WO2005121174A2 - Novel g-csf polypeptides, polynucleotides, modulators thereof, and methods of use - Google Patents

Novel g-csf polypeptides, polynucleotides, modulators thereof, and methods of use Download PDF

Info

Publication number
WO2005121174A2
WO2005121174A2 PCT/US2005/019491 US2005019491W WO2005121174A2 WO 2005121174 A2 WO2005121174 A2 WO 2005121174A2 US 2005019491 W US2005019491 W US 2005019491W WO 2005121174 A2 WO2005121174 A2 WO 2005121174A2
Authority
WO
WIPO (PCT)
Prior art keywords
polypeptide
composition
chosen
nucleic acid
csf
Prior art date
Application number
PCT/US2005/019491
Other languages
French (fr)
Other versions
WO2005121174A3 (en
Inventor
Thomas Linnemann
Keting Chu
Kevin Hestir
Haishan Lin
Lewis T. Williams
Ernestine Lee
Original Assignee
Five Prime Therapeutics, 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 Five Prime Therapeutics, Inc. filed Critical Five Prime Therapeutics, Inc.
Publication of WO2005121174A2 publication Critical patent/WO2005121174A2/en
Publication of WO2005121174A3 publication Critical patent/WO2005121174A3/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
    • C07K14/52Cytokines; Lymphokines; Interferons
    • C07K14/53Colony-stimulating factor [CSF]
    • C07K14/535Granulocyte CSF; Granulocyte-macrophage CSF

Definitions

  • G-CSF granulocyte colony stimulating factor
  • G-CSF granulocyte colony stimulating factor
  • G-CSF is produced by a number of cell types including fibroblasts, macrophages, T-cells, trophoblasts, endothelial cells, and epithelial cells, and is the expression product of a single copy gene with four exons and five introns located on chromosome seventeen.
  • Human G-CSF can be obtained and purified from a number of sources.
  • natural human G-CSF nhG-CSF
  • nhG-CSF natural human G-CSF
  • G-CSF molecule in glycosylated form as a product of eukaryotic host cell expression
  • a G-CSF molecule in non- glycosylated form as a product of prokaryotic host cell expression
  • G-CSF is administered to cancer patients to stimulate neutrophil production in an effort to compensate for hematopoietic deficits resulting from chemotherapy or radiation therapy.
  • the half-life of G- CSF in these patients is relatively short.
  • undesirable side effects remain problematic upon administration of the G-CSF molecule.
  • the reported side effects include mild to moderate bone and/or musculoskeletal pain, anemia, thrombocytopenia, and injection site reactions.
  • some of the reported side effects are more severe and include, for example, spleen enlargement and respiratory disturbances.
  • Figure 1 shows the polypeptide alignment of the novel G-CSF polypeptides of the invention, CLN00489695_5pvl and CLN00493523_5pvl (SEQ. ID. NOS.:9 and 10, respectively) with the amino acid sequences of known variants of G-CSF.
  • Sequence 23331380 is SEQ. ID. NO.: 148 from U.S. Patent No. 6,436,387.
  • Sequence 23331383 is SEQ. ID. NO.:151 from U.S. Patent No. 6,436,387.
  • Sequence 23331390 is SEQ. ID. NO.: 158 from U.S. Patent No. 6,436,387.
  • Sequence 23331381 is SEQ. ID.
  • Sequence 23331388 is SEQ. ID. NO.:156 from U.S. Patent No. 6,436,387.
  • Sequence 23331378 is SEQ. JD. NO.: 146 from U.S. Patent No. 6,436,387.
  • Sequence 23331379 is SEQ. ID. NO.: 147 from U.S. Patent No. 6,436,387.
  • Sequence 23331382 is SEQ. ID. NO.:150 from U.S. Patent No. 6,436,387.
  • Sequence 56626517 is SEQ. ID. NO.:18 from U.S. Patent No. 6,797,493.
  • Figure 2 shows the polypeptide alignment of the novel G-CSF polypeptides of the invention, CLN00489695_5pvl and CLN00493523_5pvl (SEQ. ID. NOS.:9 and 10, respectively) with predicted translations of known variants of G-CSF.
  • NP_000750_NM_000759 is the National Center for Biotechnology Information (NCBI) identification number for human colony stimulating factor 3 isoform a precursor.
  • Sequence 21261230 is a predicted translation of SEQ. ID. NO.:8 from WO 02/20751.
  • Sequence 42285214 is a predicted translation of SEQ. ID. NO.:10291 from WO 02/068579.
  • Sequence 21261234 is a predicted translation of SEQ. ID. NO.:12 from WO 02/20751.
  • Sequence 21261235 is a predicted translation of SEQ. ID. NO.:13 from WO 02/20751.
  • Sequence 21261226 is a predicted translation of SEQ. ID. NO.:4 from WO 02/20751.
  • Sequence 53922219 is a predicted translation of SEQ. ID. NO.:95 from U.S Patent No. 6,730,303.
  • Sequence 21261228 is a predicted translation of SEQ. ID. NO.:6 from WO 02/20751.
  • Sequence 21261237 is a predicted translation of SEQ.
  • Sequence 21261227 is a predicted translation of SEQ. ID. NO.:5 from WO 02/20751.
  • Sequence 53922218 is apredicted translation of SEQ. ID. NO.:94 from U.S. Patent No. 6,730,303.
  • Sequence 21261229 is a predicted translation of SEQ. ID. NO.:7 from WO 02/20751.
  • Sequence 3977652 is a predicted translation of SEQ. ID. NO.:l from U.S. Patent No. 5,795,968.
  • Sequence 6088626 is SEQ. ID. NO.:2 from WO 93/15211.
  • Figure 3 [009] Figure 3 shows the NMR structure of the G-CSF molecule (FEBS Lett.
  • Figure 4 compares the amino acid sequences of human and mouse G-CSF.
  • Table 1 shows the correlation between the Sequence Listing and the G-CSF molecules discussed herein.
  • Column 1 shows an internal designation identification number (FP ID).
  • Column 2 shows the nucleotide sequence identification number for the open reading frame of the nucleic acid sequence (SEQ. ID. NO.:Nl).
  • Column 3 shows the amino acid sequence identification number for the polypeptide sequence (SEQ. ID. NO..P1).
  • Column 4 shows the nucleotide sequence identification number for the entire nucleic acid sequence, including both coding and non-coding sequences (SEQ. ID. NO.:N0).
  • Column 5 shows the polypeptide identification number of the source clone or sequence (Source ID).
  • Table 2 [012] Table 2 shows a publicly available annotation of the polypeptide sequences of the Sequence Listing.
  • Column 1 shows an internal designation identification number of the polypeptide (FP TD).
  • Column 2 shows the source identification number of the polypeptide (Source ID).
  • Column 3 shows the predicted length of the polypeptide (Pred Prot Len).
  • Columns 4 and 5 describe the characteristics of the human protein in the NCBI database with the greatest degree of similarity to polypeptides encoded by each FP JD. The NCBI protein is described by its accession number (Top Human Hit Accession ID) and by the NCBI's annotation of that sequence (Top Human Hit Annot).
  • Column 6 shows the length of the polypeptide sequence set forth in column 4 (Top Human Hit Len).
  • Column 7 shows the number of matching amino acid residues between the FP ID and the Top Human Hit sequences (Match Len).
  • Table 3 shows characteristics of the polypeptides of the Sequence Listing.
  • Column 1 shows an internal designation identification number of the polypeptide (FP ID).
  • Column 2 shows the source identification number of the polypeptide (Source ID).
  • Column 3 shows the cluster identification number of polypeptides HG1015530, HG1015531, and HG1015532 (Cluster).
  • Column 4 shows the classification of the polypeptide (Class), designating that cluster 16616 is secreted.
  • Column 5 shows the predicted protein length (Pred Prot Len).
  • Column 6 shows an internal parameter predicting the likelihood that the FP ID is secreted (Treevote), with "1" being a high likelihood that the polypeptide is secreted and "0" being a low likelihood that the polypeptide is secreted.
  • Column 1 shows an internal designation identification number of the polypeptide (FP ID).
  • Column 2 shows the source identification number of the polypeptide (Source ID).
  • Column 3 shows the predicted protein length (Pred Prot Len).
  • Column 4 shows the length of the match between the FP ID and the Source ID.
  • Column 5 shows the percent identity between the FP ID and the Source ID over the length of the FP ID.
  • Column 6 shows the percent identity between the FP ID and the Source ID over the length of Source ID.
  • the polypeptide and modulator compositions and methods of the invention are useful in the diagnosis, treatment, and/or prevention of neutropenia, including chronic and chemotherapy-associated neutropenia, mobilization of bone marrow stem cells, treatment of inflammatory, immune, and ischemic diseases, treatment of bacterial and fungal infections, and the enhancement of myocardial regeneration without aggravated inflammation in the infarcted heart.
  • DESCRIPTION OF THE INVENTION Summary of the Invention [016] The invention provides a first nucleic acid molecule comprising a first polynucleotide sequence chosen from SEQ. ID.
  • nucleic acid molecule is an isolated molecule.
  • This first nucleic acid molecule may be a cDNA molecule, a genomic DNA molecule, a cRNA molecule, a siRNA molecule, a RNAi molecule, or an mRNA molecule.
  • the invention provides a double-stranded isolated nucleic acid molecule comprising this first nucleic acid molecule and its complement.
  • the invention provides a second nucleic acid molecule comprising a second polynucleotide sequence complementary to the first nucleic acid molecule.
  • the second nucleic acid molecule may be, for example, a RNAi molecule, an anti-sense molecule, or a ribozyme.
  • the invention also provides an isolated polypeptide comprising an amino acid sequence chosen from SEQ. ID. NOS.:9-10 and active fragments of either of these. This polypeptide may be present in a cell culture, for example a bacterial cell culture, a mammalian cell culture, or an insect cell culture. It may be encoded by the first nucleic acid molecule.
  • This polypeptide may further comprise at least one fusion partner, for example, a polymer, a polypeptide, a succinyl group, or serum albumin.
  • a suitable polymer is either a branched or linear chain polyethylene glycol moiety, for example, one attached through an amino group of an amino acid of the polypeptide.
  • the invention further provides a vector comprising the first nucleic acid molecule and a promoter that regulates its expression.
  • the promoter may or may not be naturally contiguous to the first nucleic acid molecule. It may be an inducible promoter, a conditionally-active promoter, for example the cre-lox promoter, a constitutive promoter, or a tissue-specific promoter.
  • the invention yet further provides a recombinant host cell comprising the first nucleic acid molecule or a polypeptide encoded therein, an isolated polypeptide comprising an amino acid sequence chosen from SEQ. ID. NOS.: 9- 10 and active fragments of either of these, and/or a vector comprising the first nucleic acid molecule and a promoter that regulates its expression.
  • the host cell may be a prokaryotic cell. It may also be a eukaryotic cell, for example, a human cell, a non-human mammalian cell, an insect cell, a fish cell, a plant cell, or a fungal cell.
  • the invention provides a non-human animal injected with the first nucleic acid molecule. It also provides a non-human animal transformed with the first nucleic acid molecule or a polypeptide encoded thereby. It further provides a non-human animal injected with an isolated polypeptide comprising an amino acid sequence chosen from SEQ. ID. NOS.:9-10 and active fragments of either of these. [020]
  • the invention also provides nucleic acid compositions, polypeptide compositions, vector compositions, and host cell compositions comprising the nucleic acid molecules, polypeptides, vectors, and host cells of the invention and a carrier, for example, a pharmaceutically acceptable carrier or excipient.
  • the invention provides a method of producing a recombinant host cell comprising providing a composition comprising a vector that comprises the first nucleic acid molecule and allowing the host cell to come into contact with the vector to form a recombinant host cell.
  • the invention provides a method of producing a polypeptide comprising providing a composition comprising a recombinant host cell of the invention and culturing the recombinant host cell to produce the polypeptide.
  • the invention also provides a method of producing a polypeptide comprising providing the first nucleic acid and expressing it in a cell free expression system to produce the polypeptide. Suitable cell free expression systems include a wheat germ lysate expression system, a rabbit reticulocyte expression system, and an E. coli lysate expression system.
  • the invention also provides a diagnostic kit comprising a composition comprising a polynucleotide molecule complementary to the first nucleic acid molecule and a vehicle.
  • Kits of the invention may comprise an antibody that specifically binds to the first nucleic acid molecule or a polypeptide encoded thereby.
  • Kits of the invention may comprise an antibody that specifically binds to an isolated polypeptide comprising an amino acid sequence chosen from SEQ. ID. NOS.: 9- 10 and active fragments of either of these.
  • Kits of the invention may be diagnostic. They may include instructions for use. [024]
  • the invention provides a method of determining the presence of the first nucleic acid molecule or its complement comprising providing a complement to the first nucleic acid molecule or the complement of the first nucleic acid molecule, allowing the molecules to interact, and determining whether interaction has occurred.
  • the invention provides a method of determining the presence of an antibody specific to a polypeptide encoded by the first nucleic acid molecule or an isolated polypeptide comprising an amino acid sequence chosen from SEQ. ID. NOS.:9-10 and active fragments of either of these in a sample comprising providing a composition comprising the polypeptide, allowing the polypeptide to interact with the sample, and determining whether interaction has occurred between the polypeptide and the antibody. It also provides an antibody that specifically binds to or interferes with the activity of one or more of these polypeptides.
  • This antibody may be a polyclonal antibody, a monoclonal antibody, a single chain antibody, or an active fragments of any of these.
  • the antibody may be an antigen binding fragment, an Fc fragment, a cdr fragment, or a framework fragment.
  • the invention provides a method for treating or preventing disease in a subject comprising providing a composition comprising a polypeptide chosen from SEQ. DD. NOS.:9-10 and active fragments of either of these, and a carrier; and administering the composition to the subject.
  • Polypeptides encoded by a nucleic acid molecule comprising a nucleotide sequence chosen from SEQ. ID. NOS.: 1-2, 19-20, and active fragments thereof are also suitable for practicing the invention.
  • the polypeptides used in treating or preventing disease may comprise at least one fusion partner. Suitable fusion partners include, for example, polymers, polypeptides, succinyl groups, and serum albumin.
  • the polymer may comprise either a branched or linear chain polyethylene glycol moiety, which may, for example, be attached through an amino group of an amino acid of the polypeptide.
  • the method can be used to treat or prevent neutropenia, for example, neutropenia induced by chemotherapy or radiotherapy.
  • the method can be used to treat or prevent infection, for example, a bacterial, fungal, parasitic, or viral infection; this method may further comprise administering granulocyte transfusion therapy.
  • the method can be used to treat or prevent septic shock. It can be used to treat or prevent an inflammatory disease, for example, rheumatoid arthritis, myocarditis, inflammatory bowel disease, psoriasis, multiple sclerosis, systemic lupus erythematosus, intraocular inflammation, surgery, injury related inflammation, osteoarthritis, or central nervous system inflammation.
  • ocular inflammation for example, scleritis, orbital pseudotumor, corneal melt, ocular cicatricial pemphigoid, or Sjogren's syndrome.
  • the method can be used to treat or prevent ischemic diseases, for example, stroke, myocardial infarction, and fulminant liver failure.
  • ischemic diseases for example, stroke, myocardial infarction, and fulminant liver failure.
  • immune diseases for example, B-lymphocytic disease. It can be used to suppress immune activity.
  • This method can be practiced with a polypeptide that further comprises at least one fusion partner, for example, a polymer, for example, a polyethylene glycol moiety; a polypeptide; a succinyl group; or serum albumin.
  • the polyethylene glycol moiety may be attached through an amino group of an amino acid of the polypeptide. It may be either a branched or linear chain polymer.
  • the method may include administering the composition to the subject locally or systemically.
  • the invention provides a method for mobilizing peripheral blood stem cells and/or bone marrow stem cells in a subject comprising providing a composition comprising a polypeptide chosen from SEQ. TD.
  • the invention also provides a method for modulator screening comprising providing a polypeptide chosen from an isolated polypeptide comprising an amino acid sequence chosen from SEQ. ID. NOS.: 9- 10 and active fragments of either of these, or an isolated polypeptide encoded by the first nucleic acid molecule, and using the polypeptide as a target in an assay that detects the modulator.
  • This assay can detect modulators which are, for example, small molecule drugs or antibodies.
  • the invention provides a polypeptide wherein at least one cysteine residue is replaced by a polar amino acid, a basic amino acid, an acidic amino acid, and/or a neutral amino acid, for example, serine or alanine.
  • This polypeptide may further comprise at least one fusion partner. It may further comprise a growth factor, for example, GM-CSF or M-CSF.
  • polynucleotide refers to polymeric forms of nucleotides of any length.
  • the polynucleotides can contain deoxyribonucleotides, ribonucleotides, and/or their analogs or derivatives.
  • a "complement" of a nucleic acid molecule is a one that is comprised of its complementary base pairs.
  • Deoxyribonucleotides with the base adenine are complementary to those with the base thymidine, and deoxyribonucleotides with the base thymidine are complementary to those with the base adenine.
  • Deoxyribonucleotides with the base cytosine are complementary to those with the base guanine, and deoxyribonucleotides with the base guanine are complementary to those with the base cytosine.
  • Ribonucleotides with the base adenine are complementary to those with the base uracil, and deoxyribonucleotides with the base uracil are complementary to those with the base adenine.
  • Ribonucleotides with the base cytosine are complementary to those with the base guanine, and deoxyribonucleotides with the base guanine are complementary to those with the base cytosine.
  • a "vector” is a plasmid that can be used to transfer DNA sequences from one organism to another or to express a gene of interest.
  • a "promoter,” as used herein, is a DNA regulatory region capable of binding
  • RNA polymerase in a mammalian cell and initiating transcription of a downstream (3' direction) coding sequence operably linked thereto.
  • a promoter sequence includes the minimum number of bases or elements necessary to initiate transcription of a gene of interest at levels detectable above background. Within the promoter sequence is a transcription initiation site, as well as protein binding domains (consensus sequences) responsible for the binding of RNA polymerase. Eucaryotic promoters will often, but not always, contain "TATA" boxes and "CAT” boxes. Promoters include those that are naturally contiguous to a nucleic acid molecule and those that are not naturally contiguous to a nucleic acid molecule.
  • a promoter includes inducible promoters, conditionally active promoters, such as a cre-lox promoter, constitutive promoters, and tissue specific promoters.
  • "Expression of a nucleic acid molecule” refers to the conversion of the information contained in the molecule, into a gene product.
  • a gene product can be the direct transcriptional product of a gene (e.g., mRNA, tRNA, rRNA, antisense RNA, ribozyme, structural RNA, or any other type of RNA) or a peptide or polypeptide produced by translation of an mRNA.
  • Gene products also include RNAs which are modified, by processes such as capping, polyadenylation, methylation, and editing, and proteins modified by, for example, methylation, acetylation, phosphorylation, ubiquitination, ADP-ribosylation, myristilation, and glycosylation.
  • polypeptide refers to a polymeric form of amino acids of any length, which can include naturally-occurring amino acids, coded and non-coded amino acids, chemically or biochemically modified, derivatized, or designer amino acids, amino acid analogs, peptidomimetics, and depsipeptides, and polypeptides having modified, cyclic, bicyclic, depsicyclic, or depsibicyclic peptide backbones.
  • the term includes single chain protein as well as multimers.
  • the term also includes conjugated proteins, fusion proteins, including, but not limited to, glutathione S-transferase (GST) fusion proteins, fusion proteins with a heterologous amino acid sequence, fusion proteins with heterologous and homologous leader sequences, fusion proteins with or without N-terminal methionine residues, pegolyated proteins, and immunologically tagged, or his-tagged proteins.
  • GST glutathione S-transferase
  • the term also includes peptide aptamers.
  • a "pfam domain” is a protein or a portion of a protein with a tertiary structure. Pfams may have characteristic functional activities, such as enzymatic or binding activities. Multiple pfam domains can be connected by flexible polypeptide regions within a protein, pfam domains can comprise the N-terminus or the C-terminus of a protein, or can be situated at any point between.
  • a "non-transmembrane domain” is a portion of a transmembrane protein that does not span the membrane. It may be extracellular, cytoplasmic, or luminal.
  • An “isolated,” “purified,” “substantially isolated,” or “substantially purified” molecule is one that has been manipulated to exist in a higher concentration than in nature. For example, a subject antibody is isolated, purified, substantially isolated, or substantially purified when at least 10%, or 20%, or 40%, or 50%, or 70%, or 90% of non-subject-antibody materials with which it is associated in nature have been removed.
  • an “isolated,” “purified,” “substantially isolated,” or “substantially purified” molecule includes recombinant molecules.
  • fragment is intended a polynucleotide or polypeptide consisting of only a part of the intact full-length or naturally occurring polynucleotide or polypeptide sequence and structure.
  • a fragment may comprise or encode any portion of the molecules described herein.
  • a fragment may comprise or encode the amino acids at the splice sites of SEQ. ID. NOS.:l, 2, 7, 8, 9, 10, 17, 18, 19 and 20.
  • a polypeptide fragment can include e.g., a C-terminal deletion, an N-terminal deletion, and/or an internal deletion of a native polypeptide or an extracellular domain of a transmembrane protein.
  • a “biologically active” entity, or an entity having “biological activity,” is one or more entity having structural, regulatory, or biochemical functions of a naturally occurring molecule or any function related to or associated with a metabolic or physiological process. Biological activity can be determined by protein folding or protein conformation. Biologically active polynucleotide fragments are those exhibiting activity similar, but not necessarily identical, to an activity of a polynucleotide of the present invention. The biological activity can include an improved desired activity, or a decreased undesirable activity.
  • an entity demonstrates biological activity when it participates in a molecular interaction with another molecule, such as hybridization, when it has therapeutic value in alleviating a disease condition, when it has prophylactic value in inducing an immune response, when it has diagnostic value in determining the presence of a molecule, such as a biologically active fragment of a polynucleotide that can, for example, be detected as unique for the polynucleotide molecule, or that can be used as a primer in a polymerase chain reaction.
  • a molecule such as a biologically active fragment of a polynucleotide that can, for example, be detected as unique for the polynucleotide molecule, or that can be used as a primer in a polymerase chain reaction.
  • a biologically active polypeptide or fragment thereof includes one that can participate in a biological reaction, for example, one that can serve as an epitope or immunogen to stimulate an immune response, such as production of antibodies, or that can participate in stimulating or inhibiting signal transduction by binding to ligands receptors or other proteins, or nucleic acids; or activating enzymes or substrates.
  • antibody and "immunoglobulin” refer to a protein, for example, one generated by the immune system, synthetically, or recombinantly, that is capable of recognizing and binding to a specific antigen; antibodies are commonly known in the art. Antibodies may recognize polypeptide or polynucleotide antigens.
  • the term includes active fragments, including for example, an antigen binding fragment of an immunoglobulin, a variable and/or constant region of a heavy chain, a variable and/or constant region of a light chain, a complementarity determining region (cdr), and a framework region.
  • a "host cell” is an individual cell or cell culture which can be or has been a recipient of any recombinant vector(s) or isolated polynucleotide.
  • Host cells include progeny of a single host cell, and the progeny may not necessarily be completely identical (in morphology or in total DNA complement) to the original parent cell due to natural, accidental, or deliberate mutation and/or change.
  • a host cell includes cells transfected or infected in vivo or in vitro with a recombinant vector or a polynucleotide of the invention.
  • a host cell which comprises a recombinant vector of the invention may be called a "recombinant host cell.”
  • a “stem cell” is a pluripotent or multipotent cell with the ability to self- renew, to remain undifferentiated, and to become differentiated.
  • a stem cell can divide without limit, for at least the lifetime of the animal in which it naturally resides.
  • a stem cell is not terminally differentiated; it is not at the end stage of a differentiation pathway. When a stem cell divides, each daughter cell can either remain a stem cell or embark on a course that leads toward terminal differentiation.
  • Subject “individual,” “host,” and “patient” are used interchangeably herein to refer to mammals, including, but not limited to, rodents, simians, humans, felines, canines, equines, bovines, porcines, ovines, caprines, mammalian laboratory animals, mammalian farm animals, mammalian sport animals, and mammalian pets.
  • modulate refers to the production, either directly or indirectly, of an increase or a decrease, a stimulation, inhibition, interference, or blockage in a measured activity when compared to a suitable control.
  • a “modulator” of a polypeptide or polynucleotide or an “agent” are terms used interchangeably herein to refer to a substance that affects, for example, increases, decreases, stimulates, inhibits, interferes with, or blocks a measured activity of the polypeptide or polynucleotide, when compared to a suitable control.
  • Modulating a level of an active subject polypeptide includes increasing or decreasing, blocking, or interfering with the expression or activity of a subject polypeptide, increasing or decreasing a level of an active polypeptide, and increasing or decreasing the level of mRNA encoding an active subject polypeptide. Modulation can occur directly or indirectly.
