 A host cell transfected with the vector of claim 9
11 An isolated DNA molecule hybridizable to a polynucleotide of any one of claims 1 to 8 and encoding a polypeptide having VIGF activity
12 A pharmaceutical composition comprising a polynucleotide or any one of claims 1 to 8 and 11 and a pharmaceutically acceptable carrier, excipient or diluent
13 A process for diagnosing a disease or susceptibility to a disease related to a mutation m VIGF nucleic acid sequence comprising isolating a nucleic acid sequence encoding VIGF from a sample derived from a host and determining a mutation m said VIGF nucleic acid sequence with reference to a polynucleotide of any one of claims 1 to 8 and 11. / 'fJitLLEf/u! L,1'V U,-| Lfc - 4 5 - I OF (v / 8 SEP 1339 | .... RECFn/^o
14. A diagnostic process comprising analysing for the presence of a polynucleotide of any one of claims 1 to 8 and 11 in a sample derived from a host group consisting of (a) a polypeptide comprising amino acids 22-184 of SEQ ID NO 2, (b) a polypeptide comprising amino acids 1-18 4 of SEQ ID NO 2; (c) a polypeptide comprising amino acids 51-64 of SEQ ID NO 2, (d) a polypeptide comprising amino acids 76-90 of SEQ ID NO 2, (e) a polypeptide fragment of SEQ ID NO:2 wherein said fiagment has the ability to stimulate the proliferation of endothelial cells in the presence of comitogen Con A, (c) , (d) or (e) resulting from conservative substitutions, and (g) an antigenic polypeptide fragment of the polypeptide of (a), (b), (c), (d) , (e) or (f)
15 An isolated polypeptide selected from the (f) a polypeptide variant of (a), (b) ,
16. The polypeptide of claim 15 fused to a heterologous polypeptide
17 The isolated polypeptide of claim 15, wherein said polypeptide is (a)
18. The isolated polypeptide of claim 15, wherein said polypeptide is (b)
19. The isolated polypeptide of claim 15, wherein said polypeptide is (c)
20 The isolated polypeptide of claim 15, wherein said polypeptide is (d)
21 The isolated polypeptide of claim 15, wherein said polypeptide is (e) 46
22. The isolated polypeptide of claim 15, wherein said polypeptide is (f)
23 The isolated polypeptide of claim 15, wherein said polypeptide is (g)
24 A pharmaceutical composition comprising an isolated polypeptide of claim 15, together with a pharmaceutical^ acceptable carrier, excipient or diluent.
25 A method of diagnosing a pathological condition of a susceptibility to a pathological condition in a subject related to expression or activity of a protein comprising (a) determining the presence or amount of expression of the polypeptide of claim 15 m a biological sample, (b) diagnosing a pathological condition or a susceptibility to a pathological condition based on the presence or amount of expression of the polypeptide
26 A method for identifying binding partner to the polypeptide of claim 15 comprising (a) contacting the polypeptide of claim 15 with a binding partner, and (b) determining whether the binding partner binds to the polypeptide
27 An isolated polypeptide comprising a member selected from the group consisting of (a) a polypeptide encoded by the human cDNA contained m the ATCC Deposit No 75874; (b) a polypeptide encoded by the human cDNA contained in the ATCC Deposit No 75874, excepting the N-termmal methionine, (c) a polypeptide encoded by a polynucleotide which hybridizes in a solution *8 SfP 199$ C? nrr~ , - rj consisting of 0 2.\SSC, 0 1% SDS at 60°C to the human cDNA contained m the ATCC Deposit No 7587 4, wherein said encoded polypeptide has the ability to stimulate proliferation of endothelial cells in the presence of comitogen Con A, (d) a polypeptide encoded by a polynucleotide which hybridizes m a solution consisting of 0.2xSSC, 0 1% SDS at 60°C to the complement of SEQ ID NO 1, wherein said encoded polypeptide has the ability to stimulate proliferation of endothelial cells m the presence of comitogen Con A; (e) a polypeptide fragment of (a), (b), (c) or (d) wherein said fragment retains the ability to stimulate proliferation of endothelial cells in the presence of comitogen Con A, (f) a polypeptide variant of (a), (b), (c), (d) or (e) resulting from conservative substitutions, and (g) an antigenic polypeptide fragment of the polypeptide of (a), (b) , (c), (d), (e) or (f)