  • Disease refers to any condition, infection, disorder, or syndrome that requires medical intervention or for which medical intervention is desirable. Such medical intervention can include treatment, diagnosis, and/or prevention.
  • Treatment covers any administration or application of remedies for disease in a mammal, including a human, and includes inhibiting the disease, i.e., arresting its development, or relieving the disease, i.e., causing regression, or restoring or repairing a lost, missing, or defective function; or stimulating an inefficient process.
  • Preventing includes providing prophylaxis with respect to the occurrence or recurrence of a disease in a subject that may be predisposed to the disease but has not yet been diagnosed with the disease.
  • Treatment and prophylaxis can be administered to an organism, including a human, or to a cell in vivo, in vitro, or ex vivo, and the cell subsequently administered to the subject.
  • compositions refers to a composition that usually contains a carrier, such as a pharmaceutically acceptable carrier or excipient that is conventional in the art and which is suitable for administration into a subject for therapeutic, diagnostic, or prophylactic purposes. It may include a cell culture in which the polypeptide or polynucleotide is present in the cells or in the culture medium.
  • a carrier such as a pharmaceutically acceptable carrier or excipient that is conventional in the art and which is suitable for administration into a subject for therapeutic, diagnostic, or prophylactic purposes. It may include a cell culture in which the polypeptide or polynucleotide is present in the cells or in the culture medium.
  • compositions for oral administration can form solutions, suspensions, tablets, pills, capsules, sustained release formulations, oral rinses, or powders.
  • a "pharmaceutically acceptable carrier” refers to a non-toxic solid, semisolid or liquid filler, diluent, encapsulating material, formulation auxiliary, or excipient of any conventional type.
  • a pharmaceutically acceptable carrier is non-toxic to recipients at the dosages and concentrations employed and is compatible with other ingredients of the formulation.
  • Nucleic Acids and Polypeptides [055] The invention provides two novel G-CSF molecules.
  • the wild-type G-CSF polypeptide structure includes a four helix bundle (FEBS Lett., 314(3):435-439 (1992)), wherein the first two helices, known as helices A and B, are connected via a longer loop containing a short helix E, which allows helix A and B to align in a parallel manner ( Figure 3A).
  • the first G-CSF polypeptide described herein, CLN00489695 lacks the connective helix E and a portion of helix A ( Figure 4A).
  • the second G-CSF polypeptide molecule, CLN004493523 lacks a portion of helix B ( Figure 4B).
  • the invention provides nucleic acid molecules containing a polynucleotide encoding these two newly identified G-CSF variant polypeptides, which have the amino acid sequences shown in the Sequence Listing (SEQ. ID. NOS.:9-10).
  • Fragments of the full length G-CSF variants may be used as hybridization probes for cDNA libraries to isolate the full length gene and to isolate other genes which have a high sequence similarity or a similar biological activity.
  • Probes of this type can have at least 30 bases and may comprise, for example, 50 or more bases.
  • the probe may also be used in a screening procedure to identify a cDNA clone corresponding to a full length transcript and a genomic clone or clones that contain complete G-CSF genes, including regulatory and promoter regions, exons, and introns.
  • An example of such a screen would include isolating the coding regions of G-CSF genes by using a known nucleic acid sequence to synthesize an oligonucleotide probe.
  • Labeled oligonucleotides having a sequence complementary to a gene of the present invention can be used to screen a human cDNA, a genomic DNA, or a mRNA library to identify complementary library components.
  • the present invention further relates to polynucleotides which hybridize to the described sequences if there is at least 91%, at least 92%, or at least 95% identity between the sequences.
  • the present invention relates to polynucleotides which hybridize under stringent conditions to the described polynucleotides. Stringent conditions generally include condition under which hybridization will occur only if there is at least 95%, or at least 97% identity between the sequences.
  • polynucleotides which hybridize to the hereinabove described polynucleotides encode polypeptides which may retain substantially the same biological function or activity as the mature polypeptide.
  • the polynucleotide may have at least 20 bases, at least 30 bases, or at least 50 bases which hybridize to a polynucleotide of the present invention and which has an identity thereto, as hereinabove described, and which may or may not retain activity.
  • the present invention is directed to polynucleotides having at least a
  • nucleic acid molecules of the present invention encoding a G-CSF polypeptide may be obtained using standard cloning and screening procedures, such as those for cloning cDNAs using mRNA as starting material.
  • Nucleic acids of the invention are useful as hybridization probes for differential identification of the tissue(s) or cell type(s) present in a biological sample.
  • Polypeptides and antibodies directed to those polypeptides are useful for providing immunological probes for the differential identification of tissues or cell types.
  • the present invention also relates to vectors which include the isolated nucleic acid molecules of the present invention, host cells which are genetically engineered with the recombinant vectors, and the production of G-CSF polypeptides or fragments thereof by recombinant techniques.
  • the vector may be, for example, a phage, plasmid, viral, or retroviral vector. Retroviral vectors may be replication competent or replication defective. In the latter case, viral propagation generally will occur only in complementing host cells.
  • the present invention provides recombinant vectors that contain, for example, nucleic acid constructs that encode secretory leader sequences and a selected heterologous polypeptide of interest, and host cells that are genetically engineered with the recombinant vectors.
  • Selected heterologous polypeptides of interest in the present invention include, for example, an extracellular fragment of a secreted protein, a type I membrane protein, a type II membrane protein, a multi-membrane protein, and a soluble receptor.
  • These vectors and host cells can be used for the production of polypeptides described herein, including fragments thereof by conventional recombinant techniques.
  • the vector may be, for example, a phage, plasmid, viral or retroviral vector.
  • Retroviral vectors may be replication competent or replication defective. As above, in the latter case, viral propagation generally will occur only in complementing host cells.
  • the polynucleotides may be joined to a vector containing a secretory leader sequence and a selectable marker for propagation in a host.
  • a. plasmid vector is introduced in a precipitate, such as a calcium phosphate precipitate, or in a complex with a charged lipid. If the vector is a virus, it may be packaged in vitro using an appropriate packaging cell line and then transduced into host cells.
  • the polynucleotides may be joined to a vector containing a selectable marker for propagation in a host.
  • a plasmid vector is introduced in a precipitate, such as a calcium phosphate precipitate, or in a complex with a charged lipid.
  • a virus If the vector is a virus, it may be packaged in vitro using an appropriate packaging cell line and then transduced into host cells.
  • the DNA insert can be operatively linked to an appropriate promoter, such as the phage lambda PL promoter; the E. coli lac, t ⁇ , phoA and tac promoters; the SV40 early and late promoters; and promoters of retroviral LTRs, to name a few.
  • an appropriate promoter such as the phage lambda PL promoter; the E. coli lac, t ⁇ , phoA and tac promoters; the SV40 early and late promoters; and promoters of retroviral LTRs, to name a few.
  • Other suitable promoters will be known to the skilled artisan.
  • the expression constructs will further contain sites for transcription initiation, termination, and, in the transcribed region, a ribosome binding site for translation.
  • the coding portion of the transcripts expressed by the constructs can include a translation initiating codon at the beginning and a termination codon (UAA, UGA, or UAG) appropriately positioned at the end of the polypeptide to be translated.
  • the expression vectors may include at least one selectable marker.
  • markers include dihydrofolate reductase, G418 or neomycin resistance for eukaryotic cell culture and tetracycline, kanamycin or ampicillin resistance genes for culturing in E. coli and other bacteria.
  • Representative examples of appropriate hosts include, but are not limited to, bacterial cells, such as E. coli, Streptomyces and Salmonella typhimurium cells; fungal cells, such as yeast cells; insect cells such as Drosophila S2 and Spodoptera Sf9 cells; animal cells such as CHO, COS, 293, and Bowes melanoma cells; and plant cells. Appropriate culture mediums and conditions for the above-described host cells are known in the art.
  • the selectable markers are genes that confer a phenotype on a cell expressing the marker, so that the cell can be identified under appropriate conditions.
  • a selectable marker allows the selection of transformed cells based on their ability to thrive in the presence or absence of a chemical or other agent that inhibits an essential cell function.
  • Suitable markers therefore, include genes coding for proteins which confer drug resistance or sensitivity thereto, impart color to, or change the antigenic characteristics of those cells transfected with a molecule encoding the selectable marker, when the cells are grown in an appropriate selective medium.
  • selectable markers include cytotoxic markers and drug resistance markers, whereby cells are selected by their ability to grow on media containing one or more of the cytotoxins or drugs; auxotrophic markers by which cells are selected for their ability to grow on defined media with or without particular nutrients or supplements, such as thymidine and hypoxanthine; metabolic markers for which cells are selected, e.g., their ability to grow on defined media containing the appropriate sugar as the sole carbon source, and markers which confer the ability of cells to form colored colonies on chromogenic substrates or cause cells to fluoresce.
  • cytotoxic markers and drug resistance markers whereby cells are selected by their ability to grow on media containing one or more of the cytotoxins or drugs
  • auxotrophic markers by which cells are selected for their ability to grow on defined media with or without particular nutrients or supplements, such as thymidine and hypoxanthine
  • metabolic markers for which cells are selected, e.g., their ability to grow on defined media containing the appropriate sugar as the sole carbon source, and markers which confer the ability of cells
  • vectors suitable for use in bacteria include pQE70, pQE60, and pQE-
  • pBS vectors Phagescript vectors, Bluescript vectors, pNH8A, pNH6a, pNH18A, pNH46A, available from Stratagene (La Jolla, CA); and ptrc99a, pKK223-3, pKK233-3, pDR540, pRIT5 available from Pharmacia (Peapack, NJ).
  • suitable eukaryotic vectors are pWLNEO, pSV2CAT, pOG44, pXTl, and pSG available from Stratagene; and pSVK3, pBPV, pMSG and pSVL, available from Pharmacia.
  • Suitable viral vectors include the retroviral LTR; the S V40 promoter; and the human cytomegalovirus (CMV) promoter described in Miller, et al., Biotechniques, Vol. 7, No. 9, 980-990 (1989), or any other homologous or heterologous promoter, for example, cellular promoters such as eukaryotic cellular promoters including, but not limited to, the histone, pol HI, ⁇ -actin promoters, adenovirus promoters, e.g., the adenoviral major late promoter; thymidine kinase (TK) promoters; and B19 parvo virus promoters.
  • cellular promoters such as eukaryotic cellular promoters including, but not limited to, the histone, pol HI, ⁇ -actin promoters, adenovirus promoters, e.g., the adenoviral major late promoter; thymidine kinase
  • Suitable vectors include those employing a pTT vector backbone
  • the pTT vector backbone may be prepared by obtaining pIRESpuro/EGFP (pEGFP) and pSEAP basic vector(s), for example from Clontech (Palo Alto, CA), and pcDNA3.1, ⁇ CDNA3.1/Myc-(His) 6 and pCEP4 vectors can be obtained from, for example, Invitrogen.
  • pEGFP pIRESpuro/EGFP
  • pSEAP basic vector(s) for example from Clontech (Palo Alto, CA)
  • pcDNA3.1, ⁇ CDNA3.1/Myc-(His) 6 and pCEP4 vectors can be obtained from, for example, Invitrogen.
  • SuperGlo GFP variant sgGFP
  • Q-Biogene Carlsbad, CA.
  • Preparing a pCEP5 vector can be accomplished by removing the CMV promoter and polyadenylation signal of pCEP4 by sequential digestion and self-ligation using Sail and Xbal enzymes resulting in plasmid pCEP4 ⁇ .
  • a GblH fragment from pAdCMV5 Massie et al., J. Virol, 72: 2289- 2296 (1998)), encoding the CMV5-poly(A) expression cassette may be ligated in BglH- linearized ⁇ CEP4 ⁇ , resulting in pCEP5 vector.
  • the pTT vector can be prepared by deleting the hygromycin (55 l and Sail excision followed by fill-in and ligation) and EBNA1 (Clal and Nsi ⁇ excision followed by fill-in and ligation) expression cassettes.
  • the ColEI origin Fspl-Sall fragment, including the 3' end of ⁇ -lactamase ORF
  • Fspl-Sall fragment from pcDNA3.1 containing the pMBI origin (and the same 3' end of ⁇ -lactamase ORF).
  • a Myc-(His) 6 C-terminal fusion tag can be added to SEAP (Hind ⁇ -Hpa ⁇ fragment from pSEAP-basic) following in-frame ligation in pcDNA3.1/Myc-His digested with HindJI ⁇ and EcoRV. Plasmids can subsequently be amplified in E. coli (DH5 ⁇ ) grown in LB medium and purified using MAXI prep columns (Qiagen, Mississauga, Ontario, Canada). To quantify, plasmids can be subsequently diluted in 50 mM Tris-HCl pH 7.4 and absorbencies can be measured at 260 nm and 280 nm. Plasmid preparations with A 6 o/A 28 o ratios between about 1.75 and about 2.00 are suitable.
  • Introduction of the construct into the host cell can be effected by calcium phosphate transfection, D ⁇ A ⁇ -dextran mediated transfection, cationic lipid-mediated transfection, electroporation, transduction, infection, or other methods. Such methods are described in many standard laboratory manuals, such as Davis et al., Basic Methods In Molecular Biology (1986).
  • the polypeptides may be expressed in a modified form, such as a fusion protein, and may include not only secretion signals, but also additional heterologous functional regions. For instance, a region of additional amino acids, particularly charged amino acids, may be added to the N-terminus of the polypeptide to improve stability and persistence in the host cell, during purification, or during subsequent handling and storage. Also, peptide moieties may be added to the polypeptide to facilitate purification. Such regions may be removed prior to final preparation of the polypeptide. [072] The addition of peptide moieties to polypeptides to engender secretion or excretion, to improve stability and to facilitate purification, among others, are familiar and routine techniques in the art.
  • a suitable fusion protein may comprise a heterologous region from immunoglobulin that is useful to stabilize and purify proteins.
  • ⁇ P-A-O 464 533 (Canadian counterpart 2045869) discloses fusion proteins containing various portions of constant region of immunoglobulin molecules together with another human protein or part thereof.
  • the Fc part in a fusion protein is thoroughly advantageous for use in therapy and diagnosis and thus results, for example, in improved pharmacokinetic properties ( ⁇ P-A 0232262).
  • Fc portion proves to be a hindrance to use in therapy and diagnosis, for example when the fusion protein is to be used as an antigen for immunizations.
  • human proteins such as hIL-5
  • Fc portions have been fused with Fc portions for the purpose of high-throughput screening assays to identify antagonists. See, Bennett et al., J. Molec. Recog., 8:52-58 (1995) and Johanson et al, J. Biol Chem., 270:9459-9471 (1995).
  • the G-CSF polypeptides can be recovered and purified from recombinant cell cultures by well-known methods, including ammonium sulfate or ethanol precipitation, acid extraction, anion or cation exchange chromatography, phosphocellulose chromatography, hydrophobic interaction chromatography, affinity chromatography, hydroxylapatite chromatography, and lectin chromatography.
  • High performance liquid chromatography (HPLC) can be employed for purification.
  • Polypeptides of the present invention include products purified from natural sources, including bodily fluids, tissues and cells, whether directly isolated or cultured; products of chemical synthetic procedures; and products produced by recombinant techniques from a prokaryotic or eukaryotic host, including, for example, bacterial, yeast, higher plant, insect, and mammalian cells. Depending upon the host employed in a recombinant production procedure, the polypeptides of the present invention may be glycosylated or may be non-glycosylated. In addition, polypeptides of the invention may also include an initial modified methionine residue, in some cases as a result of host-mediated processes.
  • N-terminal methionine encoded by the translation initiation codon generally is removed with high efficiency from any protein after translation in eukaryotic cells. While the N-terminal methionine on most proteins also is efficiently removed in most prokaryotes, for some proteins this prokaryotic removal process is inefficient, depending on the nature of the amino acid to which the N- terminal methionine is covalently linked.
  • a heterologous polypeptide may be expressed as described above, or as a fusion protein, and may include not only secretion signals, but also a secretory leader sequence.
  • a secretory leader sequence of the invention directs certain proteins to the endoplasmic reticulum (ER). The ER separates the membrane-bound proteins from other proteins. Once localized to the ER, proteins can be further directed to the Golgi apparatus for distribution to vesicles; including secretory vesicles; the plasma membrane, lysosomes, and other organelles.
  • Proteins targeted to the ER by a secretory leader sequence can be released into the extracellular space as a secreted protein.
  • vesicles containing secreted proteins can fuse with the cell membrane and release their contents into the extracellular space—a process called exocytosis. Exocytosis can occur constitutively or after receipt of a triggering signal. In the latter case, the proteins may be stored in secretory vesicles (or secretory granules) until exocytosis is triggered. Similarly, proteins residing on the cell membrane can also be secreted into the extracellular space by proteolytic cleavage of a "linker" holding the protein to the membrane. [076] Additionally, peptide moieties and/or purification tags may be added to the polypeptide to facilitate purification. Such regions may be removed prior to final preparation of the polypeptide.
  • Suitable purification tags include, for example, V5, HISX6, HISX8, avidin, and biotin.
  • the invention provides a fusion protein comprising a heterologous region from an immunoglobulin that is useful to stabilize and purify proteins.
  • EP- A-O 464533 (Canadian counterpart 2045869) discloses fusion proteins containing various portions of constant region of immunoglobulin molecules together with another human protein or part thereof.
  • the Fc part of a fusion protein is advantageous for use in therapy and diagnosis and thus results, for example, in improved pharmacokinetic properties (EP-A 0232 262).
  • Fc portion proves to be a hindrance to use in therapy and/or diagnosis, for example, when the fusion protein is to be used as an antigen for immunizations.
  • human proteins such as hIL-5
  • Fc portions have been fused with Fc portions for the purpose of high-throughput screening assays to identify antagonists of hIL-5. See, Bennett et al., J. Molec. Recog., 8:52-58 (1995) and Johanson et al, J. Biol. Chem., 270:9459-9471 (1995).
  • a heterologous polypeptide of the invention can be recovered and purified from recombinant cell cultures by well-known methods including ammonium sulfate or ethanol precipitation, acid extraction, anion or cation exchange chromatography, phosphocellulose chromatography, hydrophobic interaction chromatography, affinity chromatography, hydroxylapatite chromatography, lectin chromatography, and high performance liquid chromatography (HPLC).
  • Polypeptides of the present invention include products purified from natural sources, including bodily fluids, tissues and cells, whether directly isolated or cultured; products of chemical synthetic procedures; and products produced by recombinant techniques from a prokaryotic or eukaryotic host, including, for example, bacterial, yeast, higher plant, insect, and mammalian cells, or from a cell free expression system.
  • a prokaryotic or eukaryotic host including, for example, bacterial, yeast, higher plant, insect, and mammalian cells, or from a cell free expression system.
  • the polypeptides of the present invention may be glycosylated or may be non-glycosylated.
  • polypeptides of the invention may also include an initial modified methionine residue, in some cases as a result of host-mediated processes.
  • the invention further provides isolated G-CSF polypeptides containing the amino acid sequences encoded by the nucleotide sequences set forth in the Sequence Listing, the amino acid sequences set forth in the Sequence Listing, and polypeptides comprising a fragment of any of these.
  • the invention provides secreted proteins, which are capable of being directed to the endoplasmic reticulum (ER), secretory vesicles, or the extracellular space as a result of a secretory leader, signal peptide, or leader sequence, as well as proteins released into the extracellular space without necessarily containing a signal sequence.
  • K a secreted protein is released into the extracellular space, it may undergo extracellular processing to a mature polypeptide. Release into the extracellular space can occur by many mechanisms, including exocytosis and proteolytic cleavage.
  • the sequences of the invention encompass a variety of different types of nucleic acids and polypeptides with different structures and functions.
  • Protein domains are portions of proteins that have a tertiary structure and sometimes have enzymatic or binding activities; multiple domains can be connected by flexible polypeptide regions within a protein.
  • Pfam domains can comprise the N-terminus or the C-terminus of a protein, or can be situated at any point in between.
  • the pfam system identifies protein families based on these domains and provides an annotated, searchable database that classifies proteins into families (Bateman et al., Nucl. Acids Res. 30:276-280 (2002)). Sequences of the invention can encode or be comprised of more than one pfam.
  • Sequences of the invention may comprise an interleukin-6/G-CSF/MGF
  • IL-6 pfam domain.
  • IL-6 is a chemokine that has been reported, inter alia, to play a role in the differentiation of B-cells into immunoglobulin-secreting cells, as well as in the induction of myeloma and plasmacytoma growth, nerve cell differentiation, and acute phase reactants in hepatocytes (Hirano et al., Nature 324:73-76 (1986); Clogston et al., Arch. Biochem. Biophys. 272:144-151 (1989)).
  • Variant and Mutant Polypeptides Protein engineering may be employed to improve or alter the characteristics of G-CSF polypeptides of the invention. Recombinant DNA technology known to those skilled in the art can be used to create novel mutant proteins or "muteins" including single or multiple amino acid substitutions, deletions, additions, or fusion proteins. Such modified polypeptides can show, e.g., enhanced activity or increased stability. In addition, they may be purified in higher yields and show better solubility than the corresponding natural polypeptide, at least under certain purification and storage conditions.
  • the present invention further relates to variants of the nucleic acid molecules of the present invention, which encode portions, analogs, or derivatives of the G-CSF molecules.
  • Variants may occur naturally, such as a natural allelic variant, i.e., one of several alternate forms of a gene occupying a given chromosomal locus Genes II, Lewin, B., ed., John Wiley & Sons, New York (1985)).
  • Non-naturally occurring variants may be produced using art-known mutagenesis techniques.
  • Such variants include those produced by nucleotide substitutions, deletions, or additions.
  • the substitutions, deletions, or additions may involve one or more nucleotides.
  • the variants may be altered in coding regions, non-coding regions, or both. Alterations in the coding regions may produce conservative or non-conservative amino acid substitutions, deletions or additions. These may take the form of silent substitutions, additions, or deletions which do not alter the properties or activities of the described G- CSF proteins, or portions thereof.
  • the invention provides nucleic acid molecules encoding mature proteins, i.e., those with cleaved signal peptide or leader sequences, e.g., as shown in the Sequence Listing.
  • Further embodiments include an isolated nucleic acid molecule comprising a polynucleotide having a nucleotide sequence at least 93% identical, or at least 95%, 96%, 97%, 98%, or 99% identical to a polynucleotide from the Sequence Listing, a polypeptide encoded by a polynucleotide shown in the Sequence Listing, a polypeptide shown in the Sequence Listing, or a biologically active fragment of any of these.
  • a polynucleotide having a nucleotide sequence at least, for example, 95% identical to a reference nucleotide sequence encoding an G-CSF polypeptide is one in which the nucleotide sequence is identical to the reference sequence except that it may include up to five point mutations per each 100 nucleotides of the reference nucleotide sequence.
  • nucleotide having a nucleotide sequence at least 95% identical to a reference nucleotide sequence up to 5% of the nucleotides in the reference sequence may be deleted or substituted with another nucleotide, or a number of nucleotides up to 5% of the total nucleotides in the reference sequence may be inserted into the reference sequence.
  • mutations of the reference sequence may occur at the 5' or 3' terminal positions of the reference nucleotide sequence or anywhere between those terminal positions, interspersed either individually among nucleotides in the reference sequence or in one or more contiguous groups within the reference sequence.
  • nucleotide sequences set forth in the Sequence Listing can be determined conventionally using known computer programs such as the Bestfit program (Wisconsin Sequence Analysis Package, Version 8 for Unix, Genetics Computer Group, Madison, Wl). Bestfit uses the local homology algorithm of Smith and Waterman, Advances in Applied Mathematics 2:482- 489 (1981), to find the best segment of homology between two sequences.
  • the parameters are set, of course, such that the percentage of identity is calculated over the full length of the reference nucleotide sequence and that gaps in homology of up to 5% of the total number of nucleotides in the reference sequence are allowed.
  • the present application is directed to nucleic acid molecules at least 93%
  • nucleic acid sequences set forth in the Sequence Listing irrespective of whether they encode a polypeptide having G-CSF activity. Even where a particular nucleic acid molecule does not encode a polypeptide having G-CSF activity, one of skill in the art would know how to use the nucleic acid molecule, for instance, as a hybridization probe or a polymerase chain reaction (PCR) primer.
  • PCR polymerase chain reaction
  • nucleic acid molecules of the present invention that do not encode a polypeptide having G-CSF activity include, inter alia, (1) isolating the G-CSF gene or allelic variants thereof in a cDNA library; (2) in situ hybridization (e.g., "FISH") to metaphase chromosomal spreads to provide the precise chromosomal location of the G- CSF genes, as described in Verna et al., Human Chromosomes: A Manual of Basic Techniques, Pergamon Press, New York (1988); and Northern blot analysis for detecting G-CSF mRNA expression in specific tissues.
  • FISH in situ hybridization
  • the present application is also directed to nucleic acid molecules having sequences at least 93%, 95%, 96%, 97%, 98%, or 99% identical to a nucleic acid sequence of the Sequence Listing which, encode a polypeptide having G-CSF polypeptide activity, i.e., a polypeptide exhibiting activity either identical to or similar, but not identical, to an activity of the G-CSF polypeptides of the invention, as measured in a particular biological assay.
  • the G-CSF polypeptides of the present invention may either stimulate or inhibit the proliferation of various mammalian cells, as demonstrated below.
  • nucleic acid molecules having a sequence at least 93%, 95%, 96%, 97%, 98%, or 99% identical to the nucleic acid sequence of the nucleic acid sequences set forth in the Sequence Listing will encode a polypeptide having G-CSF polypeptide activity.
  • multiple degenerate variants of these nucleotide sequences encode the same polypeptide, this will be clear to the skilled artisan even without performing the above described comparison assay.
  • nucleic acid molecules that are not degenerate variants will also encode a polypeptide having G-CSF polypeptide activity
  • the skilled artisan is fully aware of amino acid substitutions that are either less likely or not likely to significantly affect protein function (e.g., replacing one aliphatic amino acid with a second aliphatic amino acid), as further described below.
  • N-Terminal and C-Terminal Deletion Mutants [092] For instance, for many proteins, including the extracellular domain of a membrane associated protein or the mature form(s) of a secreted protein, it is known in the art that one or more amino acids may be deleted from the N-terminus or C-terminus without substantial loss of biological function.
  • the present invention further provides polypeptides having one or more residues deleted from the amino terminus of the amino acid sequences of the G-CSF molecules as shown in the Sequence Listing.
  • polypeptides having one or more residues deleted from the amino terminus of the amino acid sequences of the G-CSF molecules as shown in the Sequence Listing.
  • many examples of biologically functional C-terminal deletion muteins are known. For instance, interferon gamma increases in activity as much as ten fold when 8-10 amino acid residues are deleted from the carboxy terminus of the protein, see, for example, Dobeli et al., /. Biotechnology, 7:199-216 (1988).
  • the invention further includes variations of the G-CSF polypeptides which show substantial G-CSF polypeptide activity or which include regions of the G- CSF proteins such as the protein portions discussed below.
  • Such mutants include deletions, insertions, inversions, repeats, and type substitutions, selected according to general rules known in the art, so as have little effect on activity.
  • conservative substitutions are the replacements, one for another, among the aliphatic amino acids Ala, Val, Leu, and lie; interchange of the hydroxyl residues Ser and Thr, exchange of the acidic residues Asp and Glu, substitution between the amide residues Asn and Gin, exchange of the basic residues Lys and Arg, and replacements between the aromatic residues Phe and Tyr.
  • polypeptide encoded by a nucleic acid sequence of the Sequence Listing 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; such a substituted amino acid residue may or may not be one encoded by the genetic code; (ii) one in which one or more of the amino acid residues includes a substituent group; (iii) 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 in which the additional amino acids are fused to the above form of the polypeptide, such as an IgG Fc fusion region peptide, a leader or secretory sequence, a sequence employed to purify the above form of the polypeptide, or a proprotein sequence.
  • the G-CSF polypeptides of the present invention may include one or more amino acid substitutions, deletions, or additions, either from natural mutations or human manipulation. As indicated, these changes may be of a minor nature, such as conservative amino acid substitutions, that do not significantly affect the folding or activity of the protein.
  • Conservative amino acid substitutions include the aromatic substitutions Phe, Trp, and Tyr; the hydrophobic substitutions Leu, Iso, and Val; the polar substitutions Glu and Asp; the basic substitutions Arg, Lys, and His; the acidic substitutions Asp and Glu; and the small amino acid substations Ala, Ser, Thr, Met, and Gly.
  • Amino acids essential for the functions of G-CSF polypeptides can be identified by methods known in the art, such as site-directed mutagenesis or alanine- scanning mutagenesis, see, for example, Cunningham and Wells, Science, 244:1081-1085 (1989). The latter procedure introduces single alanine mutations. The resulting mutant molecules are then tested for biological activity such as receptor binding, or in vitro or in vitro proliferative activity.
  • Replacing amino acids can also change the selectivity of the binding of a ligand to cell surface receptors.
  • Ostade et al. Nature, 361:266-268 (1993) describes certain mutations resulting in selective binding of TNF- ⁇ to only one of the two known types of TNF receptors.
  • Sites that are critical for ligand-receptor binding can also be determined by structural analysis such as crystallization, nuclear magnetic resonance, or photoaffinity labeling, for example, Smith et al., J. Mol. Biol, 224:899-904 (1992) and de Vos et al., Science, 255:306-312 (1992).
  • polypeptides of the present invention can be provided in an isolated form, and can be substantially purified.
  • a recombinantly produced version of the herein described G-CSF polypeptides can be substantially purified, e.g., by the one-step method described in Smith and Johnson, Gene, 67:31-40 (1988).
  • Polypeptides of the invention also can be purified from natural or recombinant sources using anti- G-CSF antibodies of the invention using methods which are well known in the art of protein purification.
  • the polypeptides herein may be purified or isolated in the presence of ions or agents that aid in the refolding of the molecules or aid in dimerizing or trimerizing the molecules as conventional in the art.
  • polypeptides of the present invention include polypeptides which have at least 93%, 95%, 96%, 97%, 98%, or 99% similarity to those described above.
  • the polypeptides of the invention also contain those which are at least 93%, 94%, or 95%, 96%, 97%, 98%, or 99% identical to a polypeptide encoded by a nucleic acid sequence of the Sequence Listing.
  • the percent similarity of two polypeptides can be measured by a similarity score determined by comparing the amino acid sequences of the two polypeptides using the Bestfit program with the default settings for determining similarity. Bestfit uses the local homology algorithm of Smith and Waterman, Advances in Applied Mathematics 2:482-489 (1981) to find the best segment of similarity between two sequences.
  • the polypeptides of the present invention can be used to raise polyclonal and monoclonal antibodies, which are useful in assays for detecting G-CSF protein expression, also as described below, or as agonists and/or antagonists capable of enhancing or inhibiting G-CSF protein function.
  • polypeptides can also be used in a yeast two-hybrid system to capture G-CSF protein binding proteins, which are also candidate agonists and antagonists, according to the present invention.
  • the yeast two hybrid system is described in Fields and Song, Nature, 340:245-246 (1989).
  • Aptamers [0108] Another suitable agent for modulating an activity of a subject polypeptide is an aptamer.
  • Aptamers of the invention include both nucleotide and peptide aptamers.
  • Nucleotide aptamers of the invention include double stranded DNA and single stranded RNA molecules that bind to G-CSF proteins or fragments thereof.
  • Peptide aptamers are peptides or small polypeptides that act as dominant inhibitors of protein function. Peptide aptamers specifically bind to target proteins, blocking their functional ability (Kolonin et al., Proc. Natl. Acad. Sci. 95:14,266-14,271 (1998)). [0109] Due to the highly selective nature of peptide aptamers, they can be used not only to target a specific protein, but also to target specific functions of a given protein (for example, a signaling function). Further, peptide aptamers can be expressed in a controlled fashion by use of promoters which regulate expression in a temporal, spatial, or inducible manner.
  • Peptide aptamers act dominantly, therefore, they can be used to analyze proteins for which loss-of-function mutants are not available.
  • Aptamers of the invention may bind nucleotide cofactors (Latham et al., Nucl. Acids Res. 22:2817-2822 (1994)).
  • Peptide aptamers that bind with high affinity and specificity to a target protein can be isolated by a variety of techniques known in the art. Peptide aptamers can be isolated from random peptide libraries by yeast two-hybrid screens (Xu et al., Proc. Natl. Acad. Sci. 94:12,473-12,478. (1997)). They can also be isolated from phage libraries (Hoogenboom et al., Immunotechnology 4:1-20 (1998)) or chemically generated peptides/libraries. Epitope-Bearing Portions [0111] In another aspect, the invention provides a polypeptide comprising an epitope-bearing portion of a polypeptide of the invention.
  • the epitope of this polypeptide portion is an immunogenic or antigenic epitope of a polypeptide of the invention.
  • Immunogenic epitopes are those parts of a protein that elicit an antibody response when the whole protein is provided as the immunogen.
  • a region of a protein molecule to which an antibody can bind is an antigenic epitope.
  • the number of immunogenic epitopes of a protein generally is less than the number of antigenic epitopes. See, for instance, Geysen et al., Proc. Natl. Acad. Sci., USA 81:3998-4002 (1983).
  • polypeptides bearing an antigenic epitope i.e., that contain a region of a protein molecule to which an antibody can bind
  • relatively short synthetic peptides that mimic part of a protein sequence are routinely capable of eliciting an antiserum that reacts with the partially mimicked protein. See, for instance, Sutcliffe et al., Science, 219:660-666 (1983).
  • Peptides capable of eliciting protein-reactive sera are frequently represented in the primary sequence of a protein, can be characterized by a set of simple chemical rules, and are confined neither to immunodominant regions of intact proteins (i.e., immunogenic epitopes) nor to the amino or carboxyl terminals.
  • Antigenic epitope-bearing peptides and polypeptides of the invention are therefore useful for raising antibodies, including monoclonal antibodies, that bind specifically to a polypeptide of the invention. See, for instance, Wilson et al., Cell, 37:767-778 (1984).
  • the epitope-bearing peptides and polypeptides of the invention may be produced by any conventional means. See, for example, Houghten, Proc. Natl.
  • Epitope-bearing peptides and polypeptides of the invention can be used to induce antibodies according to methods well known in the art. See, for instance, Bittle, et al, J. Gen. Virol, 66:2347-2354 (1985).
  • Immunogenic epitope-bearing peptides of the invention i.e., those parts of a protein that elicit an antibody response when the whole protein is the immunogen, are identified according to methods known in the art. See, for instance, U.S. Pat. No.
  • 5,480,971 discloses linear Cl-C7-alkyl peralkylated oligopeptides, and sets and libraries of such peptides, as well as methods for using such oligopeptide sets and libraries for determining the sequence of a peralkylated oligopeptide that preferentially binds to an acceptor molecule of interest.
  • non-peptide analogs of the epitope-bearing peptides of the invention also can be made routinely by these methods.
  • G-CSF polypeptides of the present invention can be combined with parts of the constant domain of immunoglobulins, resulting in chimeric polypeptides.
  • These fusion proteins facilitate purification and show an increased half-life in vivo. This has been shown, for example, for chimeric proteins consisting of the first two domains of the human CD4-polypeptide and various domains of the constant regions of the heavy or light chains of mammalian immunoglobulins (Fc), for example, as described in EP A 394,827; Traunecker et al., Nature, 331:84-86 (1988).
  • Fusion proteins that have a disulfide-linked dimeric structure due to the IgG part can also be more efficient in binding and neutralizing other molecules than the monomeric G-CSF protein or protein fragment alone, for example, as described by Fountoulakis et al., J. Biochem., 270:3958-3964 (1995).
  • Suitable chemical moieties for derivatization of a heterologous polypeptide include, for example, polymers, such as water soluble polymers, all or part of human serum albumin, fetuin A; fetuin B; a leucine zipper domain; a tetranectin trimerization domain; mannose binding protein (also known as mannose binding lectin), for example, mannose binding protein 1 ; and an Fc region.
  • polymers such as water soluble polymers, all or part of human serum albumin, fetuin A; fetuin B; a leucine zipper domain; a tetranectin trimerization domain; mannose binding protein (also known as mannose binding lectin), for example, mannose binding protein 1 ; and an Fc region.
  • heterologous polypeptides of the present invention and the epitope-bearing fragments thereof described herein can be combined with parts of the constant domain of immunoglobulins (IgG), resulting in chimeric polypeptides.
  • IgG immunoglobulins
  • These particular fusion molecules facilitate purification and show an increased half-life in vivo. This has been shown, e.g., for chimeric proteins consisting of the first two domains of the human CD4-polypeptide and various domains of the constant regions of the heavy or light chains of mammalian immunoglobulins, for example, EP A 394,827; Traunecker et al., Nature, 331:84-86 (1988).
  • Fusion molecules that have a disulfide-linked dimeric structure due to the IgG part can also be more efficient in binding and neutralizing other molecules than, for example, a monomeric polypeptide or polypeptide fragment alone, see, for example, Fountoulakis et al., J. Biochem., 270:3958-3964 (1995).
  • a human serum albumin fusion molecule may also be prepared as described herein and as further described in U.S. Patent No. 6,686,179.
  • Peptides that bind albumin are also suitable fusion partners, as are lipids or other molecules that bind albumin.
  • polypeptides of the present invention can be fused to marker sequences, such as a peptide that facilitates purification of the fused polypeptide.
  • the marker amino acid sequence may be a hexa-histidine peptide such as the tag provided in a pQE vector (Qiagen, Inc., among others, many of which are commercially available. As described in Gentz et al., Proc. Natl Acad. Sci. USA 86:821-824 (1989), for instance, hexa-histidine provides for convenient purification of the fusion protein.
  • chemagglutinin HA tag corresponds to an epitope derived from the influenza hemagglutinin protein. (Wilson et al., Cell 37:767 (1984)).
  • Oligomeric proteins may be used as fusion partners to increase molecular weight, thus decreasing kidney filtration.
  • coiled-coil domains such as the tetramectin coiled-coil domain, the coiled-coil domain of cartilage oligomeric matrix protein, and angiopoietin coiled-coil domains, and leucine zipper domains are suitable fusion partners.
  • collagen and collagen-like domains including those found in collagens, mannose-binding lectin, lung surfactant proteins A and D, adiponectin, ficolin, conglutinin, macrophage scavenger receptor, and emilin.
  • Dimeric immunoglobulin domains for example antibody CH3 domains, are also suitable.
  • Covalent modifications can also increase molecular size. PEGylation, described in more detail above, and hyperglycosylation are examples of suitable covalent modifications.
  • Polymers e.g., water soluble polymers, are useful in the present invention as the polypeptide to which each polymer is attached will not precipitate in an aqueous environment, such as a physiological environment.
  • Polymers employed in the invention will be pharmaceutically acceptable for the preparation of a therapeutic product or composition.
  • One skilled in the art will be able to select the desired polymer based on such considerations as whether the polymer/protein conjugate will be used therapeutically and, if so, the desired dosage, circulation time, and resistance to proteolysis.
  • Suitable, clinically acceptable, water soluble polymers include, but are not limited to, polyethylene glycol (PEG), polyethylene glycol propionaldehyde, copolymers of ethylene glycol/propylene glycol, monomethoxy-polyethylene glycol, carboxymethylcellulose, dextran, polyvinyl alcohol (PVA), polyvinyl pyrrolidone, poly- 1,3-dioxolane, poly-l,3,6-trioxane, ethylene/maleic anhydride copolymer, poly ( ⁇ -amino acids) (either homopolymers or random copolymers), poly(n- vinyl pyrrolidone) polyethylene glycol, polypropylene glycol homopolymers (PPG) and other polyakylene oxides, polypropylene oxide/ethylene oxide copolymers, polyoxyethylated polyols (POG) (e.g., glycerol) and other polyoxyethylated polyols, poly
  • polyethylene glycol is meant to encompass any of the forms that have been used to derivatize other proteins, such as mono-(Cl-ClO) alkoxy- or aryloxy-polyethylene glycol.
  • Polyethylene glycol propionaldehyde may have advantages in manufacturing due to its stability in water.
  • a modified heterologous polypeptide of the invention may be prepared by attaching polyamino acids or branch point amino acids to the polypeptide.
  • the polyamino acid may be a carrier protein that serves to increase the circulation half life of the polypeptide (i.e., in addition to the advantages achieved via a fusion molecule).
  • such polyamino acids should ideally be those that have or do not create neutralizing antigenic response, or other adverse responses.
  • Such polyamino acids may be selected from serum album (such as human serum albumin), an additional antibody or portion thereof, for example the Fc region, fetuin A, fetuin B, leucine zipper nuclear factor erythroid derivative-2 (NFE2), neuroretinal leucine zipper, mannose motif (mbpl), tetranectin, or other polyamino acids, e.g. lysines.
  • serum album such as human serum albumin
  • an additional antibody or portion thereof for example the Fc region, fetuin A, fetuin B, leucine zipper nuclear factor erythroid derivative-2 (NFE2), neuroretinal leucine zipper, mannose motif (mbpl), tetranectin, or other polyamino acids, e.g. lysines.
  • Fc region Fc region
  • fetuin A fetuin A
  • fetuin B leucine zipper nuclear factor erythroid
  • Polymers used herein may be of any molecular weight and may be branched or unbranched.
  • the polymers each typically have an average molecular weight of between about 1 kDa to about 100 kDa (in reference to a polymer, the term "about” indicating that in preparations of a polymer, some molecules will weigh more, some less, than the stated molecular weight).
  • the average molecular weight of each polymer may be between about 5 kDa and about 50 kDa, or between about 12 kDa and about 25 kDa. Generally, the higher the molecular weight or the more branches, the higher the polyme ⁇ protein ratio.
  • Polymers employed in the present invention are typically attached to a heterologous polypeptide with consideration of effects on functional or antigenic domains of the polypeptide. h general, chemical derivatization may be performed under any suitable condition used to react a protein with an activated polymer molecule.
  • Activating groups which can be used to link the polymer to the active moieties include the following: sulfone, maleimide, sulfhydryl, thiol, triflate, tresylate, azidirine, oxirane, and 5-pyridyl.
  • Polymers of the invention are typically attached to a heterologous polypeptide at the alpha ( ⁇ ) or epsilon ( ⁇ ) amino groups of amino acids or a reactive thiol group, but it is also contemplated that a polymer group could be attached to any reactive group of the protein that is sufficiently reactive to become attached to a polymer group under suitable reaction conditions.
  • a polymer may be covalently bound to a heterologous polypeptide via a reactive group, such as a free amino or carboxyl group.
  • the amino acid residues having a free amino group may include lysine residues and the N-terminal amino acid residue.
  • Those having a free carboxyl group may include aspartic acid residues, glutamic acid residues, and the C-terminal amino acid residue.
  • Those having a reactive thiol group include cysteine residues.
  • Methods for preparing fusion molecules conjugated with polymers will each generally involve (a) reacting a heterologous polypeptide with a polymer under conditions whereby the polypeptide becomes attached to one or more polymers and (b) obtaining the reaction product.
  • Reaction conditions for each conjugation may be selected from any of those known in the art or those subsequently developed, but should be selected to avoid or limit exposure to reaction conditions such as temperatures, solvents, and pH levels that would inactivate the protein to be modified.
  • the optimal reaction conditions for the reactions will be determined case-by-case based on known parameters and the desired result. For example, the larger the ratio of polymer:polypeptide conjugate, the greater the percentage of conjugated product.
  • the optimum ratio in terms of efficiency of reaction in that there is no excess unreacted polypeptide or polymer
  • the ratio of polymer (e.g., PEG) to a polypeptide will generally range from 1 : 1 to 100: 1.
  • One or more purified conjugates may be prepared from each mixture by standard purification techniques, including among others, dialysis, salting-out, ultrafiltration, ion-exchange chromatography, gel filtration chromatography, and electrophoresis.
  • the method of obtaining the N-terminal chemically modified protein preparation i.e., separating this moiety from other monoderivatized moieties if necessary
  • Selective N-terminal chemical modification may be accomplished by reductive alkylation which exploits differential reactivity of different types of primary amino groups (lysine versus the N-terminal) available for derivatization in a particular protein.
  • substantially selective derivatization of the protein at the N-terminus with a carbonyl group containing polymer is achieved.
  • the conjugation with the polymer takes place predominantly at the N-terminus of the protein and no significant modification of other reactive groups, such as the lysine side chain amino groups, occurs.
  • the polymer may be of the type described above and should have a single reactive aldehyde for coupling to the protein.
  • Polyethylene glycol propionaldehyde, containing a single reactive aldehyde, may also be used.
  • the present invention contemplates the chemically derivatized polypeptide to include mono- or poly- (e.g., 2-4) PEG moieties.
  • Pegylation may be carried out by any of the pegylation reactions known in the art.
  • Methods for preparing a pegylated protein product will generally include (a) reacting a polypeptide with polyethylene glycol (such as a reactive ester or aldehyde derivative of PEG) under conditions whereby the protein becomes attached to one or more PEG groups; and (b) obtaining the reaction product(s).
  • polyethylene glycol such as a reactive ester or aldehyde derivative of PEG
  • the optimal reaction conditions for the reactions will be determined case by case based on known parameters and the desired result.
  • heterologous secretory leader sequences are advantageous in that a resulting mature amino acid sequence, of the secreted polypeptide is not altered as the secretory leader sequence is removed in the ER during the secretion process.
  • addition of a heterologous secretory leader is often required to express and secrete.
  • extracellular domains of Type II single transmembrane proteins (STM) as the secretory leader, which is also a transmembrane spanning domain, must typically be removed so that they may be soluble.
  • the invention encompasses polypeptides which are differentially modified during or after translation, e.g., by glycosylation, acetylation, phosphorylation, amidation, derivatization by known protecting/blocking groups, proteolytic cleavage, linkage to an antibody molecule or other cellular ligand. Any of numerous chemical modifications may be carried out by known techniques, including but not limited to specific chemical cleavage by cyanogen bromide, trypsin, chymotrypsin, papain, V8 protease; NABH 4; acetylation; formylation; oxidation; reduction; and/or metabolic synthesis in the presence of tunicamycin.
  • Additional post-translational modifications encompassed by the invention include, for example, e.g., N-linked or O-linked carbohydrate chains, processing of N- terminal or C-terminal ends), attachment of chemical moieties to the amino acid backbone, chemical modifications of N-linked or O-linked carbohydrate chains, and addition or deletion of an N-terminal methionine residue as a result of prokaryotic host cell expression.
  • the polypeptides may also be modified with a detectable label, such as an enzymatic, fluorescent, isotopic, or affinity label to allow for detection and isolation of the protein.
  • G-CSF compositions are provided in formulation with pharmaceutically acceptable excipients, a wide variety of which are known in the art (Gennaro, Remington: The Science and Practice of Pharmacy with Facts and Comparisons: Drugfacts Plus, 20th ed. (2003); Ansel et al., Pharmaceutical Dosage Forms and Drug Delivery Systems, 7 th ed., Lippencott Williams and Wilkins (2004); Kibbe et al., Handbook of Pharmaceutical Excipients, 3 r ed., Pharmaceutical Press (2000)).
  • Pharmaceutically acceptable excipients such as vehicles, adjuvants, carriers or diluents, are readily available to the public.
  • compositions for oral administration can form solutions, suspensions, tablets, pills, granules, capsules, sustained release formulations, oral rinses, or powders.
  • the agents, polynucleotides, and polypeptides can be used alone or in combination with appropriate additives, for example, with conventional additives, such as lactose, mannitol, corn starch, or potato starch; with binders, such as crystalline cellulose, cellulose derivatives, acacia, corn starch, or gelatins; with disintegrators, such as corn starch, potato starch, or sodium carboxymethylcellulose; with lubricants, such as talc or magnesium stearate; and if desired, with diluents, buffering agents, moistening agents, preservatives, and flavoring agents.
  • conventional additives such as lactose, mannitol, corn starch, or potato starch
  • binders such as crystalline cellulose, cellulose derivatives, acacia, corn starch, or gelatins
  • disintegrators such as corn starch, potato starch, or sodium carboxymethylcellulose
  • lubricants such as talc or magnesium stea
  • Suitable excipient vehicles are, for example, water, saline, dextrose, glycerol, ethanol, or the like, and combinations thereof.
  • the vehicle can contain minor amounts of auxiliary substances such as wetting or emulsifying agents or pH buffering agents. Actual methods of preparing such dosage forms are known, or will be apparent, to those skilled in the art (Gennaro, 2003).
  • the composition or formulation to be administered will, in any event, contain a quantity of the agent adequate to achieve the desired state in the subject being treated.
  • the agents, polynucleotides, and polypeptides can be formulated into preparations for injection by dissolving, suspending or emulsifying them in an aqueous or nonaqueous solvent, such as vegetable or other similar oils, synthetic aliphatic acid glycerides, esters of higher aliphatic acids or propylene glycol; and if desired, with conventional additives such as solubilizers, isotonic agents, suspending agents, emulsifying agents, stabilizers and preservatives.
  • an aqueous or nonaqueous solvent such as vegetable or other similar oils, synthetic aliphatic acid glycerides, esters of higher aliphatic acids or propylene glycol
  • solubilizers isotonic agents
  • suspending agents emulsifying agents
  • stabilizers and preservatives emulsifying agents
  • the antibodies, agents, polynucleotides, and polypeptides can be utilized in aerosol formulation to be administered via inhalation.