28 The polypeptide of claim 27 fused to a heterologous polypeptide.
29 The isolated polypeptide of claim 27, wherein said polypeptide is (a).
30 The isolated polypeptide of claim 27, wherein said polypeptide is (b)
31 The isolated polypeptide of claim 27, wherein said polypeptide is (c)
32 The isolated polypeptide of claim 27, wherein said polypeptide is (d)
33 The isolated polypeptide of claim 27, wherein said polypeptide is (e) '8 SEP J999 - El
34. The isolated polypeptide of claim 27, wherein said polypeptide is (f)
35 The isolated polypeptide of claim 27, wherein said polypeptide is (g)
36 A pharmaceutical composition comprising an isolated polypeptide of claim 27, together with a pharmaceutically acceptable carrier, excipient or diluent
37 A method of diagnosing a pathological condition or a susceptibility to a pathological condition m a subject related to expression or activity of a protein comprising (a) determining the presence or amount of expression of the polypeptide of claim 27 m a biological sample, (b) diagnosing a pathological condition or a susceptibility to a pathological condition based on the presence or amount of expression of the polypeptide
38 A method of identifying binding partner to the polypeptide of claim 27 comprising* (a) contacting the polypeptide of claim 27 with a binding partner, and (b) determining whether the binding partner binds to the polypeptide
39 An isolated polypeptide selected from the group consisting of- (a) a polypeptide consisting of ammo acids 22-184 of SEQ ID NO 2, (b) a polypeptide consisting of amino acids 1-184 of SEQ ID N0:2, (c) a polypeptide consisting of amino acids 51-64 of SEQ ID N0:2; - 49 - 8 SEP W99 R '<"£lVPn (d) a polypeptide consisting of ammo acids 76-90 of SEQ ID NO 2, (e) a polypeptide variant of (a), (b), (c) or (d) resulting from conservative substitutions, and (f) an antigenic polypeptide fragment of the polypeptide of (a), (b) , (c) or (d)
40 The polypeptide of claim 39 fused to a heterologous polypeptide
41 The isolated polypeptide of claim 39, wherein said polypeptide is (a)
42 The isolated polypeptide of claim 39, wherein said polypeptide is (b)
43 The isolated polypeptide of claim 39, wherein said polypeptide is (c)
44 The isolated polypeptide of claim 39, wherein said polypeptide is (d)
45 The isolated polypeptide of claim 39, wherein said polypeptide is (e)
46 The isolated polypeptide of claim 39, wherein said polypeptide is (f)
47 A pharmaceutical composition comprising an isolated polypeptide of claim 39, together with a pharmaceutically acceptable carrier, excipient or diluent
48 A method of diagnosing a pathological condition or a susceptibility to a pathological condition m a subject related to expression or activity of a protein comprising (a) determining the presence or amount of expression of the polypeptide of claim 39 in a biological sample, (b) diagnosing a pathological condition or a susceptibility to a pathological condition based on 50 " ! OF hjz f ~3 SEP 1999 j the presence or amount of expression of the polypeptide
49 A method for identifying binding partner to the polypeptide of claim 39 comprising (a) contacting the polypeptide of claim 39 with a binding partner, and (b) determining whether the binding partner binds to the polypeptide.
50 A method as claimed m claim 49 wherein the binding partner is an antibody
51 An antibody against a polypeptide claimed m any one of claims 15 to 23
52 An isolated polynucleotide as defined m claim 1 or claim 8 substantially as herein described with reference to any example thereof and with or without reference to the accompanying drawings
53 A vector as claimed m claim 9 substantially as herein described with reference to any example thereof and with or without reference to the accompanying drawings
54 A host cell as claimed m claim 10 substantially as herein described with reference to any example thereof and with or without reference to the accompanying drawings.