  • the compounds of the present invention can be formulated into pressurized acceptable propellants such as dichlorodifluoromethane, propane, nitrogen, and the like.
  • the agent, polynucleotides, or polypeptide composition may be converted to powder form for administration intranasally or by inhalation, as conventional in the art.
  • the agents can be made into suppositories by mixing with a variety of bases such as emulsifying bases or water-soluble bases.
  • the compounds of the present invention can be administered rectally via a suppository.
  • the suppository can include vehicles such as cocoa butter, carbowaxes and polyethylene glycols, which melt at body temperature, yet are solidified at room temperature.
  • a polynucleotide, polypeptide, or other modulator can also be introduced into tissues or host cells by other routes, such as viral infection, microinjection, or vesicle fusion.
  • expression vectors can be used to introduce nucleic acid compositions into a cell as described above.
  • jet injection can be used for intramuscular administration (Furth et al., Anal. Biochem. 205:365-368 (1992)).
  • the DNA can be coated onto gold microparticles, and delivered intradermally by a particle bombardment device, or "gene gun” as described in the literature (Tang et al., Nature 356:152-154 (1992)), where gold microprojectiles are coated with the DNA, then bombarded into skin cells.
  • Unit dosage forms for oral or rectal administration such as syrups, elixirs, and suspensions can be provided wherein each dosage unit, for example, teaspoonful, tablespoonful, tablet, or suppository, contains a predetermined amount of the composition containing one or more agents.
  • unit dosage forms for injection or intravenous administration can comprise the agent(s) in a composition as a solution in sterile water, normal saline or another pharmaceutically acceptable carrier.
  • Chromosome Assays [0143] In certain embodiments relating to chromosomal mapping, a cDNA herein disclosed is used to clone the genomic nucleic acid of the G-CSF. This can be accomplished using a variety of well known techniques and libraries, which generally are commercially available.
  • the genomic DNA then is used for in situ chromosome mapping using techniques well known for this purpose. Therefore, the nucleic acid molecules 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 the 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 is an important first step in correlating those sequences with genes associated with disease.
  • sequences can be mapped to chromosomes by preparing PCR primers 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. [0145] PCR mapping of somatic cell hybrids is a rapid procedure for assigning a particular DNA to a particular chromosome.
  • 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.
  • FISH Fluorescence in situ hybridization
  • cDNA or genomic sequences of affected and unaffected individuals differences can be determined in the cDNA or genomic sequences of affected and unaffected individuals. If a mutation is observed in some or all of the 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 physical 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 genes (assuming 1 megabase mapping resolution and one gene per 20 kb). Identification of Agonists and Antagonists [0149] This invention provides modulators, i.e., polypeptides, polynucleotides, or other agents that increase or decrease the activity of their target.
  • modulators i.e., polypeptides, polynucleotides, or other agents that increase or decrease the activity of their target.
  • an agent is a subject polypeptide, where the subject polypeptide itself is administered to an individual.
  • an agent is an antibody specific for a subject "target" polypeptide.
  • an agent is a chemical compound such as a small molecule that may be useful as an orally available drug.
  • Such modulation includes the recruitment of other molecules that directly effect the modulation.
  • an antibody that modulates the activity of a subject polypeptide that is a receptor on a cell surface may bind to the receptor and fix complement, activating the complement cascade and resulting in lysis of the cell.
  • An agent which modulates a biological activity of a subject polypeptide or polynucleotide increases or decreases the activity or binding at least about 10%, at least about 15%, at least about 20%, at least about 25%, at least about 50%, at least about 80%, or at least about 2-fold, at least about 5-fold, or at least about 10-fold or more when compared to a suitable control.
  • This invention also provides a method of screening compounds to identify those which modulate the action of the polypeptide of the present invention.
  • An example of such an assay comprises combining a mammalian fibroblast cell and the polypeptide(s) of the present invention, the compound to be screened and 3 [H] thymidine under cell culture conditions where the fibroblast cell would normally proliferate.
  • a control assay may be performed in the absence of the compound to be screened and compared to the amount of fibroblast proliferation in the presence of the compound to determine if the compound stimulates proliferation by determining the uptake of 3 [H] thymidine in each case.
  • fibroblast cell proliferation is measured by liquid scintillation chromatography, which measures the incorporation of [H] thymidine. Both agonistic and antagonistic compounds may be identified by this procedure.
  • a mammalian cell or membrane preparation expressing a receptor for a polypeptide of the present invention, as described above, is incubated with a labeled polypeptide of the present invention in the presence of the compound. The ability of the compound to enhance or block this interaction could then be measured.
  • the response of a known second messenger system following interaction of a compound to be screened and the G-CSF receptor is measured and the ability of the compound to bind to the receptor and elicit a second messenger response is measured to determine if the compound is a potential agonist or antagonist.
  • second messenger systems include, but are not limited to, cAMP, guanylate cyclase, ion channels, and phosphoinositide hydrolysis.
  • antagonistic compounds include antibodies, or in some cases, oligonucleotides, which bind to a receptor of a polypeptide of the present invention but elicit no second messenger response, or which bind to the G-CSF polypeptide itself.
  • a potential antagonist may be a mutant form of the polypeptide which binds to the receptors but elicits no second messenger response, thus effectively blocking the action of the polypeptide.
  • Another compound antagonistic to G-CSF genes and gene products 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 methods are based on the binding of a polynucleotide to DNA or RNA.
  • a 5' coding portion of the polynucleotide sequence, which encodes mature polypeptides of the present invention can be used to design an antisense RNA oligonucleotide of from about 10 to about 40 base pairs in length.
  • a DNA oligonucleotide is designed to be complementary to a region of the gene involved in transcription, for example, a triple helix (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 the polypeptides of the present invention.
  • the antisense RNA oligonucleotide hybridizes to the mRNA in vivo and blocks translation of the mRNA molecule into the polypeptide, as described by Okano, J.
  • oligonucleotides as Antisense Inhibitors of Gene Expression, CRC Press, Boca Raton, Fla. (1988).
  • the oligonucleotides described above can also be delivered to cells such that the antisense RNA or DNA is expressed in vivo to inhibit polypeptide production.
  • Potential antagonist compounds also include small molecules which bind to and occupy the binding site of the receptors, thereby making the receptor inaccessible to its polypeptide such that normal biological activity is prevented.
  • small molecules include, but are not limited to, small peptides or peptide-like molecules.
  • Antagonist compounds may be employed to inhibit the cell growth and proliferation effects of the polypeptides of the present invention on neoplastic cells and tissues, i.e. stimulation of angiogenesis of tumors, and, therefore, retard or prevent abnormal cellular growth and proliferation, for example, in tumor formation or growth.
  • the present invention also provides methods for identifying agents, such as antibodies, which enhance or block the actions of G-CSF molecules on cells.
  • agents of interest include both agonists and antagonists.
  • the invention provides agonists which increase the natural biological functions of G-CSF or which function in a manner similar to G-CSF.
  • the invention also provides antagonists, which decrease or eliminate the functions of G-CSF.
  • a cellular compartment such as a membrane or cytosolic preparation may be prepared from a cell that expresses a molecule that binds G-CSF molecules, such as a molecule of a signaling or regulatory pathway modulated by G-CSF molecules.
  • Subcellular fractionation methods are known in the art of cell biology, and can be tailored to produce crude fractions with discrete and defined components, e.g., organelles or organellar membranes. The preparation is incubated with labeled G-CSF molecules in the absence or the presence of a candidate molecule which may be an G-CSF agonist or antagonist.
  • the ability of the candidate molecule to interact with the binding molecule or an G-CSF molecules is reflected in decreased binding of the labeled ligand.
  • Molecules which bind gratuitously, that is, without inducing the effects of G-CSF molecules are most likely antagonists. Molecules that bind well and elicit effects that are the same as or closely related to G-CSF molecules may potentially prove to be agonists.
  • the effects of potential agonists and antagonists may by measured, for instance, by determining an activity of one or more components of a second messenger system following interaction of the candidate molecule with a cell or appropriate cell preparation, and comparing the effect with that of G-CSF molecules, or with that of molecules that elicit the same effects as G-CSF.
  • Second messenger systems which may be useful in this regard include, but are not limited to, cAMP, cGMP, ion channel, and phosphoinositide hydrolysis second messenger systems.
  • a competitive assay for G-CSF antagonists combines a mixture of G-CSF molecules and a potential antagonist with membrane-bound G-CSF receptor molecules. Under appropriate conditions, this assay can also be performed with recombinant G-CSF receptor molecules. G-CSF molecules can be labeled, such as by radioactivity, such that the number of G-CSF molecules bound to a receptor molecule can be determined accurately to assess the effectiveness of the potential antagonist.
  • Potential antagonists include small organic molecules, polypeptides, and antibodies that bind to a polypeptide of the invention, and thereby inhibit or extinguish its activity. Potential antagonists also may be small organic molecules, polypeptides such as closely related proteins or antibodies that bind the same sites on a binding molecule, such as a receptor molecule, without inducing G-CSF -induced activities, thereby preventing the action of G-CSF molecules by excluding G-CSF molecules from binding.
  • Antagonists of the invention include fragments of the G-CSF molecules having the nucleic acid and amino acid sequences shown in the Sequence Listing.
  • G-CSF polynucleotides, polypeptides, agonists, and/or antagonists of the invention may be used to treat neutropenia, including febrile neutropenia, sepsis, and other bacterial-mediated infections. They may be administered, for example, to enhance the efficacy of neutrophils or to expand the neutrophil compartment to reduce inflammation mediated by defenders, such as lymphocytes. They can be used to treat inflammatory disease caused by or characterized by an inability to eradicate pathogenic bacteria via the innate immune system, such that inappropriate reactions to self-antigens ensue when the adaptive immune response, such as a lymphocytic response, occurs. Crohn's disease is an example of such an inflammatory disease. In Crohn's disease, the innate immune response fails to remove bacteria that breach the intestinal epithelium. An adaptive immune response, characterized by inflammation resulting from lymphocytes secreting cytokines results.
  • Antisense molecules can be used to control gene expression through, e.g., antisense DNA or RNA, or through triple-helix formation. Antisense techniques are discussed, for example, in Okano, J. Neurochem., 56:560 (1991); Oligodeoxynucleotides as Antisense Inhibitors of Gene Expression, CRC Press, Boca Raton, Fla. (1988). Triple helix formation is discussed in, for instance, Lee et al., Nucleic Acids Research, 3:173 (1979); Cooney et al., Science, 241:456 (1988); and Dervan et al., Science, 251:1360 (1991).
  • the methods are based on the binding of a polynucleotide to a complementary DNA or RNA.
  • the 5' coding portion of a polynucleotide that encodes the mature polypeptide of the present invention may be used to design an antisense RNA oligonucleotide of from about 10 to about 40 base pairs in length.
  • a DNA oligonucleotide is designed to be complementary to a region of the gene involved in transcription, thereby preventing transcription and the subsequent production of G-CSF molecules.
  • the antisense RNA oligonucleotide hybridizes to the mRNA in vivo and blocks translation of the mRNA molecule into an G-CSF polypeptide.
  • G-CSF polynucleotides, polypeptides, agonists, and/or antagonists of the invention may be used in developing treatments for any disorder mediated (directly or indirectly) by defective G-CSF molecules, or insufficient GSF.
  • G-CSF polypeptides, agonists, and/or antagonists may be administered to a patient (e.g., a mammal, such as human) afflicted with such a disorder.
  • G-CSF nucleotide sequences permits the detection of defective G-CSF genes, and the replacement thereof with normal G-CSF- encoding genes.
  • Defective genes may be detected in in vitro diagnostic assays, and by comparison of the G-CSF nucleotide sequences disclosed herein with that of an G-CSF gene derived from a patient suspected of harboring a defect in this gene.
  • the G-CSF molecules of the present invention may be employed to treat lymphoproliferative disease which results in lymphadenopathy.
  • the G-CSF molecules may further be employed as a research tool in elucidating the biology of autoimmune disorders, including systemic lupus erythematosus (SLE), Graves' disease, immunoproliferative disease lymphadenopathy (IPL), angioimmunoproliferative lymphadenopathy (AIL), immunoblastic lymphadenopathy (H3L), rheumatoid arthritis, diabetes, and multiple sclerosis. It also finds use in treating allergies and graft versus host disease.
  • G-CSF polynucleotides, polypeptides, and/or agonists or antagonists of the invention may also be used to treat, prevent, diagnose, and/or prognose diseases which include, but are not limited to, autoimmune disorders, immunodeficiency disorders, and graft versus host disease.
  • Th2 lymphocyte related disorders e.g., atopic dermatitis, atopic asthma, rhinoconjunctivitis, allergic rhinitis, Omenn's syndrome, systemic sclerosis, and graft versus host disease
  • Th-1 lymphocyte-related disorders e.g., rheumatoid arthritis, multiple sclerosis, psoriasis, Sjogren's syndrome, Hashimoto's thyroiditis, Grave's disease, primary biliary cirrhosis, Wegener's granulomatosis, and tuberculosis
  • activated B lymphocyte-related disorders e.g., systemic lupus erythematosus, Goodpasture's syndrome, rheumatoid arthritis, and type I diabetes.
  • Molecules of the invention are useful for killing or inhibiting the multiplication of a cell that produces an infectious disease or for treating an infectious disease.
  • the molecules of the invention can be used accordingly in a variety of settings for the treatment of an infectious disease in an animal.
  • the term "treating" includes any or all of preventing the growth, multiplication or replication of the pathogen that causes the infectious disease and ameliorating one or more symptoms of an infectious disease.
  • the G-CSF polypeptides of the present invention may be employed to inhibit neoplasia, such as tumor cell growth. They may be responsible for tumor destruction through apoptosis and cytotoxicity to certain cells.
  • Diseases associated with increased cell survival, or the inhibition of apoptosis that may be treated, prevented, diagnosed and/or prognosed with the G-CSF polynucleotides, polypeptides and/or agonists or antagonists of the invention include, but are not limited to, cancers (such as follicular lymphomas, carcinomas with p53 mutations, and hormone-dependent tumors, including, but not limited to colon cancer, cardiac tumors, pancreatic cancer, melanoma, retinoblastoma, glioblastoma, lung cancer, intestinal cancer, testicular cancer, stomach cancer, neuroblastoma, myxoma, myoma, lymphoma, endothelioma, osteoblastoma, osteoclastoma, osteosarcoma, chondrosarcoma, adenoma, breast cancer, prostate cancer, Kaposi's sarcoma, and ovarian cancer); autoimmune disorders (such as, multiple sclerosis
  • G-CSF polynucleotides and/or polypeptides, and their agonists, and/or antagonists may be used to inhibit growth, progression, and/or metastasis of cancers, in particular those listed above or in the paragraph that follows.
  • Additional diseases or conditions associated with increased cell survival that may be treated, prevented, diagnosed, and/or prognosed with the G-CSF polynucleotides and/or polypeptides and their agonists and/or antagonists include, but are not limited to, progression and/or metastases of malignancies and related disorders such as leukemia (including acute leukemias, (e.g., acute lymphocytic leukemia and acute myelocytic leukemia (including myeloblastic, promyelocytic, myelomonocytic, monocytic, and erythroleukemia) and chronic leukemias (e.g., chronic myelocytic (granulocytic) leukemia and chronic lymphocytic leukemia)), polycythemia vera, lymphomas (e.g., Hodgkin's disease and non-Hodgkin's disease), multiple myeloma, Waldenstrom's macroglobulinemia, heavy chain diseases,
  • Diseases associated with increased apoptosis that may be treated, prevented, diagnosed and/or prognosed with the G-CSF polynucleotides, polypeptides and/or agonists or antagonists of the invention include, but are not limited to, AIDS, neurodegenerative disorders (such as Alzheimer's disease, Parkinson's disease, amyotrophic lateral sclerosis, retinitis pigmentosa, cerebellar degeneration, and brain tumor or prior associated disease); diabetes, autoimmune disorders (such as, multiple sclerosis, Sj ⁇ gren's syndrome, Graves' disease, Hashimoto's thyroiditis, autoimmune diabetes, biliary cirrhosis, Behcet's disease, Crohn's disease, polymyositis, SLE, immune- related glomerulonephritis, autoimmune gastritis, thrombocytopenic purpura, and rheumatoid arthritis) myelodysplastic syndromes (such as aplastic anemia),
  • AIDS neuro
  • G-CSF polynucleotides, polypeptides, agonists, and/or antagonists are used to treat the diseases and disorders listed above.
  • G-CSF polynucleotides, polypeptides, agonists or antagonists of the invention are used to treat AIDS and pathologies associated with AIDS.
  • Another embodiment of the present invention is directed to the use of G-CSF polynucleotides, polypeptides, or antagonists to reduce G-CSF-mediated death of T cells in HlV-infected patients.
  • the role of T cell apoptosis in the development of AIDS has been the subject of a number of studies (for example, Meyaard et al., Science, 257:217- 219 (1992); Groux et al., J. Exp. Med., 175:331 (1992); and Oyaizu et al, in Cell Activation and Apoptosis in HIV Infection, Andrieu and Lu, Eds., Plenum Press, New York, pp. 101-114 (1995)).
  • Fas-mediated apoptosis has been implicated in the loss of T cells in HTV positive individuals (Katsikis et al., J. Exp. Med. 181:2029-2036 (1995). It is also likely that T cell apoptosis occurs through multiple mechanisms.
  • Activated human T cells are induced to undergo programmed cell death (apoptosis) upon triggering through the CD3/T cell receptor complex, a process termed activated-induced cell death (AICD).
  • AICD activated-induced cell death
  • AICD may play a role in the depletion of CD4+ T cells and the progression to AIDS in HiV-infected individuals.
  • the present invention provides a method of inhibiting G-CSF- mediated T cell death in HIV patients, comprising administering G-CSF polynucleotides, polypeptides, or antagonists of the invention to the patients.
  • the patient is asymptomatic when treatment with G-CSF polynucleotides, polypeptides, or antagonists commences.
  • peripheral blood T cells may be extracted from an HIV patient, and tested for susceptibility to G-CSF-mediated cell death by procedures known in the art.
  • a patient's blood or plasma is contacted with G-CSF antagonists, anti-G-CSF antibodies, of the invention ex vivo.
  • the G-CSF antagonists may be bound to a suitable chromatography matrix by procedures known in the art.
  • the patient's blood or plasma flows through a chromatography column containing G-CSF antagonist bound to the matrix, before being returned to the patient.
  • the immobilized G-CSF antagonist binds G-CSF, thus removing G-CSF protein from the patient's blood.
  • an G-CSF polynucleotide, polypeptide, or antagonist of the invention is administered in combination with other inhibitors of T cell apoptosis.
  • Fas-mediated apoptosis also has been implicated in loss of T cells in HIV individuals (Katsikis et al., J. Exp. Med., 181:2029- 2036 (1995)).
  • a patient susceptible to both Fas ligand mediated and G-CSF- mediated T cell death may be treated with both an agent that blocks G-CSF/G-CSF receptor interactions and an agent that blocks Fas-ligand/Fas interactions.
  • Suitable agents for blocking binding of Fas-ligand to Fas include, but are not limited to, soluble Fas polypeptides; multimeric forms of soluble Fas polypeptides (e.g., dimers of sFas/Fc); anti-Fas antibodies that bind Fas without transducing the biological signal that results in apoptosis; anti-Fas-ligand antibodies that block binding of Fas-ligand to Fas; and muteins of Fas-ligand that bind Fas but do not transduce the biological signal that results in apoptosis.
  • the antibodies employed according to this method are monoclonal antibodies. Examples of suitable agents for blocking Fas-ligand/Fas interactions, including blocking anti-Fas monoclonal antibodies, are described in WO 95/10540, hereby incorporated by reference.
  • agents which block binding of G-CSF to an G-CSF receptor are administered with the G-CSF polynucleotides, polypeptides, or antagonists of the invention.
  • agents include, but are not limited to, soluble G-CSF receptor polypeptides; multimeric forms of soluble G-CSF receptor polypeptides; and G-CSF receptor antibodies that bind the G-CSF receptor without transducing the biological signal that results in apoptosis, anti- G-CSF antibodies that block binding of G-CSF to one or more G-CSF receptors, and muteins of G-CSF that bind G-CSF receptors but do not transduce the biological signal that results in apoptosis.
  • G-CSF polypeptides of the invention may also be employed to regulate hematopoeisis and, in particular, erythropoiesis.
  • Hematopoeisis is a multi-step cell proliferation and differentiation process which begins with a pool of multipotent stem cells. These cells can proliferate and differentiate into hematopoietic progenitors in reply to different stimuli.
  • the G-CSF polypeptides of the invention, as well as agonists and antagonists thereof, may be used to either stimulate or inhibit development of hematopoietic cells and, in particular, erythropoietic precursor cells.
  • G-CSF may be used to treat B-cell deficiencies, including those arising in the context of infection, cancer treatment, transplant, immunodeficiency, immune disorder, agammaglobulinemia, hypogammaglobulinemia, defects in B cell development, defects in B cell function, defects in B-cell regulation, and shortened B cell lifespan.
  • IL-7 is one of the factors involved in mediating B cell development (Stoddart et al., Immunol. Rev. 175:47-58 (2000)).
  • G-CSF may be used to treat Bruton agammaglobulinemia, an X chromosome-linked agammaglobulinemia conventionally understood as a life-threatening disease that involves a failure in normal development of B lymphocytes and is associated with missense mutations in BTK, a gene encoding a cytoplasmic tyrosine kinase (Bruton agammaglobulinemia tyrosine kinase, EC 2.7.1.112), a member of the Tec family of protein-tyrosine kinases (Ohta et al., Proc. Natl. Acad. Sci. 91:9062-9066 (1994).
  • Bruton agammaglobulinemia an X chromosome-linked agammaglobulinemia conventionally understood as a life-threatening disease that involves a failure in normal development of B lymphocytes and is associated with missense mutations in BTK
  • G-CSF may be used to treat platelet deficiencies, such as manifest by thrombocytopenia, for example, immune-mediated thrombocytopenia. It can stimulate the growth and proliferation of lymphocytes, and can promote angiogenesis. G-CSF is useful in the treatment of diabetes. It can modulate the immune response of an organism, can treat allergies, and can be used to treat and/or prevent infection. G-CSF can inhibit tumor growth and can be used to treat cancer.
  • molecules of the invention may be employed as agents to boost immunoresponsiveness among individuals having a temporary immune deficiency.
  • Conditions resulting in a temporary immune deficiency that may be ameliorated or treated by administering the G-CSF polypeptides or polynucleotides of the invention, or agonists thereof, include, but are not limited to, recovery from viral infections (e.g., influenza), conditions associated with malnutrition, recovery from infectious mononucleosis, or conditions associated with stress, recovery from measles, recovery from blood transfusion, and recovery from surgery.
  • the term "treating" includes any or all of preventing replication of cells associated with an autoimmune disease state including, but not limited to, cells capable of producing an autoimmune antibody, lessening the autoimmune-antibody burden, and ameliorating one or more symptoms of an autoimmune disease.
  • G-CSF polynucleotides or polypeptides of the invention may be used to diagnose, prognose, treat, or prevent one or more of the following diseases or disorders, or conditions associated therewith: primary immunodeficiencies, Kawasaki syndrome, bone marrow transplant (e.g., recent bone marrow transplant in adults or children), chronic B-cell lymphocytic leukemia, chronic inflammatory demyelinating polyneuropathy, and post-transfusion purpura, anemia (e.g., anemia associated with parvovirus B19, patients with stable multiple myeloma who are at high risk for infection (e.g., recurrent infection), autoimmune hemolytic anemia (e.g., warm-type autoimmune hemolytic anemia), thrombocytopenia (e.g., neonatal thrombocytopenia), and immune-mediated neutropenia), transplantation (e.g., cytomegalovirus (CMV)-negative recipients of CMV
  • CMV cytomegalovirus
  • Autoimmune disorders and conditions associated with these disorders that may be treated, prevented, and/or diagnosed with the G-CSF polynucleotides, polypeptides, and/or antagonist of the invention (e.g., anti- G-CSF antibodies), include, but are not limited to, autoimmune hemolytic anemia, autoimmune neonatal thrombocytopenia, idiopathic thrombocytopenia purpura, autoimmunocytopenia, hemolytic anemia, antiphospholipid syndrome, dermatitis, allergic encephalomyelitis, myocarditis, relapsing polychondritis, rheumatic heart disease, glomerulonephritis (e.g., IgA nephropathy), multiple sclerosis, neuritis, uveitis ophthalmia, polyendocrinopathies, purpura (e.g., Henloch-Scoenlein purpura), Reiter's disease, stiff-man syndrome, autoimmune
  • autoimmune thyroiditis i.e., Hashimoto's thyroiditis
  • hypothyroidism i.e., Hashimoto's thyroiditis
  • systemic lupus erythematosus often characterized, e.g., by circulating and locally generated immune complexes
  • Goodpasture's syndrome characterized, e.g., by anti-basement membrane antibodies
  • pemphigus often characterized, e.g., by epidermal acantholytic antibodies
  • receptor autoimmunities such as, for example, (a) Graves' disease (often characterized, e.g., by TSH receptor antibodies), (b) myasthenia gravis (often characterized, e.g., by acetylcholine receptor antibodies), and (c) insulin resistance (often
  • Additional autoimmune disorders which may be treated, prevented, and/or diagnosed with the compositions of the invention include, but are not limited to, rheumatoid arthritis (often characterized, e.g., by immune complexes in joints), scleroderma with anti-collagen antibodies (often characterized, e.g., by nucleolar and other nuclear antibodies), mixed connective tissue disease (often characterized, e.g., by antibodies to extractable nuclear antigens (e.g., ribonucleoprotein)), polymyositis/dermatomyositis (often characterized, e.g., by nonhistone anti-nuclear antibodies), pernicious anemia (often characterized, e.g., by antibodies to parietal cells, microsomes, and intrinsic factor), idiopathic Addison's disease (often characterized, e.g., by humoral and cell-mediated adrenal cytotoxicity, infertility (often characterized,
  • compositions of the invention include, but are not limited to, chronic active hepatitis (often characterized, e.g. by smooth muscle antibodies), primary biliary cirrhosis (often characterized, e.g., by mitochondria!