55 An isolated DNA molecule as claimed m claim 11 substantially as herein described with reference to any example thereof and with or without reference to the accompanying drawings
56 A pharmaceutical composition as claimed m claim 12 substantially as herein described with reference to any example thereof and with or without reference to the accompanying drawings
57 A process as claimed in claim 13 for diagnosing a disease or susceptibility to a disease related to a mutation m VIGF nucleic acid sequence substantially as herein described with reference to any example thereof and with or without reference to the accompanying drawings
58 A diagnostic process as claimed m claim 14 substantially as herein described with reference to any example thereof and with or without reference to the accompanying drawings
59 An isolated polypeptide as defined m claim 15 substantially as herein described with reference to any example thereof and with or without reference to the accompanying drawings
60 A pharmaceutical composition as claimed m claim 24 substantially as herein described with reference to any example thereof and with or without reference to the accompanying drawings
61 A method as claimed in claim 25 of diagnosing a pathological condition of a susceptibility to a pathological condition in a subject related to expression or activity of a protein substantially as herein described with reference to any example thereof and with or without reference to the accompanying drawings
62 A method as claimed in claim 26 for identifying binding partner to the polypeptide of claim 15 substantially as herein described with reference to any example thereof and with or without reference to the accompanying drawings
63 An isolated polypeptide as defined m claim 27 substantially as herein described with reference to any example thereof and with or without reference to the accompanying drawings
64 A pharmaceutical composition as claimed in claim 36 substantially as herein described with _ 52 - J ''Jn1 -,L of N- -c,nf or,K j " 8 SEP 1399 '—-dF-Ce ivfd reference to any example thereof and with or without reference to the accompanying drawings
65. A method as claimed in claim 37 of diagnosing a pathological condition substantially as herein described with reference to any example thereof and with or without reference to the accompanying drawings.
66. A method as claimed m claim 38 for identifying binding partner to the polypeptide of claim 27 substantially as herein described with reference to any example thereof and with or without reference to the accompanying drawings
67 An isolated polypeptide as defined in claim 39 substantially as herein described with reference to any example thereof and with or without reference to the accompanying drawings
68. A pharmaceutical composition as claimed in claim 47 substantially as herein described with reference to any example thereof and with or without reference to the accompanying drawings
69. A method as claimed in claim 48 of diagnosing a pathological condition substantially as herein described with reference to any example thereof and with or without reference to the accompanying drawings 7 0 A method as claimed in claim 4 9 for identifying binding partner to the polypeptide of claim 39 substantially as herein described with reference to any example thereof and with or without reference to the accompanying drawings 71 An antibody against a polypeptide as claimed m claim 51 substantially as herein described with reference to any example thereof and with or without reference to the accompanying drawings riNitLLEC| _ -Hi ,, Cr-c-CC*C^t<tS^ | By the authorised agents _Kin np CJAIMS8 SEP E39 SJ PA?s& S0N Lsecfivp..
NZ278504A 1994-12-09 1994-12-09 Human vascular ibp-like growth factor polypeptide (vigf) NZ278504A (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
NZ278504A NZ278504A (en) 1994-12-09 1994-12-09 Human vascular ibp-like growth factor polypeptide (vigf)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
NZ278504A NZ278504A (en) 1994-12-09 1994-12-09 Human vascular ibp-like growth factor polypeptide (vigf)
PCT/US1994/014388 WO1996017931A1 (en) 1994-12-09 1994-12-09 Human vascular ibp-like growth factor