  • antibodies other endocrine gland failure (often characterized, e.g., by specific tissue antibodies in some cases), vitiligo (often characterized, e.g., by melanocyte antibodies), vasculitis (often characterized, e.g., by Ig and complement in vessel walls and/or low serum complement), post-myocardial infarction (often characterized, e.g., by myocardial antibodies), cardiotomy syndrome (often characterized, e.g., by myocardial antibodies), urticaria (often characterized, e.g., by IgG and IgM antibodies to IgE), atopic dermatitis (often characterized, e.g., by IgG and IgM antibodies to IgE), asthma (often characterized, e.g., by IgG and IgM antibodies to IgE), inflammatory myopathies, and many other inflammatory, granulomatous, degenerative, and atrophic disorders.
  • G-CSF polynucleotides or polypeptides, or antagonists thereof are used to treat or prevent systemic lupus erythematosus and/or diseases, disorders or conditions associated therewith.
  • Lupus-associated diseases, disorders, or conditions that may be treated or prevented with G-CSF polynucleotides or polypeptides, or antagonists of the invention include, but are not limited to, hematologic disorders (e.g., hemolytic anemia, leukopenia, lymphopenia, and thrombocytopenia), immunologic disorders (e.g., anti-DNA antibodies, and anti-Sm antibodies), rashes, photosensitivity, oral ulcers, arthritis, fever, fatigue, weight loss, serositis (e.g., pleuritus (pleuricy)), renal disorders (e.g., nephritis), neurological disorders (e.g., seizures, peripheral neuropathy, CNS related disorders), gastrointestinal disorders, Raynaud phenomenon, and pericarditis.
  • hematologic disorders e.g., hemolytic anemia, leukopenia, lymphopenia, and thrombocytopenia
  • immunologic disorders e.g., anti-DNA antibodies, and anti-Sm
  • G-CSF polypeptides, agonists, or antagonists of the invention may be used to treat diseases associated with ischemia, e.g., cardiovascular disorders, including peripheral artery disease, such as limb ischemia. They may also include stroke, vascular disease, and fulminant liver failure.
  • ischemia e.g., cardiovascular disorders, including peripheral artery disease, such as limb ischemia. They may also include stroke, vascular disease, and fulminant liver failure.
  • the term "treating” includes any or all of preventing the growth, multiplication, or replication of the pathogen that causes the ischemic disease and ameliorating one or more symptoms of an ischemic disease.
  • G-CSF induces therapeutic stem cell homing to injured myocardium.
  • G-CSF has been observed to be up-regulated following myocardial infarction (Askari et al., supra).
  • the invention provides compositions and methods for providing G-CSF to the heart.
  • autologous cells genetically modified with molecules of the Sequence Listing can be transplanted to a subject that can benefit from such cells. Bone marrow stimulation may be performed in conjunction with the transplantation.
  • Cardiac fibroblasts and cardiac myoblasts can be transfected with G-CSF as described by Askari et al., supra, or other means known in the art, and provided in vivo by local administration.
  • a rodent model with a ligation of the left anterior descending artery is suitable for studying and practicing this method.
  • the method is suitable for therapeutic purposes in humans.
  • This method of promoting tissue regeneration may be accomplished by stem cell engraftment. It is applicable to the heart, and other organs, including the brain, pancreas, lung, liver, skin, and bone.
  • G-CSF can be delivered to the tissues by conventional local administration of a molecule described herein or a composition comprising such molecule.
  • G-CSF can also be delivered to the tissues by local administration of a cell comprising a molecule described herein or a composition comprising such molecule.
  • Suitable cells include heart cells, brain cells, pancreatic cells, lung cells, liver cells, skin cells, bone cells, mesenchymal stem cells, progenitor cells, adult stem cells, and embryonic stem cells.
  • Cardiovascular disorders include cardiovascular abnormalities, such as arterio-arterial fistula, arteriovenous fistula, cerebral arteriovenous malformations, congenital heart defects, pulmonary atresia and scimitar syndrome.
  • Congenital heart defects include aortic coarctation, cor triatriatum, coronary vessel anomalies, crisscross heart, dextrocardia, patent ductus arteriosus, Ebstein's anomaly, Eisenmenger complex, hypoplastic left heart syndrome, levocardia, tetralogy of Fallot, transposition of great vessels, double outlet right ventricle, tricuspid atresia, persistent truncus arteriosus, and heart septal defects, such as aortopulmonary septal defect, endocardial cushion defects, Lutembacher's syndrome, trilogy of Fallot, and ventricular heart septal defects.
  • Cardiovascular disorders also include heart disease, such as arrhythmias, carcinoid heart disease, high cardiac output, low cardiac output, cardiac tamponade, endocarditis (including bacterial), heart aneurysm, cardiac arrest, congestive heart failure, congestive cardiomyopathy, paroxysmal dyspnea, cardiac edema, heart hypertrophy, congestive cardiomyopathy, left ventricular hypertrophy, right ventricular hypertrophy, post-infarction heart rupture, ventricular septal rupture, heart valve diseases, myocardial diseases, myocardial ischemia, pericardial effusion, pericarditis (including constrictive and tuberculous), pneumopericardium, postpericardiotomy syndrome, pulmonary heart disease, rheumatic heart disease, ventricular dysfunction, hyperemia, cardiovascular pregnancy complications, scimitar syndrome, cardiovascular syphilis, and cardiovascular tuberculosis.
  • heart disease such as arrhythmias, carcinoid heart disease, high cardiac output, low cardiac
  • Arrhythmias include sinus arrhythmia, atrial fibrillation, atrial flutter, bradycardia, extrasystole, Adams-Stokes syndrome, bundle-branch block, sinoatrial block, long QT syndrome, parasystole, Lown-Ganong-Levine syndrome, Mahaim-type pre-excitation syndrome, Wolff-Parkinson- White syndrome, sick sinus syndrome, tachycardias, and ventricular fibrillation.
  • Tachycardias include paroxysmal tachycardia, supraventricular tachycardia, accelerated idioventricular rhythm, atrioventricular nodal reentry tachycardia, ectopic atrial tachycardia, ectopic junctional tachycardia, sinoatrial nodal reentry tachycardia, sinus tachycardia, Torsades de Pointes, and ventricular tachycardia.
  • Heart valve diseases include aortic valve insufficiency, aortic valve stenosis, heart murmurs, aortic valve prolapse, mitral valve prolapse, tricuspid valve prolapse, mitral valve insufficiency, mitral valve stenosis, pulmonary atresia, pulmonary valve insufficiency, pulmonary valve stenosis, tricuspid atresia, tricuspid valve insufficiency, and tricuspid valve stenosis.
  • Myocardial diseases include alcoholic cardiomyopathy, congestive cardiomyopathy, hypertrophic cardiomyopathy, aortic subvalvular stenosis, pulmonary subvalvular stenosis, restrictive cardiomyopathy, Chagas cardiomyopathy, endocardial fibroelastosis, endomyocardial fibrosis, Kearns syndrome, myocardial reperfusion injury, and myocarditis.
  • Myocardial ischemias include coronary disease, such as angina pectoris, coronary aneurysm, coronary arteriosclerosis, coronary thrombosis, coronary vasospasm, myocardial infarction and myocardial stunning.
  • coronary disease such as angina pectoris, coronary aneurysm, coronary arteriosclerosis, coronary thrombosis, coronary vasospasm, myocardial infarction and myocardial stunning.
  • Cardiovascular diseases also include vascular diseases such as aneurysms, angiodysplasia, angiomatosis, bacillary angiomatosis, Hippel-Lindau disease, Klippel-
  • Aneurysms include dissecting aneurysms, false aneurysms, infected aneurysms, ruptured aneurysms, aortic aneurysms, cerebral aneurysms, coronary aneurysms, heart aneurysms, and iliac aneurysms.
  • Arterial occlusive diseases include arteriosclerosis, intermittent claudication, carotid stenosis, fibromuscular dysplasias, mesenteric vascular occlusion, Moyamoya disease, renal artery obstruction, retinal artery occlusion, and thromboangitis obliterans.
  • Cerebrovascular disorders include carotid artery diseases, cerebral amyloid angiopathy, cerebral aneurysm, cerebral anoxia, cerebral arteriosclerosis, cerebral arteriovenous malformation, cerebral artery diseases, cerebral embolism and thrombosis, carotid artery thrombosis, sinus thrombosis, Wallenberg's syndrome, cerebral hemorrhage, epidural hematoma, subdural hematoma, subarachnoid hemorrhage, cerebral infarction, cerebral ischemia (including transient), subclavian steal syndrome, periventricular leukomalacia, vascular headache, cluster headache, migraine, and vertebrobasilar insufficiency.
  • Embolisms include air embolisms, amniotic fluid embolisms, cholesterol embolisms, blue toe syndrome, fat embolisms, pulmonary embolisms, and thromboembolisms.
  • Thromboses include coronary thrombosis, hepatic vein thrombosis, retinal vein occlusion, carotid artery thrombosis, sinus thrombosis, Wallenberg's syndrome, and thrombophlebitis.
  • Ischemia includes cerebral ischemia, ischemic colitis, compartment syndromes, anterior compartment syndrome, myocardial ischemia, reperfusion injuries, and peripheral limb ischemia.
  • Vasculitis includes aortitis, arteritis, Behcet's syndrome, Churg-Strauss syndrome, mucocutaneous lymph node syndrome, thromboangiitis obliterans, hypersensitivity vasculitis, Schoenlein-Henoch purpura, allergic cutaneous vasculitis, and Wegener's granulomatosis.
  • ocular disorders associated with neovascularization which can be treated with the G-CSF polynucleotides and polypeptides of the present invention (including G-CSF agonists and G-CSF antagonists) include, but are not limited to neovascular glaucoma, diabetic retinopathy, retinoblastoma, retrolental fibroplasia, uveitis, retinopathy of prematurity, macular degeneration, corneal graft neovascularization, as well as other eye inflammatory diseases, ocular tumors, and diseases associated with choroidal or iris neovascularization. See, e.g., reviews by Waltman et al., Am. J.
  • disorders which can be treated with the G-CSF polynucleotides and polypeptides of the present invention include, but are not limited to, hemangioma, arthritis, psoriasis, angiofibroma, atherosclerotic plaques, delayed wound healing, granulations, hemophilic joints, hypertrophic scars, nonunion fractures, Osier- Weber syndrome, pyogenic granuloma, scleroderma, trachoma, and vascular adhesions.
  • Polynucleotides and/or polypeptides of the invention, and/or agonists and/or antagonists thereof, are useful in the diagnosis and treatment or prevention of a wide range of diseases and/or conditions.
  • diseases and conditions include, but are not limited to, cancer (e.g., immune cell related cancers, breast cancer, prostate cancer, ovarian cancer, follicular lymphoma, cancer associated with mutation or alteration of p53, brain tumor, bladder cancer, uterocervical cancer, colon cancer, colorectal cancer, non-small cell carcinoma of the lung, small cell carcinoma of the lung, stomach cancer, etc.), lymphoproliferative disorders (e.g., lymphadenopathy), microbial (e.g., viral, bacterial, etc.) infection (e.g., HIV-1 infection, HIV-2 infection, herpes virus infection (including, but not limited to, HSV-1, HSV-2, CMV, VZV, HHV-6, HHV-7, EBV), adenovirus infection
  • autoimmune disease e.g., multiple sclerosis, rheumatoid arthritis, systemic lupus erythematosus, immune complex glomerulonephritis, autoimmune diabetes, autoimmune thrombocytopenic purpura, Grave's disease, Hashimoto's thyroiditis, etc.
  • cardiomyopathy e.g., dilated cardiomyopathy
  • diabetes diabetic complications (e.g., diabetic nephropathy, diabetic neuropathy, diabetic retinopathy), influenza, asthma, psoriasis, glomerulonephritis, septic shock, and ulcerative colitis.
  • Polynucleotides and/or polypeptides of the invention and/or agonists and/or antagonists thereof are also useful as an adjuvant to enhance immune responsiveness to specific antigen and/or anti- viral immune responses.
  • polynucleotides and/or polypeptides of the invention and/or agonists and/or antagonists thereof are useful in regulating (i.e., elevating or reducing) the immune response.
  • polynucleotides and/or polypeptides of the invention may be useful in preparation or recovery from surgery, trauma, radiation therapy, chemotherapy, and transplantation, or may be used to boost immune response and/or recovery in the elderly and immunocompromised individuals.
  • polynucleotides and/or polypeptides of the invention and/or agonists and/or antagonists thereof are useful as immunosuppressive agents, for example in the treatment or prevention of autoimmune disorders.
  • polynucleotides and/or polypeptides of the invention are used to treat or prevent chronic inflammatory, allergic or autoimmune conditions, such as those described herein or otherwise known in the art.
  • the uses of the G-CSF polypeptides include, but are not limited to, the treatment or prevention of viral hepatitis, herpes viral infections, allergic reactions, adult respiratory distress syndrome, neoplasia, anaphylaxis, allergic asthma, allergen rhinitis, drug allergies (e.g., to penicillin, cephalosporins), primary central nervous system lymphoma (PCNSL), glioblastoma, chronic lymphocytic leukemia (CLL), lymphadenopathy, autoimmune disease, graft versus host disease, rheumatoid arthritis, osteoarthritis, Graves' disease, acute lymphoblastic leukemia (ALL), lymphomas (Hodgkin's disease and non-Hodgkin's lymphoma (
  • G-CSF polypeptides of the present invention may be employed to inhibit neoplasia, such as tumor cell growth.
  • the combination of G-CSF protein with immunotherapeutic agents such as IL-2 or IL-12 may result in synergistic or additive effects that would be useful for the treatment of established cancers.
  • Antibodies [0207] G-CSF-protein specific antibodies for use in the present invention can be raised against the intact G-CSF protein or an antigenic polypeptide fragment thereof, for example a polypeptide fragment of approximately six or more amino acids.
  • the protein or fragment may be presented with or without a carrier protein, such as an albumin, to an animal system (such as rabbit or mouse); in general the polypeptide fragments are sufficiently immunogenic to produce a satisfactory immune response without a carrier if they are at least about 25 amino acids in length.
  • a carrier protein such as an albumin
  • Antibodies of the invention include polyclonal and monoclonal antibody preparations, as well as preparations including hybrid antibodies, altered antibodies, chimeric antibodies and, humanized antibodies, as well as: hybrid (chimeric) antibody molecules (see, for example, Winter et al., Nature 349:293-299 (1991)); and U.S. Patent No. 4,816,567); F(ab') 2 and F(ab) fragments; Fv molecules (noncovalent heterodimers, see, for example, Inbar et al., Proc. Natl. Acad. Sci. 69:2659-2662 (1972)); and Ehrlich et al.
  • the invention also provides monoclonal antibodies and G-CSF protein binding fragments thereof.
  • Methods of making monoclonal and polyclonal antibodies are known in the art.
  • Monoclonal antibodies are generally antibodies having a homogeneous antibody population. The term is not limited regarding the species or source of the antibody, nor is it intended to be limited by the manner in which it is made. The term encompasses whole immunoglobulins.
  • Monoclonal antibodies of the invention can be prepared using hybridoma technology, for example, Kohler et al., Nature, 256:495 (1975); Kohler et al., Eur. J. Immunol, 6:511 (1976); Kohler et. al., Eur. J.
  • Such cells may be cultured in any suitable tissue culture medium; e.g., in Earle's modified Eagle's medium supplemented with 10% fetal bovine serum (inactivated at about 56°C), and supplemented with about 10 grams/liter of nonessential amino acids, about 1,000 U/ml of penicillin, and about 100 ⁇ g/ml of streptomycin.
  • the splenocytes of such mice are extracted and fused with a suitable myeloma cell line.
  • Any suitable myeloma cell line may be employed in accordance with the present invention; e.g., the parent myeloma cell line (SP20), available from the American Type Culture Collection, Manassas, VA. After fusion, the resulting hybridoma cells are selectively maintained in HAT medium, and then cloned by limiting dilution as described by Wands et al., Gastroenterology, 80:225-232 (1981).
  • Polyclonal antibodies are generated by immunizing a suitable animal, such as a mouse, rat, rabbit, sheep or goat, with an antigen of interest, such as a stem cell transformed with a gene encoding an antigen.
  • an antigen of interest such as a stem cell transformed with a gene encoding an antigen.
  • the antigen can be linked to a carrier prior to immunization.
  • Suitable carriers are typically large, slowly metabolized macromolecules such as proteins, polysaccharides, polylactic acids, polyglycolic acids, polymeric amino acids, amino acid copolymers, lipid aggregates (such as oil droplets or liposomes), and inactive virus particles.
  • the antigen may be conjugated to a bacterial toxoid, such as toxoid from diphtheria, tetanus, cholera, etc., in order to enhance the immunogenicity thereof.
  • Chimeric antibodies i.e., antibodies in which different portions are derived from different animal species, such as those having a variable region derived from a murine monoclonal antibody and a human immunoglobulin constant region, for example, humanized antibodies, and insertion/deletions relating to cdr and framework regions, re suitable for use in the invention.
  • the invention also includes humanized antibodies, i.e., those with mostly human immunoglobulin sequences.
  • Humanized antibodies of the invention generally refer to non-human immunoglobulins that have been modified to incorporate portions of human sequences.
  • a humanized antibody may include a human antibody that contains entirely human immunoglobulin sequences.
  • the antibodies of the invention may be prepared by any of a variety of methods.
  • cells expressing the G-CSF protein or an antigenic fragment thereof can be administered to an animal in order to induce the production of sera containing polyclonal antibodies.
  • a preparation of G-CSF protein can be prepared and purified to render it substantially free of natural contaminants, and the preparation introduced into an animal in order to produce polyclonal antisera with specific binding activity.
  • Antibodies of the invention specifically bind to their respective antigen(s); they may display high avidity and/or high affinity to a specific polypeptide, or more accurately, to an epitope of an antigen. Antibodies of the invention may bind to one epitope, or to more than one epitope. They may display different affinities and/or avidities to different epitopes on one or more molecules. When an antibody binds more strongly to one epitope than to another, adjusting the binding conditions can, in some instances, result in antibody binding almost exclusively to the specific epitope and not to any other epitopes on the same polypeptide, and not to a polypeptide that does not comprise the epitope.
  • G-CSF Protein Antigen Alternatively, antibodies capable of binding to the G-CSF protein antigen may be produced in a two-step procedure through the use of anti-idiotypic antibodies. Such a method makes use of the fact that antibodies are themselves antigens, and that, therefore, it is possible to obtain an antibody which binds to a second antibody.
  • G-CSF -protein specific antibodies are used to immunize an animal, e.g., a mouse. The splenocytes of such an animal are then used to produce hybridoma cells, and the hybridoma cells are screened to identify clones which produce an antibody whose ability to bind to the G-CSF protein-specific antibody can be blocked by the G-CSF protein antigen.
  • Such antibodies comprise anti-idiotypic antibodies to the G-CSF protein-specific antibody and can be used to immunize an animal to induce formation of further G-CSF protein-specific antibodies.
  • Fab and F(ab')2 and other fragments of the antibodies of the present invention may be used according to the methods disclosed herein. Such fragments are typically produced by proteolytic cleavage, using enzymes such as papain (to produce Fab fragments) or pepsin (to produce F(ab')2 fragments). Alternatively, G-CSF protein-binding fragments can be produced through the application of recombinant DNA technology or through synthetic chemistry. Humanized chimeric monoclonal antibodies are suitable for in vivo use of anti-G-CSF in humans. Such humanized antibodies can be produced using genetic constructs derived from hybridoma cells producing the monoclonal antibodies described above.
  • This invention is also related to the use of the genes of the present invention as part of a diagnostic assay for detecting diseases or susceptibility to diseases related to the presence of mutations in the nucleic acid sequences encoding the polypeptide of the present invention.
  • Individuals carrying mutations in a gene of the present invention may be detected at the DNA level by a variety of techniques. Nucleic acids for diagnosis may be obtained from a patient's cells, such 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 PCR, for example, as described by Saiki et al., Nature, 324: 163-166 (1986), prior to analysis.
  • RNA or cDNA may also be used for the same purpose.
  • PCR primers complementary to the nucleic acid encoding a polypeptide of the present invention can be used to identify and analyze mutations. For example, deletions and insertions can be detected by a change in size of the amplified product in comparison to the normal genotype. Point mutations can be identified by hybridizing amplified DNA to radiolabeled RNA or alternatively, radiolabeled antisense DNA sequences. Perfectly matched sequences can be distinguished from mismatched duplexes by RNase A digestion or by differences in melting temperatures. [0218] Genetic testing based on DNA sequence differences may be achieved by detecting alterations in electrophoretic mobility of DNA fragments in gels run with or without denaturing agents.
  • DNA fragments of different sequences may be distinguished on denaturing formamide 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, for example, as described by Myers et al., Science, 230:1242 (1985).
  • Sequence changes at specific locations may also be revealed by nuclease protection assays, such as RNase and SI protection or the chemical cleavage method as shown in Cotton et al., Proc. Natl. Acad. Set, USA, 85:4397-4401 (1985).
  • nuclease protection assays such as RNase and SI protection or the chemical cleavage method as shown in Cotton et al., Proc. Natl. Acad. Set, USA, 85:4397-4401 (1985).
  • 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.
  • restriction enzymes e.g., Restriction Fragment Length Polymorphisms (RFLP)
  • RFLP Restriction Fragment Length Polymorphisms
  • the present invention also relates to a diagnostic assay for detecting altered levels of G-CSF proteins in various tissues.
  • An over-expression of these proteins compared to normal control tissue samples may detect the presence of abnormal cellular proliferation, for example, a tumor.
  • Assays used to detect protein levels in a host-derived sample are well-known to those of skill in the art and include radioimmunoassays, competitive-binding assays, Western Blot analysis, ELISA assays, "sandwich” assays, and other assays for the expression levels of the genes encoding the G-CSF proteins known in the art.
  • Expression can be assayed by qualitatively or quantitatively measuring or estimating the level of G-CSF protein, or the level of mRNA encoding G-CSF protein, in a biological sample.
  • Assays may be performed directly, for example, by determining or estimating absolute protein level or mRNA level, or relatively, by comparing the G- CSF protein or mRNA to a second biological sample.
  • the G- CSF protein or mRNA level in the first biological sample is measured or estimated and compared to a standard G-CSF protein level or mRNA level; suitable standards include second biological samples obtained from an individual not having the disorder of interest.
  • Standards may be obtained by averaging levels of G-CSF in a population of individuals not having a disorder related to G-CSF expression. As will be appreciated in the art, once a standard G-CSF protein level or mRNA level is known, it can be used repeatedly as a standard for comparison.
  • An ELISA assay for example, as described by Coligan, et al., Current Protocols in Immunology, 1(2), Chap. 6, (1991), utilizes an antibody prepared with specificity to a polypeptide antigen of the present invention.
  • a reporter antibody is prepared against the monoclonal antibody.
  • a detectable reagent such as a radioactive tag, a fluorescent tag, or an enzymatic tag, e.g., a horseradish peroxidase.
  • a sample is removed from a host and incubated on a solid support, e.g. a polystyrene dish, that binds the proteins in the sample.
  • any free protein binding sites on the dish are then covered by incubating with a non-specific protein, e.g., bovine serum albumin.
  • a non-specific protein e.g., bovine serum albumin.
  • the specific antibody e.g., a monoclonal antibody
  • the reporter antibody i.e., one linked to horseradish peroxidase is placed in the dish, resulting in the binding of the reporter antibody to any antibody bound to the protein of interest; unattached reporter antibody is then removed.
  • Substrate e.g., peroxidase
  • the amount of signal produced color e.g., developed in a given time period provides a measurement of the amount of a polypeptide of the present invention present in a given volume of patient sample when compared against a standard.
  • a competition assay may be employed wherein antibodies specific to a polypeptide of the present invention are attached to a solid support, and labeled G-CSF, along with a sample derived from the host, are passed over the solid support.
  • the label can be detected and quantified, for example, by liquid scintillation chromatography, and the measurement can be correlated to the quantity of the polypeptide of interest present in the sample.
  • a "sandwich" assay similar to an ELISA assay, may be employed, wherein a polypeptide of the present invention is passed over a solid support and binds to antibody modules attached to the solid support. A second antibody is then bound to the polypeptide of interest. A third antibody, which is labeled and specific to the second antibody is then passed over the solid support and binds to the second antibody. The amount of antibody binding can be quantified; it correlates with the amount of the polypeptide of interest.
  • Biological samples of the invention can include any biological sample obtained from a subject, body fluid, cell line, tissue culture, or other source which contains G-CSF protein or mRNA.
  • biological samples include body fluids (such as sera, plasma, urine, synovial fluid, and spinal fluid) which contain free G-CSF protein, ovarian or renal system tissue, and other tissue sources found to express complete or mature G-CSF polypeptide or an G-CSF receptor. Methods for obtaining tissue biopsies and body fluids from mammals are well known in the art. Where the biological sample is to include mRNA, a tissue biopsy may provide the source.
  • Total cellular RNA can be isolated from a biological sample using any suitable technique such as the single-step guanidinium-thiocyanate-phenol-chloroform method described in Chomczynski and Sacchi, Anal. Biochem., 162:156-159 (1987). Levels of mRNA encoding the G-CSF protein are then assayed using any appropriate method. These include Northern blot analysis, S 1 nuclease mapping, PCR, reverse transcription in combination with PCR (RT-PCR), and reverse transcription in combination with the ligase chain reaction (RT-LCR).
  • any suitable technique such as the single-step guanidinium-thiocyanate-phenol-chloroform method described in Chomczynski and Sacchi, Anal. Biochem., 162:156-159 (1987).
  • Levels of mRNA encoding the G-CSF protein are then assayed using any appropriate method. These include Northern blot analysis, S 1 nuclease mapping, PCR, reverse transcription in combination with
  • Assaying G-CSF protein levels in a biological sample can be performed using antibody-based techniques.
  • G-CSF protein expression in tissues can be studied with classical immunohistological methods, for example, Jalkanen, M., et al., J. Cell. Biol, 101:976-985 (1985); Jalkanen, M., et al., J. Cell. Biol, 105:3087-3096 (1987).
  • Other antibody-based methods useful for detecting G-CSF protein gene expression include immunoassays, such as the enzyme linked immunosorbent assay (ELISA) and the radioimmunoassay (RIA).
  • ELISA enzyme linked immunosorbent assay
  • RIA radioimmunoassay
  • Suitable antibody assay labels are known in the art and include enzyme labels, such as glucose oxidase, radioisotopes, and fluorescent labels, such as fluorescein and rhodamine, and biotin.
  • G-CSF protein can also be detected in vivo by imaging.
  • Antibody labels or markers for in vivo imaging of G-CSF protein include those detectable by X- radiography, NMR, or ESR.
  • suitable labels include radioisotopes such as barium or cesium, which emit detectable radiation but are not overtly harmful to a subject.
  • suitable markers for NMR and ESR include those with a detectable characteristic spin, such as deuterium, which may be incorporated into the antibody by labeling of nutrients for the relevant hybridoma.
  • G-CSF protein-specific antibody or antibody fragment which has been labeled with an appropriate detectable imaging moiety, such as a radioisotope, a radio- opaque substance, or a material detectable by nuclear magnetic resonance, is introduced, for example, parenterally, subcutaneously or intraperitoneally, into the subject to be examined for an immune system disorder. It will be understood in the art that the size of the subject and the imaging system used will determine the quantity of imaging moiety needed to produce diagnostic images. The labeled antibody or antibody fragment will then preferentially accumulate at the location of cells which contain G-CSF protein.
  • an appropriate detectable imaging moiety such as a radioisotope, a radio- opaque substance, or a material detectable by nuclear magnetic resonance
  • G-CSF polypeptide compositions will be formulated and dosed in a fashion consistent with good medical practice, taking into account the clinical condition of the individual subject, the site of delivery of the G-CSF polypeptide composition, the method of administration, the scheduling of administration, and other factors known to practitioners.
  • the effective amount of G-CSF polypeptide for purposes herein is thus determined by such considerations.
  • the polypeptides, agonists, and antagonists of the present invention may be employed in combination with a suitable pharmaceutical carrier to comprise a pharmaceutical composition for parenteral administration.
  • a suitable pharmaceutical carrier to comprise a pharmaceutical composition for parenteral administration.
  • Such compositions comprise a therapeutically effective amount of the polypeptide, agonist, or antagonist and a pharmaceutically acceptable carrier or excipient.
  • 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(s) can be a notice in the form prescribed by a governmental agency regulating the manufacture, use or sale of pharmaceuticals or biological products, which notice reflects approval by the agency of manufacture, use or sale for human administration.
  • the polypeptides, agonists and antagonists of the present invention may be employed in conjunction with other therapeutic compounds.
  • compositions of the invention can be administered in the form of their pharmaceutically acceptable salts, or they can also be used alone or in appropriate association, as well as in combination, with other pharmaceutically active compounds.
  • the subject compositions are formulated in accordance to the mode of potential administration. Administration of the agents can be achieved in various ways, including oral, buccal, nasal, rectal, parenteral, intraperitoneal, intradermal, transdermal, subcutaneous, intravenous, intra-arterial, intracardiac, intraventricular, intracranial, intratracheal, and intrathecal administration, etc., or otherwise by implantation or inhalation.
  • compositions can be formulated into preparations in solid, semi-solid, liquid or gaseous forms, such as tablets, capsules, powders, granules, ointments, solutions, suppositories, enemas, injections, inhalants and aerosols.
  • the following methods and excipients are merely exemplary and are in no way limiting. In general, they are administered in an amount of at least about 10 micrograms/kg body weight and in most cases they will be administered in an amount not in excess of about 8 milligrams/kg body weight per day.
  • polypeptides of the invention and agonist and antagonist compounds which are polypeptides, may also be employed in accordance with the present invention by expression of such polypeptides in vivo, i.e., gene therapy.
  • cells may be engineered with a polynucleotide (DNA or RNA) encoding for the polypeptide ex vivo; the engineered cells are then provided to a patient.
  • a polynucleotide DNA or RNA
  • RNA polynucleotide
  • cells may be engineered by procedures known in the art by use of a retroviral particle containing RNA encoding for the polypeptide of the present invention.
  • cells may be engineered in vivo for expressing the polypeptide in vivo, for example, by procedures known in the art.
  • a cell producing a retroviral particle containing RNA encoding the polypeptide of the present invention may be administered to a patient for the purpose of engineering cells in vivo and expressing the polypeptide in vivo.
  • the expression vehicle for engineering cells may be other than a retroviral particle, for example, an adenovirus, which may be used to engineer cells in vivo after combination with a suitable delivery vehicle.
  • Retroviruses from which the retroviral plasmid vectors hereinabove mentioned may be derived include, but are not limited to, Moloney Murine Leukemia virus, spleen necrosis virus, retroviruses such as Rous sarcoma virus, Harvey sarcoma virus, avian leukosis virus, gibbon ape leukemia virus, human immunodeficiency virus, adenovirus, myeloproliferative sarcoma virus, and mammary tumor virus.
  • the retroviral plasmid vector is derived from Moloney murine leukemia virus.
  • Promoters suitable for these formulations include the viral vectors listed above, as well as include the respiratory syncytial virus (RSV) promoter; inducible promoters, such as the MMT promoter, the metallothionein promoter; heat shock promoters; the albumin promoter; the ApoAl promoter; human globin promoters; viral thymidine kinase promoters, such as the Herpes Simplex thymidine kinase promoter; retroviral LTRs (including the modified retroviral LTRs hereinabove described); the beta- actin promoter; and human growth hormone promoters.
  • the promoter also may be the native promoter which controls the gene encoding the polypeptide. The selection of a suitable promoter will be apparent to those skilled in the art from the teachings contained herein.
  • a retroviral plasmid vector can be employed to transduce packaging cell lines to form producer cell lines.
  • packaging cells which may be transfected include, but are not limited to, the PE501, PA317, -2, -AM, PA12, T19-14X, VT-19-17- H2, CRE, CRIP, GP+E-86, GP+envAml2, and DAN cell lines as described in Miller, Human Gene Therapy, 1:5-14 (1990).
  • the vector may transduce the packaging cells through any means known in the art. Such means include, but are not limited to, electroporation, the use of liposomes, and CaPO 4 precipitation.
  • the retroviral plasmid vector may be encapsulated into a liposome, or coupled to a lipid, and then administered to a host.
  • the producer cell line generates infectious retroviral vector particles which include the nucleic acid sequence(s) encoding the polypeptides. Such retroviral vector particles then may be employed to transduce eukaryotic cells, either in vitro or in vivo.
  • the transduced eukaryotic cells will express the nucleic acid sequence(s) encoding the polypeptide.
  • Eukaryotic cells which may be transduced include, but are not limited to, embryonic stem cells, embryonic carcinoma cells, as well as hematopoietic stem cells, hepatocytes, fibroblasts, myoblasts, keratinocytes, endothelial cells, and bronchial epithelial cells.
  • G-CSF G-CSF "Knock-outs" and Homologous Recombination
  • Endogenous gene expression can be reduced by inactivating or "knocking out” a gene of interest and/or its promoter using targeted homologous recombination, (e.g., see Smithies et al., Nature, 317:230-234 (1985); Thomas & Capecchi, Cell, 51:503- 512 (1987); Thompson et al., Cell, 5:313-321 (1989); each of which is incorporated by reference herein in its entirety).
  • targeted homologous recombination e.g., see Smithies et al., Nature, 317:230-234 (1985); Thomas & Capecchi, Cell, 51:503- 512 (1987); Thompson et al., Cell, 5:313-321 (1989); each of which is incorporated by reference herein in its entirety).
  • a mutant, non-functional polynucleotide of the invention flanked by DNA homologous to the endogenous polynucleotide sequence (either the coding regions or regulatory regions of the gene) can be used, with or without a selectable marker and/or a negative selectable marker, to transfect cells that express polypeptides of the invention in vivo.
  • techniques known in the art are used to generate knockouts in cells that contain, but do not express, the gene of interest, insertion of the DNA construct, via targeted homologous recombination, results in inactivation of the targeted gene.
  • Such cells may be obtained from the patient (i.e., animal, including human) or an MHC compatible donor and can include, but are not limited to fibroblasts, bone marrow cells, blood cells (e.g., lymphocytes), adipocytes, muscle cells, endothelial cells, etc.
  • the cells are genetically engineered in vitro using recombinant DNA techniques to introduce the coding sequence of polypeptides of the invention into the cells, or alternatively, to disrupt the coding sequence and/or endogenous regulatory sequence associated with the polypeptides of the invention, e.g., by transduction (using viral vectors, and/or vectors that integrate the transgene into the cell genome) or transfection procedures, including, but not limited to, the use of plasmids, cosmids, YACs, naked DNA, electroporation, liposomes, etc.
  • the coding sequence of the polypeptides of the invention can be placed under the control of a strong constitutive or inducible promoter or promoter/enhancer to achieve expression, and secretion, of the polypeptides of the invention.
  • the engineered cells which express and secrete the polypeptides of the invention can be introduced into the patient systemically, e.g., in the circulation, or intraperitoneally.
  • the cells can be incorporated into a matrix and implanted in the body, e.g., genetically engineered fibroblasts can be implanted as part of a skin graft; genetically engineered endothelial cells can be implanted as part of a lymphatic or vascular graft.
  • genetically engineered fibroblasts can be implanted as part of a skin graft
  • genetically engineered endothelial cells can be implanted as part of a lymphatic or vascular graft.
  • the cells to be administered are non-autologous or non-MHC compatible cells, they can be administered using well known techniques which prevent the development of a host immune response against the introduced cells.
  • the cells may be introduced in an encapsulated form which, while allowing for an exchange of components with the immediate extracellular environment, does not allow the introduced cells to be recognized by the host immune system.
  • Transgenic Non-Human Animals [0241] The polypeptides of the invention can also be expressed in transgenic animals.
  • mice Animals of any species, including, but not limited to, mice, rats, rabbits, hamsters, guinea pigs, pigs, micro-pigs, goats, sheep, cows, and non-human primates, e.g., baboons, monkeys, and chimpanzees may be used to generate transgenic animals.
  • techniques described herein or otherwise known in the art are used to express polypeptides of the invention in humans, as part of a gene therapy protocol.
  • transgene i.e., polynucleotides of the invention
  • transgene i.e., polynucleotides of the invention
  • Such techniques include, but are not limited to, pronuclear microinjection (Paterson et al., Appl Microbiol. Biotechnol. 40:691-698 (1994); Carver et al., Biotechnology (NY) 11: 1263-1270 (1993); Wright et al., Biotechnology (NY) 9:830-834 (1991); and Hoppe et al., U.S. Pat. No. 4,873,191 (1989)); retrovirus mediated gene transfer into germ lines (Van der Putten et al., Proc.
  • transgenic clones containing polynucleotides of the invention for example, nuclear transfer into enucleated oocytes of nuclei from cultured embryonic, fetal, or adult cells induced to quiescence (Campbell et al., Nature 380:64-66 (1996); Wilmut et al., Nature 385:810-813 (1997)), each of which is herein incorporated by reference in its entirety).
  • the present invention provides for transgenic animals that carry the transgene in all their cells, as well as animals which carry the transgene in some, but not all their cells, i.e., mosaic or chimeric animals.
  • the transgene may be integrated as a single transgene or as multiple copies such as in concatamers, e.g., head-to-head tandems or head-to-tail tandems.
  • the transgene may also be selectively introduced into and activated in a particular cell type by following, for example, the teaching of Lasko et al. (Proc. Natl. Acad. Sci. USA 89:6232-6236 (1992)).
  • the regulatory sequences required for such a cell-type specific activation will depend upon the particular cell type of interest, and will be apparent to those of skill in the art. It may be desired that the polynucleotide transgene be integrated into the chromosomal site of the endogenous gene, gene targeting is then suitable. Briefly, when such a technique is to be utilized, vectors containing some nucleotide sequences homologous to the endogenous gene are designed for the purpose of integrating, via homologous recombination with chromosomal sequences, into and disrupting the function of the nucleotide sequence of the endogenous gene.
  • the transgene may also be selectively introduced into a particular cell type, thus inactivating the endogenous gene in only that cell type, by following, for example, the teaching of Gu et al. (Science 265:103-106 (1994)).
  • the regulatory sequences required for such a cell-type specific inactivation will depend upon the particular cell type of interest, and will be apparent to those of skill in the art.
  • the expression of the recombinant gene may be assayed utilizing standard techniques. Initial screening may be accomplished by Southern blot analysis or PCR techniques to analyze animal tissues to verify that integration of the transgene has taken place.
  • the level of mRNA expression of the transgene in the tissues of the transgenic animals may also be assessed using techniques which include, but are not limited to, Northern blot analysis of tissue samples obtained from the animal, in situ hybridization analysis, and reverse transcriptase-PCR (rt-PCR). Samples of transgenic gene-expressing tissue may also be evaluated immunocytochemically or immunohistochemically using antibodies specific for the transgene product.
  • founder animals may be bred, inbred, outbred, or crossbred to produce colonies of the particular animal.
  • breeding strategies include, but are not limited to outbreeding of founder animals with more than one integration site in order to establish separate lines; inbreeding of separate lines in order to produce compound transgenics that express the transgene at higher levels because of the effects of additive expression of each transgene; crossing of heterozygous transgenic animals to produce animals homozygous for a given integration site in order to both augment expression and eliminate the need for screening of animals by DNA analysis; crossing of separate homozygous lines to produce compound heterozygous or homozygous lines; and breeding to place the transgene on a distinct background that is appropriate for an experimental model of interest.
  • kits that can be used in the above methods.
  • a kit comprises an antibody of the invention, e.g., a purified antibody, in one or more containers.
  • the kits of the present invention contain a substantially isolated polypeptide comprising an epitope which is specifically immunoreactive with an antibody included in the kit.
  • the kits of the present invention may also comprise a control antibody which does not react with the polypeptide of interest.
  • kits of the present invention comprise a means for detecting the binding of an antibody to a polypeptide of interest (e.g., the antibody may be conjugated to a detectable substrate such as a fluorescent compound, an enzymatic substrate, a radioactive compound or a luminescent compound, or a second antibody which recognizes the first antibody may be conjugated to a detectable substrate).
  • a detectable substrate such as a fluorescent compound, an enzymatic substrate, a radioactive compound or a luminescent compound, or a second antibody which recognizes the first antibody may be conjugated to a detectable substrate.
  • the kit is a diagnostic kit for use in screening serum containing antibodies specific against proliferative and/or cancerous polynucleotides and polypeptides.
  • a kit may include a control antibody that does not react with the polypeptide of interest.
  • a kit may include a substantially isolated polypeptide antigen comprising an epitope which is specifically immunoreactive with at least one anti-polypeptide antigen antibody.
  • a kit includes means for detecting the binding of said antibody to the antigen (e.g., the antibody may be conjugated to a fluorescent compound such as fluorescein or rhodamine which can be detected by flow cytometry).
  • the kit may include a recombinantly produced or chemically synthesized polypeptide antigen.
  • the polypeptide antigen of the kit may also be attached to a solid support.
  • the detecting means of the above-described kit includes a solid support to which said polypeptide antigen is attached.
  • a kit may also include a non-attached reporter-labeled anti-human antibody.
  • binding of the antibody to the polypeptide antigen can be detected by binding of the said reporter-labeled antibody.
  • the invention includes a diagnostic kit for use in screening serum containing antigens of the polypeptide of the invention.
  • the diagnostic kit includes a substantially isolated antibody specifically immunoreactive with polypeptide or polynucleotide antigens, and means for detecting the binding of the polynucleotide or polypeptide antigen to the antibody.
  • the antibody is attached to a solid support.
  • the antibody is a monoclonal antibody.
  • the detecting means of the kit may include a second, labeled monoclonal antibody. Alternatively, or in addition, the detecting means may include a labeled, competing antigen.
  • test serum is reacted with a solid phase reagent having a surface-bound antigen obtained by the methods of the present invention.
  • the reagent After binding with specific antigen antibody to the reagent and removing unbound serum components by washing, the reagent is reacted with reporter-labeled anti-human antibody to bind reporter to the reagent in proportion to the amount of bound anti-antigen antibody on the solid support.
  • the reagent is again washed to remove unbound labeled antibody, and the amount of reporter associated with the reagent is determined.
  • the reporter is an enzyme which is detected by incubating the solid phase in the presence of a suitable fluorometric, luminescent or colorimetric substrate (Sigma, St. Louis, Mo.).
  • the solid surface reagent in the above assay is prepared by known techniques for attaching protein material to solid support material, such as polymeric beads, dip sticks, 96-well plates and/or filter material. These attachment methods generally include non-specific adsorption of the protein to the support or covalent attachment of the protein, typically through a free amine group, to a chemically reactive group on the solid support, such as an activated carboxyl, hydroxyl, or aldehyde group. Alternatively, streptavidin coated plates can be used in conjunction with biotinylated antigen(s). [0254] All publications, patents and patent applications cited herein, whether supra or infra, are hereby incoiporated by reference in their entirety.
  • an antigen includes a mixture of two or more antigens
  • a reference to “a subject polypeptide” includes a plurality of such polypeptides
  • reference to “the agent” includes reference to one or more agents and equivalents thereof known to those skilled in the art, and so forth.

Landscapes

  • Health & Medical Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Biophysics (AREA)
  • Medicinal Chemistry (AREA)
  • Zoology (AREA)
  • Biochemistry (AREA)
  • Toxicology (AREA)
  • General Health & Medical Sciences (AREA)
  • Genetics & Genomics (AREA)
  • Gastroenterology & Hepatology (AREA)
  • Molecular Biology (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Immunology (AREA)
  • Peptides Or Proteins (AREA)
  • Preparation Of Compounds By Using Micro-Organisms (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)

Abstract

Disclosed herein are two newly identified G-CSF splice variant molecules, their polypeptide sequences, the polynucleotides encoding the polypeptide sequences, and active fragments thereof. Also provided is a procedure for producing such polypeptides by recombinant techniques employing, for example, vectors and host cells. Also disclosed are methods for utilizing such polypeptides and modulators thereof for the treatment of diseases, including cancer, immune diseases, infectious diseases, and ischemic diseases.

Description

NOVEL G-CSF POLYPEPTIDES, POLYNUCLEOTIDES, MODULATORS THEREOF, AND METHODS OF USE PRIORITY CLAIM
[001] This application claims the benefit of provisional application 60/577,316, filed in the United States Patent and Trademark Office on June 4, 2004, the disclosure of which is hereby incorporated by reference. TECHNICAL FIELD [002] The present invention relates to newly identified granulocyte colony stimulating factor (G-CSF) polypeptides, polynucleotides, modulators, vectors, host cells, compositions, and kits. It relates to methods of making and methods of using these polypeptides, polynucleotides, and modulators, including antibody modulators, in diagnostic, prophylactic, and therapeutic applications. BACKGROUND ART [003] It is generally known that hematopoiesis is principally controlled both by cells in the microenvironment of the bone marrow and by secreted factors. These secreted factors, for example, colony stimulating factors, stimulate committed progenitor cells to proliferate and form colonies of differentiating blood cells. One of these colony stimulating factors is granulocyte colony stimulating factor (G-CSF), which preferentially stimulates the growth and development of granulocytes, for example, neutrophils (Souza et al., Science, 232:61-65 (1986); Gabrilove, Seminars in Hematol, 26:2 1-14 (1989)). [004] G-CSF is produced by a number of cell types including fibroblasts, macrophages, T-cells, trophoblasts, endothelial cells, and epithelial cells, and is the expression product of a single copy gene with four exons and five introns located on chromosome seventeen. Human G-CSF can be obtained and purified from a number of sources. For example, natural human G-CSF (nhG-CSF) can be isolated from the . supernatants of cultured human tumor cell lines. A G-CSF molecule in glycosylated form, as a product of eukaryotic host cell expression, and a G-CSF molecule in non- glycosylated form, as a product of prokaryotic host cell expression, have been reported (U.S. Pat. No. 4,810,643).
[005] In one currently used therapeutic approach, G-CSF is administered to cancer patients to stimulate neutrophil production in an effort to compensate for hematopoietic deficits resulting from chemotherapy or radiation therapy. However, the half-life of G- CSF in these patients is relatively short. Additionally, despite an improved toxicity profile for some current G-CSF therapies, undesirable side effects remain problematic upon administration of the G-CSF molecule. For example, the reported side effects include mild to moderate bone and/or musculoskeletal pain, anemia, thrombocytopenia, and injection site reactions. After repetitive G-CSF administrations, some of the reported side effects are more severe and include, for example, spleen enlargement and respiratory disturbances. Furthermore, there is in vitro and in vivo evidence that the current forms of G-CSF may lead to tumor progression.