Publications (1)

Publication Number Publication Date
NZ278504A true NZ278504A (en) 1999-10-28

Family

ID=22243393

Family Applications (1)

Application Number Title Priority Date Filing Date
NZ278504A NZ278504A (en) 1994-12-09 1994-12-09 Human vascular ibp-like growth factor polypeptide (vigf)

Country Status (5)

Country Link
EP (1) EP0796325A4 (en)
JP (1) JPH10510518A (en)
AU (1) AU710568B2 (en)
NZ (1) NZ278504A (en)
WO (1) WO1996017931A1 (en)

Families Citing this family (7)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6670328B1 (en) * 1997-06-24 2003-12-30 Institut Pasteur De Lille Proteins and peptides derived from protein ESM-1 and their uses in the treatment and diagnosis of diseases linked to leukocyte migration
EP1042674A4 (en) * 1997-10-24 2005-03-23 Human Genome Sciences Inc 148 human secreted proteins
FR2775691B1 (en) * 1998-03-05 2000-12-15 Pasteur Institut SPECIFIC MONOCLONAL ANTIBODIES OF ESM-1 PROTEIN, AND USE OF SUCH ANTIBODIES FOR DETECTION OF ESM-1 PROTEIN
EP2168980A1 (en) 2000-06-23 2010-03-31 Genentech, Inc. Compositions and methods for the diagnosis and treatment of disorders involving angiogensis
EP2792747A1 (en) 2000-06-23 2014-10-22 Genentech, Inc. Compositions and methods for the diagnosis and treatment of disorders involving angiogenesis
FR2816410B1 (en) 2000-11-09 2003-04-18 Pasteur Institut ESM-1 PROTEIN DETECTION KIT AND DETECTION METHOD USING THE SAME
FR2816214B1 (en) 2000-11-09 2005-10-21 Pasteur Institut USE OF ANTAGONISTIC ESM-1 PROTEIN COMPOUND FOR THE MANUFACTURE OF A MEDICAMENT FOR THE PREVENTION AND / OR TREATMENT OF CANCER

Family Cites Families (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
FR2642656B1 (en) * 1989-02-09 1991-11-15 Salomon Sa SKI SAFETY FASTENER FOR HOLDING THE FRONT OF A SHOE MOUNTED ON THE SKI
JPH06503711A (en) * 1990-08-28 1994-04-28 カイロン コーポレイション Novel insulin-like growth factor binding protein (IGFBP-4)
PT98805B (en) * 1990-08-28 1999-01-29 Chiron Corp A method of preparing a novel binding protein to the insulin-like growth factor (IGFBP-4)
WO1992012243A1 (en) * 1991-01-08 1992-07-23 Chiron Corporation New insulin-like growth factor binding protein (igfbp-6)

Also Published As

Publication number Publication date
EP0796325A4 (en) 1999-11-03
JPH10510518A (en) 1998-10-13
EP0796325A1 (en) 1997-09-24
AU710568B2 (en) 1999-09-23
WO1996017931A1 (en) 1996-06-13
AU1401495A (en) 1996-06-26

Similar Documents

Publication Publication Date Title
US5780263A (en) Human CCN-like growth factor
US7115392B2 (en) Polynucleotides encoding human vascular endothelial growth factor 2
US20040086967A1 (en) Human criptin growth factor
US7576189B2 (en) Antibodies to human vascular endothelial growth factor 2 and methods of using the same
US6534630B1 (en) Connective tissue growth factor-2
CA2210444C (en) Keratinocyte growth factor-2
EP0873348A1 (en) Human vascular endothelial growth factor 3
US20090311263A1 (en) Human vascular ibp-like growth factor
US7153827B1 (en) Vascular endothelial growth factor 2 and methods of use
AU710568B2 (en) Human vascular IBP-like growth factor
AU714165B2 (en) Human criptin growth factor
US20020049304A1 (en) Human CCN-like growth factor
US5994302A (en) Human vascular IBP-like growth factor
KR100407087B1 (en) Human Vascular IBP-like Growth Factor
AU716100B2 (en) Human vascular endothelial growth factor 3
AU762694B2 (en) Human criptin growth factor
CA2206640A1 (en) Human vascular ibp-like growth factor
AU2005200574A1 (en) Human Vascular Endothelial Factor 2
JP2002247983A (en) Human vascular ibp-like growth factor
AU1951100A (en) Human vascular endothelial growth factor 2
AU2003252757A1 (en) Human Criptin Growth Factor
CA2413012A1 (en) Human vascular endothelial growth factor 2
AU1541402A (en) Human vascular endothelial growth factor 2
AU2780600A (en) Human vascular endothelial growth factor 3
CA2215286A1 (en) Human criptin growth factor

Legal Events

Date Code Title Description
RENW Renewal (renewal fees accepted)
RENW Renewal (renewal fees accepted)
RENW Renewal (renewal fees accepted)
EXPY Patent expired