[006] Thus, current G-CSF treatment models can be improved upon. It would be desirable to provide G-CSF molecules that effectively treat a variety of diseases and conditions while minimizing harmful side effects and other undesirable occurrences upon administration. BRBEF DESCRIPTION OF THE FIGURES AND TABLES Brief Description of the Figures Figure 1 [007] Figure 1 shows the polypeptide alignment of the novel G-CSF polypeptides of the invention, CLN00489695_5pvl and CLN00493523_5pvl (SEQ. ID. NOS.:9 and 10, respectively) with the amino acid sequences of known variants of G-CSF. The alignment was performed using Clustal Format for T-COFFEE Version_1.37, CPU=0.00 sec, SCORE=64, Nseq=12, Len=494. NP_000750_NM_000759 is the National Center for Biotechnology Information (NCBI) identification number for human colony stimulating factor 3 isoform a precursor. Sequence 23331380 is SEQ. ID. NO.: 148 from U.S. Patent No. 6,436,387. Sequence 23331383 is SEQ. ID. NO.:151 from U.S. Patent No. 6,436,387. Sequence 23331390 is SEQ. ID. NO.: 158 from U.S. Patent No. 6,436,387. Sequence 23331381 is SEQ. ID. NO.:149 from U.S. Patent No. 6,436,387. Sequence 23331388 is SEQ. ID. NO.:156 from U.S. Patent No. 6,436,387. Sequence 23331378 is SEQ. JD. NO.: 146 from U.S. Patent No. 6,436,387. Sequence 23331379 is SEQ. ID. NO.: 147 from U.S. Patent No. 6,436,387. Sequence 23331382 is SEQ. ID. NO.:150 from U.S. Patent No. 6,436,387. Sequence 56626517 is SEQ. ID. NO.:18 from U.S. Patent No. 6,797,493. Figure 2 [008] Figure 2 shows the polypeptide alignment of the novel G-CSF polypeptides of the invention, CLN00489695_5pvl and CLN00493523_5pvl (SEQ. ID. NOS.:9 and 10, respectively) with predicted translations of known variants of G-CSF. The alignment was performed using Clustal Format for T-COFFEE Version_1.37, CPU=5.74 sec, SCORE=59, Nseq=16, Len=873. NP_000750_NM_000759 is the National Center for Biotechnology Information (NCBI) identification number for human colony stimulating factor 3 isoform a precursor. Sequence 21261230 is a predicted translation of SEQ. ID. NO.:8 from WO 02/20751. Sequence 42285214 is a predicted translation of SEQ. ID. NO.:10291 from WO 02/068579. Sequence 21261234 is a predicted translation of SEQ. ID. NO.:12 from WO 02/20751. Sequence 21261235 is a predicted translation of SEQ. ID. NO.:13 from WO 02/20751. Sequence 21261226 is a predicted translation of SEQ. ID. NO.:4 from WO 02/20751. Sequence 53922219 is a predicted translation of SEQ. ID. NO.:95 from U.S Patent No. 6,730,303. Sequence 21261228 is a predicted translation of SEQ. ID. NO.:6 from WO 02/20751. Sequence 21261237 is a predicted translation of SEQ. ID. NO.:15 from WO 02/20751. Sequence 21261227 is a predicted translation of SEQ. ID. NO.:5 from WO 02/20751. Sequence 53922218 is apredicted translation of SEQ. ID. NO.:94 from U.S. Patent No. 6,730,303. Sequence 21261229 is a predicted translation of SEQ. ID. NO.:7 from WO 02/20751. Sequence 3977652 is a predicted translation of SEQ. ID. NO.:l from U.S. Patent No. 5,795,968. Sequence 6088626 is SEQ. ID. NO.:2 from WO 93/15211. Figure 3 [009] Figure 3 shows the NMR structure of the G-CSF molecule (FEBS Lett.
314:435-439 (1992). The deletions of the splice variants are highlighted and described in further detail below. Figure 3A shows the site of the deletion in CLN00489695. Figure 3B shows the site of the deletion in CLN00493523. Figure 4 [010] Figure 4 compares the amino acid sequences of human and mouse G-CSF.
The position of helices A, B, C, D, and E, as shown in Figure 3, are designated by the coils in Figure 4. The first box shows the deletion of CLN00489695. The second box shows the deletion of CLN00493523. Brief Description of the Tables Table 1
[Oil] Table 1 shows the correlation between the Sequence Listing and the G-CSF molecules discussed herein. Column 1 shows an internal designation identification number (FP ID). Column 2 shows the nucleotide sequence identification number for the open reading frame of the nucleic acid sequence (SEQ. ID. NO.:Nl). Column 3 shows the amino acid sequence identification number for the polypeptide sequence (SEQ. ID. NO..P1). Column 4 shows the nucleotide sequence identification number for the entire nucleic acid sequence, including both coding and non-coding sequences (SEQ. ID. NO.:N0). Column 5 shows the polypeptide identification number of the source clone or sequence (Source ID). Table 2 [012] Table 2 shows a publicly available annotation of the polypeptide sequences of the Sequence Listing. Column 1 shows an internal designation identification number of the polypeptide (FP TD). Column 2 shows the source identification number of the polypeptide (Source ID). Column 3 shows the predicted length of the polypeptide (Pred Prot Len). Columns 4 and 5 describe the characteristics of the human protein in the NCBI database with the greatest degree of similarity to polypeptides encoded by each FP JD. The NCBI protein is described by its accession number (Top Human Hit Accession ID) and by the NCBI's annotation of that sequence (Top Human Hit Annot). Column 6 shows the length of the polypeptide sequence set forth in column 4 (Top Human Hit Len). Column 7 shows the number of matching amino acid residues between the FP ID and the Top Human Hit sequences (Match Len). Column 8 shows the percent identity between the matching amino acid residues and the amino acid residues of the FP ID (% ID Over Query Len). For example, the length of FP ID HG1015527 is 164 amino acid residues. The number of amino acid matches with the Top Hit is 164 amino acid residues. The % ID over the Query Length is 164/164 x 100% = 100%. Column 9 shows the percent identity between the matching amino acid residues and the amino acid residues of the top human hit (% ID Over Hum Hit Len). For example, 164 of the amino acid residues are identical to the 200 amino acid residues of the top human hit. The % ID over Hum Hit Length is 164/200 x 100% = 82%. Table 3
[013] Table 3 shows characteristics of the polypeptides of the Sequence Listing.
Column 1 shows an internal designation identification number of the polypeptide (FP ID). Column 2 shows the source identification number of the polypeptide (Source ID). Column 3 shows the cluster identification number of polypeptides HG1015530, HG1015531, and HG1015532 (Cluster). Column 4 shows the classification of the polypeptide (Class), designating that cluster 16616 is secreted. Column 5 shows the predicted protein length (Pred Prot Len). Column 6 shows an internal parameter predicting the likelihood that the FP ID is secreted (Treevote), with "1" being a high likelihood that the polypeptide is secreted and "0" being a low likelihood that the polypeptide is secreted. Column 7 shows the amino acid residues that comprise a mature protein (Mature Protein Coords). Column 8 specifies signal peptide coordinates (Signal Peptide Coords). Column 9 shows the positions of the amino acids that do not pass through the membrane (Non-TM Coords). Column 10 specifies the interleukin-6 (IL6) protein family (Pfam) classification. Finally, column 11 provides the pfam coordinates (Pfam Coords). Table 4 [014] Table 4 shows a comparison between the disclosed polypeptide
CLN00489695_5pvl and known variants. Column 1 shows an internal designation identification number of the polypeptide (FP ID). Column 2 shows the source identification number of the polypeptide (Source ID). Column 3 shows the predicted protein length (Pred Prot Len). Column 4 shows the length of the match between the FP ID and the Source ID. Column 5 shows the percent identity between the FP ID and the Source ID over the length of the FP ID. Column 6 shows the percent identity between the FP ID and the Source ID over the length of Source ID. INDUSTRIAL APPLICABILITY [015] The polypeptide and modulator compositions and methods of the invention are useful in the diagnosis, treatment, and/or prevention of neutropenia, including chronic and chemotherapy-associated neutropenia, mobilization of bone marrow stem cells, treatment of inflammatory, immune, and ischemic diseases, treatment of bacterial and fungal infections, and the enhancement of myocardial regeneration without aggravated inflammation in the infarcted heart. DESCRIPTION OF THE INVENTION Summary of the Invention [016] The invention provides a first nucleic acid molecule comprising a first polynucleotide sequence chosen from SEQ. ID. NOS.: 1-2 and/or 19-20, a polynucleotide sequence encoding a polypeptide of SEQ. ID. NOS..9 and/or 10, and biologically active fragments of any of these, wherein the nucleic acid molecule is an isolated molecule. This first nucleic acid molecule may be a cDNA molecule, a genomic DNA molecule, a cRNA molecule, a siRNA molecule, a RNAi molecule, or an mRNA molecule. The invention provides a double-stranded isolated nucleic acid molecule comprising this first nucleic acid molecule and its complement. The invention provides a second nucleic acid molecule comprising a second polynucleotide sequence complementary to the first nucleic acid molecule. The second nucleic acid molecule may be, for example, a RNAi molecule, an anti-sense molecule, or a ribozyme. The invention also provides an isolated polypeptide comprising an amino acid sequence chosen from SEQ. ID. NOS.:9-10 and active fragments of either of these. This polypeptide may be present in a cell culture, for example a bacterial cell culture, a mammalian cell culture, or an insect cell culture. It may be encoded by the first nucleic acid molecule. This polypeptide may further comprise at least one fusion partner, for example, a polymer, a polypeptide, a succinyl group, or serum albumin. An example of a suitable polymer is either a branched or linear chain polyethylene glycol moiety, for example, one attached through an amino group of an amino acid of the polypeptide. [017] The invention further provides a vector comprising the first nucleic acid molecule and a promoter that regulates its expression. The promoter may or may not be naturally contiguous to the first nucleic acid molecule. It may be an inducible promoter, a conditionally-active promoter, for example the cre-lox promoter, a constitutive promoter, or a tissue-specific promoter.
[018] The invention yet further provides a recombinant host cell comprising the first nucleic acid molecule or a polypeptide encoded therein, an isolated polypeptide comprising an amino acid sequence chosen from SEQ. ID. NOS.: 9- 10 and active fragments of either of these, and/or a vector comprising the first nucleic acid molecule and a promoter that regulates its expression. The host cell may be a prokaryotic cell. It may also be a eukaryotic cell, for example, a human cell, a non-human mammalian cell, an insect cell, a fish cell, a plant cell, or a fungal cell.
[019] The invention provides a non-human animal injected with the first nucleic acid molecule. It also provides a non-human animal transformed with the first nucleic acid molecule or a polypeptide encoded thereby. It further provides a non-human animal injected with an isolated polypeptide comprising an amino acid sequence chosen from SEQ. ID. NOS.:9-10 and active fragments of either of these. [020] The invention also provides nucleic acid compositions, polypeptide compositions, vector compositions, and host cell compositions comprising the nucleic acid molecules, polypeptides, vectors, and host cells of the invention and a carrier, for example, a pharmaceutically acceptable carrier or excipient. [021] In another aspect, the invention provides a method of producing a recombinant host cell comprising providing a composition comprising a vector that comprises the first nucleic acid molecule and allowing the host cell to come into contact with the vector to form a recombinant host cell.
[022] The invention provides a method of producing a polypeptide comprising providing a composition comprising a recombinant host cell of the invention and culturing the recombinant host cell to produce the polypeptide. The invention also provides a method of producing a polypeptide comprising providing the first nucleic acid and expressing it in a cell free expression system to produce the polypeptide. Suitable cell free expression systems include a wheat germ lysate expression system, a rabbit reticulocyte expression system, and an E. coli lysate expression system. [023] The invention also provides a diagnostic kit comprising a composition comprising a polynucleotide molecule complementary to the first nucleic acid molecule and a vehicle. Kits of the invention may comprise an antibody that specifically binds to the first nucleic acid molecule or a polypeptide encoded thereby. Kits of the invention may comprise an antibody that specifically binds to an isolated polypeptide comprising an amino acid sequence chosen from SEQ. ID. NOS.: 9- 10 and active fragments of either of these. Kits of the invention may be diagnostic. They may include instructions for use. [024] In a further aspect, the invention provides a method of determining the presence of the first nucleic acid molecule or its complement comprising providing a complement to the first nucleic acid molecule or the complement of the first nucleic acid molecule, allowing the molecules to interact, and determining whether interaction has occurred.
[025] The invention provides a method of determining the presence of an antibody specific to a polypeptide encoded by the first nucleic acid molecule or an isolated polypeptide comprising an amino acid sequence chosen from SEQ. ID. NOS.:9-10 and active fragments of either of these in a sample comprising providing a composition comprising the polypeptide, allowing the polypeptide to interact with the sample, and determining whether interaction has occurred between the polypeptide and the antibody. It also provides an antibody that specifically binds to or interferes with the activity of one or more of these polypeptides. This antibody may be a polyclonal antibody, a monoclonal antibody, a single chain antibody, or an active fragments of any of these. For example, the antibody may be an antigen binding fragment, an Fc fragment, a cdr fragment, or a framework fragment.
[026] In yet a further aspect, the invention provides a method for treating or preventing disease in a subject comprising providing a composition comprising a polypeptide chosen from SEQ. DD. NOS.:9-10 and active fragments of either of these, and a carrier; and administering the composition to the subject. Polypeptides encoded by a nucleic acid molecule comprising a nucleotide sequence chosen from SEQ. ID. NOS.: 1-2, 19-20, and active fragments thereof are also suitable for practicing the invention. The polypeptides used in treating or preventing disease may comprise at least one fusion partner. Suitable fusion partners include, for example, polymers, polypeptides, succinyl groups, and serum albumin. The polymer may comprise either a branched or linear chain polyethylene glycol moiety, which may, for example, be attached through an amino group of an amino acid of the polypeptide.
[027] The method can be used to treat or prevent neutropenia, for example, neutropenia induced by chemotherapy or radiotherapy. The method can be used to treat or prevent infection, for example, a bacterial, fungal, parasitic, or viral infection; this method may further comprise administering granulocyte transfusion therapy. The method can be used to treat or prevent septic shock. It can be used to treat or prevent an inflammatory disease, for example, rheumatoid arthritis, myocarditis, inflammatory bowel disease, psoriasis, multiple sclerosis, systemic lupus erythematosus, intraocular inflammation, surgery, injury related inflammation, osteoarthritis, or central nervous system inflammation. It can be used to treat or prevent ocular inflammation, for example, scleritis, orbital pseudotumor, corneal melt, ocular cicatricial pemphigoid, or Sjogren's syndrome. The method can be used to treat or prevent ischemic diseases, for example, stroke, myocardial infarction, and fulminant liver failure. It can be used to treat or prevent immune diseases, for example, B-lymphocytic disease. It can be used to suppress immune activity.
[028] This method can be practiced with a polypeptide that further comprises at least one fusion partner, for example, a polymer, for example, a polyethylene glycol moiety; a polypeptide; a succinyl group; or serum albumin. The polyethylene glycol moiety may be attached through an amino group of an amino acid of the polypeptide. It may be either a branched or linear chain polymer. The method may include administering the composition to the subject locally or systemically. [029] The invention provides a method for mobilizing peripheral blood stem cells and/or bone marrow stem cells in a subject comprising providing a composition comprising a polypeptide chosen from SEQ. TD. NOS .9-10 and active fragments of either of these, and a carrier; and administering the composition to the subject. [030] The invention also provides a method for modulator screening comprising providing a polypeptide chosen from an isolated polypeptide comprising an amino acid sequence chosen from SEQ. ID. NOS.: 9- 10 and active fragments of either of these, or an isolated polypeptide encoded by the first nucleic acid molecule, and using the polypeptide as a target in an assay that detects the modulator. This assay can detect modulators which are, for example, small molecule drugs or antibodies. [031] In another aspect, the invention provides a polypeptide wherein at least one cysteine residue is replaced by a polar amino acid, a basic amino acid, an acidic amino acid, and/or a neutral amino acid, for example, serine or alanine. This polypeptide may further comprise at least one fusion partner. It may further comprise a growth factor, for example, GM-CSF or M-CSF. Definitions
[032] The terms used herein have their ordinary meanings, as set forth below, and can be further understood in the context of the specification. [033] The terms "polynucleotide," "nucleotide," "nucleic acid," "nucleic acid molecule," "nucleic acid sequence," "polynucleotide sequence," and "nucleotide sequence" are used interchangeably herein to refer to polymeric forms of nucleotides of any length. The polynucleotides can contain deoxyribonucleotides, ribonucleotides, and/or their analogs or derivatives.
[034] A "complement" of a nucleic acid molecule is a one that is comprised of its complementary base pairs. Deoxyribonucleotides with the base adenine are complementary to those with the base thymidine, and deoxyribonucleotides with the base thymidine are complementary to those with the base adenine. Deoxyribonucleotides with the base cytosine are complementary to those with the base guanine, and deoxyribonucleotides with the base guanine are complementary to those with the base cytosine. Ribonucleotides with the base adenine are complementary to those with the base uracil, and deoxyribonucleotides with the base uracil are complementary to those with the base adenine. Ribonucleotides with the base cytosine are complementary to those with the base guanine, and deoxyribonucleotides with the base guanine are complementary to those with the base cytosine.
[035] A "vector" is a plasmid that can be used to transfer DNA sequences from one organism to another or to express a gene of interest.
[036] A "promoter," as used herein, is a DNA regulatory region capable of binding
RNA polymerase in a mammalian cell and initiating transcription of a downstream (3' direction) coding sequence operably linked thereto. For purposes of the present invention, a promoter sequence includes the minimum number of bases or elements necessary to initiate transcription of a gene of interest at levels detectable above background. Within the promoter sequence is a transcription initiation site, as well as protein binding domains (consensus sequences) responsible for the binding of RNA polymerase. Eucaryotic promoters will often, but not always, contain "TATA" boxes and "CAT" boxes. Promoters include those that are naturally contiguous to a nucleic acid molecule and those that are not naturally contiguous to a nucleic acid molecule. Additionally, a promoter includes inducible promoters, conditionally active promoters, such as a cre-lox promoter, constitutive promoters, and tissue specific promoters. [037] "Expression of a nucleic acid molecule" refers to the conversion of the information contained in the molecule, into a gene product. A gene product can be the direct transcriptional product of a gene (e.g., mRNA, tRNA, rRNA, antisense RNA, ribozyme, structural RNA, or any other type of RNA) or a peptide or polypeptide produced by translation of an mRNA. Gene products also include RNAs which are modified, by processes such as capping, polyadenylation, methylation, and editing, and proteins modified by, for example, methylation, acetylation, phosphorylation, ubiquitination, ADP-ribosylation, myristilation, and glycosylation. [038] The terms "polypeptide," "peptide," and "protein," used interchangeably herein, refer to a polymeric form of amino acids of any length, which can include naturally-occurring amino acids, coded and non-coded amino acids, chemically or biochemically modified, derivatized, or designer amino acids, amino acid analogs, peptidomimetics, and depsipeptides, and polypeptides having modified, cyclic, bicyclic, depsicyclic, or depsibicyclic peptide backbones. The term includes single chain protein as well as multimers. The term also includes conjugated proteins, fusion proteins, including, but not limited to, glutathione S-transferase (GST) fusion proteins, fusion proteins with a heterologous amino acid sequence, fusion proteins with heterologous and homologous leader sequences, fusion proteins with or without N-terminal methionine residues, pegolyated proteins, and immunologically tagged, or his-tagged proteins. The term also includes peptide aptamers.
[039] A "pfam domain" is a protein or a portion of a protein with a tertiary structure. Pfams may have characteristic functional activities, such as enzymatic or binding activities. Multiple pfam domains can be connected by flexible polypeptide regions within a protein, pfam domains can comprise the N-terminus or the C-terminus of a protein, or can be situated at any point between.
[040] A "non-transmembrane domain" is a portion of a transmembrane protein that does not span the membrane. It may be extracellular, cytoplasmic, or luminal. [041] An "isolated," "purified," "substantially isolated," or "substantially purified" molecule (such as a polypeptide, polynucleotide, or antibody) is one that has been manipulated to exist in a higher concentration than in nature. For example, a subject antibody is isolated, purified, substantially isolated, or substantially purified when at least 10%, or 20%, or 40%, or 50%, or 70%, or 90% of non-subject-antibody materials with which it is associated in nature have been removed. As used herein, an "isolated," "purified," "substantially isolated," or "substantially purified" molecule includes recombinant molecules.
[042] By "fragment" is intended a polynucleotide or polypeptide consisting of only a part of the intact full-length or naturally occurring polynucleotide or polypeptide sequence and structure. A fragment may comprise or encode any portion of the molecules described herein. For example, a fragment may comprise or encode the amino acids at the splice sites of SEQ. ID. NOS.:l, 2, 7, 8, 9, 10, 17, 18, 19 and 20. A polypeptide fragment can include e.g., a C-terminal deletion, an N-terminal deletion, and/or an internal deletion of a native polypeptide or an extracellular domain of a transmembrane protein.
[043] A "biologically active" entity, or an entity having "biological activity," is one or more entity having structural, regulatory, or biochemical functions of a naturally occurring molecule or any function related to or associated with a metabolic or physiological process. Biological activity can be determined by protein folding or protein conformation. Biologically active polynucleotide fragments are those exhibiting activity similar, but not necessarily identical, to an activity of a polynucleotide of the present invention. The biological activity can include an improved desired activity, or a decreased undesirable activity. For example, an entity demonstrates biological activity when it participates in a molecular interaction with another molecule, such as hybridization, when it has therapeutic value in alleviating a disease condition, when it has prophylactic value in inducing an immune response, when it has diagnostic value in determining the presence of a molecule, such as a biologically active fragment of a polynucleotide that can, for example, be detected as unique for the polynucleotide molecule, or that can be used as a primer in a polymerase chain reaction. A biologically active polypeptide or fragment thereof includes one that can participate in a biological reaction, for example, one that can serve as an epitope or immunogen to stimulate an immune response, such as production of antibodies, or that can participate in stimulating or inhibiting signal transduction by binding to ligands receptors or other proteins, or nucleic acids; or activating enzymes or substrates.
[044] The terms "antibody" and "immunoglobulin" refer to a protein, for example, one generated by the immune system, synthetically, or recombinantly, that is capable of recognizing and binding to a specific antigen; antibodies are commonly known in the art. Antibodies may recognize polypeptide or polynucleotide antigens. The term includes active fragments, including for example, an antigen binding fragment of an immunoglobulin, a variable and/or constant region of a heavy chain, a variable and/or constant region of a light chain, a complementarity determining region (cdr), and a framework region. The terms include polyclonal and monoclonal antibody preparations, as well as preparations including hybrid antibodies, altered antibodies, chimeric antibodies, hybrid antibody molecules, F(ab')2 and F(ab) fragments; Fv molecules (for example, noncovalent heterodimers), dimeric and trimeric antibody fragment constructs; minibodies, humanized antibody molecules, and any functional fragments obtained from such molecules, wherein such fragments retain specific binding. [045] A "host cell" is an individual cell or cell culture which can be or has been a recipient of any recombinant vector(s) or isolated polynucleotide. Host cells include progeny of a single host cell, and the progeny may not necessarily be completely identical (in morphology or in total DNA complement) to the original parent cell due to natural, accidental, or deliberate mutation and/or change. A host cell includes cells transfected or infected in vivo or in vitro with a recombinant vector or a polynucleotide of the invention. A host cell which comprises a recombinant vector of the invention may be called a "recombinant host cell."
[046] A "stem cell" is a pluripotent or multipotent cell with the ability to self- renew, to remain undifferentiated, and to become differentiated. A stem cell can divide without limit, for at least the lifetime of the animal in which it naturally resides. A stem cell is not terminally differentiated; it is not at the end stage of a differentiation pathway. When a stem cell divides, each daughter cell can either remain a stem cell or embark on a course that leads toward terminal differentiation.
[047] "Subject," "individual," "host," and "patient" are used interchangeably herein to refer to mammals, including, but not limited to, rodents, simians, humans, felines, canines, equines, bovines, porcines, ovines, caprines, mammalian laboratory animals, mammalian farm animals, mammalian sport animals, and mammalian pets.
[048] The term "modulate" refers to the production, either directly or indirectly, of an increase or a decrease, a stimulation, inhibition, interference, or blockage in a measured activity when compared to a suitable control. A "modulator" of a polypeptide or polynucleotide or an "agent" are terms used interchangeably herein to refer to a substance that affects, for example, increases, decreases, stimulates, inhibits, interferes with, or blocks a measured activity of the polypeptide or polynucleotide, when compared to a suitable control.
[049] "Modulating a level of an active subject polypeptide" includes increasing or decreasing, blocking, or interfering with the expression or activity of a subject polypeptide, increasing or decreasing a level of an active polypeptide, and increasing or decreasing the level of mRNA encoding an active subject polypeptide. Modulation can occur directly or indirectly.
[050] "Disease" refers to any condition, infection, disorder, or syndrome that requires medical intervention or for which medical intervention is desirable. Such medical intervention can include treatment, diagnosis, and/or prevention.
[051] "Treatment," as used herein, covers any administration or application of remedies for disease in a mammal, including a human, and includes inhibiting the disease, i.e., arresting its development, or relieving the disease, i.e., causing regression, or restoring or repairing a lost, missing, or defective function; or stimulating an inefficient process.
[052] "Preventing," as used herein, includes providing prophylaxis with respect to the occurrence or recurrence of a disease in a subject that may be predisposed to the disease but has not yet been diagnosed with the disease. Treatment and prophylaxis can be administered to an organism, including a human, or to a cell in vivo, in vitro, or ex vivo, and the cell subsequently administered to the subject.
[053] A "composition" herein refers to a composition that usually contains a carrier, such as a pharmaceutically acceptable carrier or excipient that is conventional in the art and which is suitable for administration into a subject for therapeutic, diagnostic, or prophylactic purposes. It may include a cell culture in which the polypeptide or polynucleotide is present in the cells or in the culture medium. For example, compositions for oral administration can form solutions, suspensions, tablets, pills, capsules, sustained release formulations, oral rinses, or powders.
[054] A "pharmaceutically acceptable carrier" refers to a non-toxic solid, semisolid or liquid filler, diluent, encapsulating material, formulation auxiliary, or excipient of any conventional type. A pharmaceutically acceptable carrier is non-toxic to recipients at the dosages and concentrations employed and is compatible with other ingredients of the formulation. Nucleic Acids and Polypeptides [055] The invention provides two novel G-CSF molecules. It is known that the wild-type G-CSF polypeptide structure includes a four helix bundle (FEBS Lett., 314(3):435-439 (1992)), wherein the first two helices, known as helices A and B, are connected via a longer loop containing a short helix E, which allows helix A and B to align in a parallel manner (Figure 3A). In contrast to wild-type G-CSF, the first G-CSF polypeptide described herein, CLN00489695, lacks the connective helix E and a portion of helix A (Figure 4A). The second G-CSF polypeptide molecule, CLN004493523, lacks a portion of helix B (Figure 4B). Accordingly, the invention provides nucleic acid molecules containing a polynucleotide encoding these two newly identified G-CSF variant polypeptides, which have the amino acid sequences shown in the Sequence Listing (SEQ. ID. NOS.:9-10).
[056] Fragments of the full length G-CSF variants may be used as hybridization probes for cDNA libraries to isolate the full length gene and to isolate other genes which have a high sequence similarity or a similar biological activity. Probes of this type can have at least 30 bases and may comprise, for example, 50 or more bases. The probe may also be used in a screening procedure to identify a cDNA clone corresponding to a full length transcript and a genomic clone or clones that contain complete G-CSF genes, including regulatory and promoter regions, exons, and introns. An example of such a screen would include isolating the coding regions of G-CSF genes by using a known nucleic acid sequence to synthesize an oligonucleotide probe. Labeled oligonucleotides having a sequence complementary to a gene of the present invention can be used to screen a human cDNA, a genomic DNA, or a mRNA library to identify complementary library components.
[057] The present invention further relates to polynucleotides which hybridize to the described sequences if there is at least 91%, at least 92%, or at least 95% identity between the sequences. The present invention relates to polynucleotides which hybridize under stringent conditions to the described polynucleotides. Stringent conditions generally include condition under which hybridization will occur only if there is at least 95%, or at least 97% identity between the sequences. For example, overnight incubation at 42°C in a solution containing 50% formamide, 5x SSC (150 mM NaCl, 15 mM trisodium citrate), 50 mM sodium phosphate (pH 7.6), 5x Denhardt's solution, 10% dextran sulfate, and 20 μg/ml denatured, sheared salmon sperm DNA, followed by washing the filters in O.lx SSC at about 65°C, constitute stringent conditions. The polynucleotides which hybridize to the hereinabove described polynucleotides encode polypeptides which may retain substantially the same biological function or activity as the mature polypeptide.
[058] Alternatively, the polynucleotide may have at least 20 bases, at least 30 bases, or at least 50 bases which hybridize to a polynucleotide of the present invention and which has an identity thereto, as hereinabove described, and which may or may not retain activity.
[059] Thus, the present invention is directed to polynucleotides having at least a
93% identity, at least a 95% identity, or at least a 99% identity to a polynucleotide which encodes the polypeptides set forth in the Sequence Listing, as well as fragments thereof, which fragments have at least 30 bases or at least 50 bases, and to polypeptides encoded by such polynucleotides.
[060] Using the information provided herein, such as the nucleotide sequences set forth in the Sequence Listing, nucleic acid molecules of the present invention encoding a G-CSF polypeptide may be obtained using standard cloning and screening procedures, such as those for cloning cDNAs using mRNA as starting material. Nucleic acids of the invention are useful as hybridization probes for differential identification of the tissue(s) or cell type(s) present in a biological sample. Polypeptides and antibodies directed to those polypeptides are useful for providing immunological probes for the differential identification of tissues or cell types. Vectors and Host Cells [061] The present invention also relates to vectors which include the isolated nucleic acid molecules of the present invention, host cells which are genetically engineered with the recombinant vectors, and the production of G-CSF polypeptides or fragments thereof by recombinant techniques. The vector may be, for example, a phage, plasmid, viral, or retroviral vector. Retroviral vectors may be replication competent or replication defective. In the latter case, viral propagation generally will occur only in complementing host cells.
[062] The present invention provides recombinant vectors that contain, for example, nucleic acid constructs that encode secretory leader sequences and a selected heterologous polypeptide of interest, and host cells that are genetically engineered with the recombinant vectors. Selected heterologous polypeptides of interest in the present invention include, for example, an extracellular fragment of a secreted protein, a type I membrane protein, a type II membrane protein, a multi-membrane protein, and a soluble receptor. These vectors and host cells can be used for the production of polypeptides described herein, including fragments thereof by conventional recombinant techniques. The vector may be, for example, a phage, plasmid, viral or retroviral vector. Retroviral vectors may be replication competent or replication defective. As above, in the latter case, viral propagation generally will occur only in complementing host cells. [063] The polynucleotides may be joined to a vector containing a secretory leader sequence and a selectable marker for propagation in a host. Generally, a. plasmid vector is introduced in a precipitate, such as a calcium phosphate precipitate, or in a complex with a charged lipid. If the vector is a virus, it may be packaged in vitro using an appropriate packaging cell line and then transduced into host cells. [064] The polynucleotides may be joined to a vector containing a selectable marker for propagation in a host. Generally, a plasmid vector is introduced in a precipitate, such as a calcium phosphate precipitate, or in a complex with a charged lipid. If the vector is a virus, it may be packaged in vitro using an appropriate packaging cell line and then transduced into host cells. [065] The DNA insert can be operatively linked to an appropriate promoter, such as the phage lambda PL promoter; the E. coli lac, tφ, phoA and tac promoters; the SV40 early and late promoters; and promoters of retroviral LTRs, to name a few. Other suitable promoters will be known to the skilled artisan. The expression constructs will further contain sites for transcription initiation, termination, and, in the transcribed region, a ribosome binding site for translation. The coding portion of the transcripts expressed by the constructs can include a translation initiating codon at the beginning and a termination codon (UAA, UGA, or UAG) appropriately positioned at the end of the polypeptide to be translated.
[066] As indicated, the expression vectors may include at least one selectable marker. Such markers include dihydrofolate reductase, G418 or neomycin resistance for eukaryotic cell culture and tetracycline, kanamycin or ampicillin resistance genes for culturing in E. coli and other bacteria. Representative examples of appropriate hosts include, but are not limited to, bacterial cells, such as E. coli, Streptomyces and Salmonella typhimurium cells; fungal cells, such as yeast cells; insect cells such as Drosophila S2 and Spodoptera Sf9 cells; animal cells such as CHO, COS, 293, and Bowes melanoma cells; and plant cells. Appropriate culture mediums and conditions for the above-described host cells are known in the art.
[067] The selectable markers are genes that confer a phenotype on a cell expressing the marker, so that the cell can be identified under appropriate conditions. Generally, a selectable marker allows the selection of transformed cells based on their ability to thrive in the presence or absence of a chemical or other agent that inhibits an essential cell function. Suitable markers, therefore, include genes coding for proteins which confer drug resistance or sensitivity thereto, impart color to, or change the antigenic characteristics of those cells transfected with a molecule encoding the selectable marker, when the cells are grown in an appropriate selective medium. For example, selectable markers include cytotoxic markers and drug resistance markers, whereby cells are selected by their ability to grow on media containing one or more of the cytotoxins or drugs; auxotrophic markers by which cells are selected for their ability to grow on defined media with or without particular nutrients or supplements, such as thymidine and hypoxanthine; metabolic markers for which cells are selected, e.g., their ability to grow on defined media containing the appropriate sugar as the sole carbon source, and markers which confer the ability of cells to form colored colonies on chromogenic substrates or cause cells to fluoresce.
[068] Among vectors suitable for use in bacteria include pQE70, pQE60, and pQE-
9, available from Qiagen, Inc., (Mississauga, Ontario, Canada); pBS vectors, Phagescript vectors, Bluescript vectors, pNH8A, pNH6a, pNH18A, pNH46A, available from Stratagene (La Jolla, CA); and ptrc99a, pKK223-3, pKK233-3, pDR540, pRIT5 available from Pharmacia (Peapack, NJ). Among suitable eukaryotic vectors are pWLNEO, pSV2CAT, pOG44, pXTl, and pSG available from Stratagene; and pSVK3, pBPV, pMSG and pSVL, available from Pharmacia. Suitable viral vectors include the retroviral LTR; the S V40 promoter; and the human cytomegalovirus (CMV) promoter described in Miller, et al., Biotechniques, Vol. 7, No. 9, 980-990 (1989), or any other homologous or heterologous promoter, for example, cellular promoters such as eukaryotic cellular promoters including, but not limited to, the histone, pol HI, β-actin promoters, adenovirus promoters, e.g., the adenoviral major late promoter; thymidine kinase (TK) promoters; and B19 parvo virus promoters. Other suitable vectors will be readily apparent to the skilled artisan.
[069] Other suitable vectors include those employing a pTT vector backbone
(Durocher et al. Nucl. Acids Res. 30 (2002)). Briefly, the pTT vector backbone may be prepared by obtaining pIRESpuro/EGFP (pEGFP) and pSEAP basic vector(s), for example from Clontech (Palo Alto, CA), and pcDNA3.1, ρCDNA3.1/Myc-(His)6 and pCEP4 vectors can be obtained from, for example, Invitrogen. SuperGlo GFP variant (sgGFP) can be obtained from Q-Biogene (Carlsbad, CA). Preparing a pCEP5 vector can be accomplished by removing the CMV promoter and polyadenylation signal of pCEP4 by sequential digestion and self-ligation using Sail and Xbal enzymes resulting in plasmid pCEP4Δ. A GblH fragment from pAdCMV5 (Massie et al., J. Virol, 72: 2289- 2296 (1998)), encoding the CMV5-poly(A) expression cassette may be ligated in BglH- linearized ρCEP4Δ, resulting in pCEP5 vector. The pTT vector can be prepared by deleting the hygromycin (55 l and Sail excision followed by fill-in and ligation) and EBNA1 (Clal and Nsiϊ excision followed by fill-in and ligation) expression cassettes. The ColEI origin (Fspl-Sall fragment, including the 3' end of β-lactamase ORF) can be replaced with a Fspl-Sall fragment from pcDNA3.1 containing the pMBI origin (and the same 3' end of β-lactamase ORF). A Myc-(His)6 C-terminal fusion tag can be added to SEAP (HindΩΪ-Hpaϊ fragment from pSEAP-basic) following in-frame ligation in pcDNA3.1/Myc-His digested with HindJIΪ and EcoRV. Plasmids can subsequently be amplified in E. coli (DH5α) grown in LB medium and purified using MAXI prep columns (Qiagen, Mississauga, Ontario, Canada). To quantify, plasmids can be subsequently diluted in 50 mM Tris-HCl pH 7.4 and absorbencies can be measured at 260 nm and 280 nm. Plasmid preparations with A 6o/A28o ratios between about 1.75 and about 2.00 are suitable.
[070] Introduction of the construct into the host cell can be effected by calcium phosphate transfection, DΕAΕ-dextran mediated transfection, cationic lipid-mediated transfection, electroporation, transduction, infection, or other methods. Such methods are described in many standard laboratory manuals, such as Davis et al., Basic Methods In Molecular Biology (1986).
[071] The polypeptides may be expressed in a modified form, such as a fusion protein, and may include not only secretion signals, but also additional heterologous functional regions. For instance, a region of additional amino acids, particularly charged amino acids, may be added to the N-terminus of the polypeptide to improve stability and persistence in the host cell, during purification, or during subsequent handling and storage. Also, peptide moieties may be added to the polypeptide to facilitate purification. Such regions may be removed prior to final preparation of the polypeptide. [072] The addition of peptide moieties to polypeptides to engender secretion or excretion, to improve stability and to facilitate purification, among others, are familiar and routine techniques in the art. A suitable fusion protein may comprise a heterologous region from immunoglobulin that is useful to stabilize and purify proteins. For example, ΕP-A-O 464 533 (Canadian counterpart 2045869) discloses fusion proteins containing various portions of constant region of immunoglobulin molecules together with another human protein or part thereof. In many cases, the Fc part in a fusion protein is thoroughly advantageous for use in therapy and diagnosis and thus results, for example, in improved pharmacokinetic properties (ΕP-A 0232262). On the other hand, for some uses it would be desirable to be able to delete the Fc part after the fusion protein has been expressed, detected, and purified in the advantageous manner described. This is the case when the Fc portion proves to be a hindrance to use in therapy and diagnosis, for example when the fusion protein is to be used as an antigen for immunizations. In drug discovery, for example, human proteins, such as hIL-5, have been fused with Fc portions for the purpose of high-throughput screening assays to identify antagonists. See, Bennett et al., J. Molec. Recog., 8:52-58 (1995) and Johanson et al, J. Biol Chem., 270:9459-9471 (1995).
[073] The G-CSF polypeptides can be recovered and purified from recombinant cell cultures by well-known methods, including ammonium sulfate or ethanol precipitation, acid extraction, anion or cation exchange chromatography, phosphocellulose chromatography, hydrophobic interaction chromatography, affinity chromatography, hydroxylapatite chromatography, and lectin chromatography. High performance liquid chromatography (HPLC) can be employed for purification. Polypeptides of the present invention include products purified from natural sources, including bodily fluids, tissues and cells, whether directly isolated or cultured; products of chemical synthetic procedures; and products produced by recombinant techniques from a prokaryotic or eukaryotic host, including, for example, bacterial, yeast, higher plant, insect, and mammalian cells. Depending upon the host employed in a recombinant production procedure, the polypeptides of the present invention may be glycosylated or may be non-glycosylated. In addition, polypeptides of the invention may also include an initial modified methionine residue, in some cases as a result of host-mediated processes. Thus, it is well known in the art that the N-terminal methionine encoded by the translation initiation codon generally is removed with high efficiency from any protein after translation in eukaryotic cells. While the N-terminal methionine on most proteins also is efficiently removed in most prokaryotes, for some proteins this prokaryotic removal process is inefficient, depending on the nature of the amino acid to which the N- terminal methionine is covalently linked.
[074] Typically, a heterologous polypeptide, whether modified or unmodified, may be expressed as described above, or as a fusion protein, and may include not only secretion signals, but also a secretory leader sequence. A secretory leader sequence of the invention directs certain proteins to the endoplasmic reticulum (ER). The ER separates the membrane-bound proteins from other proteins. Once localized to the ER, proteins can be further directed to the Golgi apparatus for distribution to vesicles; including secretory vesicles; the plasma membrane, lysosomes, and other organelles. [075] Proteins targeted to the ER by a secretory leader sequence can be released into the extracellular space as a secreted protein. For example, vesicles containing secreted proteins can fuse with the cell membrane and release their contents into the extracellular space—a process called exocytosis. Exocytosis can occur constitutively or after receipt of a triggering signal. In the latter case, the proteins may be stored in secretory vesicles (or secretory granules) until exocytosis is triggered. Similarly, proteins residing on the cell membrane can also be secreted into the extracellular space by proteolytic cleavage of a "linker" holding the protein to the membrane. [076] Additionally, peptide moieties and/or purification tags may be added to the polypeptide to facilitate purification. Such regions may be removed prior to final preparation of the polypeptide. The addition of peptide moieties to polypeptides to engender secretion or excretion, to improve stability, and to facilitate purification, among other reasons, are familiar and routine techniques in the ait. Suitable purification tags include, for example, V5, HISX6, HISX8, avidin, and biotin.
[077] The invention provides a fusion protein comprising a heterologous region from an immunoglobulin that is useful to stabilize and purify proteins. For example, EP- A-O 464533 (Canadian counterpart 2045869) discloses fusion proteins containing various portions of constant region of immunoglobulin molecules together with another human protein or part thereof. In many cases, the Fc part of a fusion protein is advantageous for use in therapy and diagnosis and thus results, for example, in improved pharmacokinetic properties (EP-A 0232 262). On the other hand, for some uses it would be desirable to be able to delete the Fc part after the fusion protein has been expressed, detected, and purified in the advantageous manner described. This is the case when the Fc portion proves to be a hindrance to use in therapy and/or diagnosis, for example, when the fusion protein is to be used as an antigen for immunizations. In drug discovery, for example, human proteins, such as hIL-5, have been fused with Fc portions for the purpose of high-throughput screening assays to identify antagonists of hIL-5. See, Bennett et al., J. Molec. Recog., 8:52-58 (1995) and Johanson et al, J. Biol. Chem., 270:9459-9471 (1995).
[078] A heterologous polypeptide of the invention can be recovered and purified from recombinant cell cultures by well-known methods including ammonium sulfate or ethanol precipitation, acid extraction, anion or cation exchange chromatography, phosphocellulose chromatography, hydrophobic interaction chromatography, affinity chromatography, hydroxylapatite chromatography, lectin chromatography, and high performance liquid chromatography (HPLC). Polypeptides of the present invention include products purified from natural sources, including bodily fluids, tissues and cells, whether directly isolated or cultured; products of chemical synthetic procedures; and products produced by recombinant techniques from a prokaryotic or eukaryotic host, including, for example, bacterial, yeast, higher plant, insect, and mammalian cells, or from a cell free expression system. Depending upon the host employed in a recombinant production procedure, the polypeptides of the present invention may be glycosylated or may be non-glycosylated. In addition, polypeptides of the invention may also include an initial modified methionine residue, in some cases as a result of host-mediated processes. Thus, it is well known in the art that the N-terminal methionine encoded by the translation initiation codon generally is removed with high efficiency from any protein after translation in all eukaryotic cells. While the N-terminal methionine on most proteins also is efficiently removed in most prokaryotes, for some proteins this prokaryotic removal process is inefficient, depending on the nature of the amino acid to which the N-terminal methionine is covalently linked. Polypeptides and Fragments [079] The invention further provides isolated G-CSF polypeptides containing the amino acid sequences encoded by the nucleotide sequences set forth in the Sequence Listing, the amino acid sequences set forth in the Sequence Listing, and polypeptides comprising a fragment of any of these.
[080] The invention provides secreted proteins, which are capable of being directed to the endoplasmic reticulum (ER), secretory vesicles, or the extracellular space as a result of a secretory leader, signal peptide, or leader sequence, as well as proteins released into the extracellular space without necessarily containing a signal sequence. K a secreted protein is released into the extracellular space, it may undergo extracellular processing to a mature polypeptide. Release into the extracellular space can occur by many mechanisms, including exocytosis and proteolytic cleavage. [081] The sequences of the invention encompass a variety of different types of nucleic acids and polypeptides with different structures and functions. They can encode or comprise polypeptides belonging to different protein families (pfam). The "pfam" system is an organization of protein sequence classification and analysis, based on conserved protein domains; it can be publicly accessed in a number of ways, for example, at http://pfam.wustl.edu. Protein domains are portions of proteins that have a tertiary structure and sometimes have enzymatic or binding activities; multiple domains can be connected by flexible polypeptide regions within a protein. Pfam domains can comprise the N-terminus or the C-terminus of a protein, or can be situated at any point in between. The pfam system identifies protein families based on these domains and provides an annotated, searchable database that classifies proteins into families (Bateman et al., Nucl. Acids Res. 30:276-280 (2002)). Sequences of the invention can encode or be comprised of more than one pfam.
[082] Sequences of the invention may comprise an interleukin-6/G-CSF/MGF
(IL6) pfam domain. IL-6 is a chemokine that has been reported, inter alia, to play a role in the differentiation of B-cells into immunoglobulin-secreting cells, as well as in the induction of myeloma and plasmacytoma growth, nerve cell differentiation, and acute phase reactants in hepatocytes (Hirano et al., Nature 324:73-76 (1986); Clogston et al., Arch. Biochem. Biophys. 272:144-151 (1989)). The IL6 pfam includes glycoproteins with disulfide bonds mediated by cysteine residues and typically comprises a bundle of four alpha helices with a left-handed twist (Zink et al., Biochemistry 33:8453-8463 (1994); http://pfam.wustl.edu/cgi-bin/getdesc? name=IL6). Variant and Mutant Polypeptides [083] Protein engineering may be employed to improve or alter the characteristics of G-CSF polypeptides of the invention. Recombinant DNA technology known to those skilled in the art can be used to create novel mutant proteins or "muteins" including single or multiple amino acid substitutions, deletions, additions, or fusion proteins. Such modified polypeptides can show, e.g., enhanced activity or increased stability. In addition, they may be purified in higher yields and show better solubility than the corresponding natural polypeptide, at least under certain purification and storage conditions.
[084] The present invention further relates to variants of the nucleic acid molecules of the present invention, which encode portions, analogs, or derivatives of the G-CSF molecules. Variants may occur naturally, such as a natural allelic variant, i.e., one of several alternate forms of a gene occupying a given chromosomal locus Genes II, Lewin, B., ed., John Wiley & Sons, New York (1985)). Non-naturally occurring variants may be produced using art-known mutagenesis techniques.
[085] Such variants include those produced by nucleotide substitutions, deletions, or additions. The substitutions, deletions, or additions may involve one or more nucleotides. The variants may be altered in coding regions, non-coding regions, or both. Alterations in the coding regions may produce conservative or non-conservative amino acid substitutions, deletions or additions. These may take the form of silent substitutions, additions, or deletions which do not alter the properties or activities of the described G- CSF proteins, or portions thereof.
[086] In an embodiment, the invention provides nucleic acid molecules encoding mature proteins, i.e., those with cleaved signal peptide or leader sequences, e.g., as shown in the Sequence Listing. Further embodiments include an isolated nucleic acid molecule comprising a polynucleotide having a nucleotide sequence at least 93% identical, or at least 95%, 96%, 97%, 98%, or 99% identical to a polynucleotide from the Sequence Listing, a polypeptide encoded by a polynucleotide shown in the Sequence Listing, a polypeptide shown in the Sequence Listing, or a biologically active fragment of any of these.
[087] A polynucleotide having a nucleotide sequence at least, for example, 95% identical to a reference nucleotide sequence encoding an G-CSF polypeptide is one in which the nucleotide sequence is identical to the reference sequence except that it may include up to five point mutations per each 100 nucleotides of the reference nucleotide sequence. In other words, to obtain a polynucleotide having a nucleotide sequence at least 95% identical to a reference nucleotide sequence, up to 5% of the nucleotides in the reference sequence may be deleted or substituted with another nucleotide, or a number of nucleotides up to 5% of the total nucleotides in the reference sequence may be inserted into the reference sequence. These mutations of the reference sequence may occur at the 5' or 3' terminal positions of the reference nucleotide sequence or anywhere between those terminal positions, interspersed either individually among nucleotides in the reference sequence or in one or more contiguous groups within the reference sequence. [088] As a practical matter, whether any particular nucleic acid molecule is at least
93%, 95%, 96%, 97%, 98%, or 99% identical to, for instance, the nucleotide sequences set forth in the Sequence Listing can be determined conventionally using known computer programs such as the Bestfit program (Wisconsin Sequence Analysis Package, Version 8 for Unix, Genetics Computer Group, Madison, Wl). Bestfit uses the local homology algorithm of Smith and Waterman, Advances in Applied Mathematics 2:482- 489 (1981), to find the best segment of homology between two sequences. When using Bestfit or any other sequence alignment program to determine whether a particular sequence is, for instance, 95% identical to a reference sequence according to the present invention, the parameters are set, of course, such that the percentage of identity is calculated over the full length of the reference nucleotide sequence and that gaps in homology of up to 5% of the total number of nucleotides in the reference sequence are allowed.
[089] The present application is directed to nucleic acid molecules at least 93%,
95%, 96%, 97%, 98%, or 99% identical to the nucleic acid sequences set forth in the Sequence Listing irrespective of whether they encode a polypeptide having G-CSF activity. Even where a particular nucleic acid molecule does not encode a polypeptide having G-CSF activity, one of skill in the art would know how to use the nucleic acid molecule, for instance, as a hybridization probe or a polymerase chain reaction (PCR) primer. Uses of the nucleic acid molecules of the present invention that do not encode a polypeptide having G-CSF activity include, inter alia, (1) isolating the G-CSF gene or allelic variants thereof in a cDNA library; (2) in situ hybridization (e.g., "FISH") to metaphase chromosomal spreads to provide the precise chromosomal location of the G- CSF genes, as described in Verna et al., Human Chromosomes: A Manual of Basic Techniques, Pergamon Press, New York (1988); and Northern blot analysis for detecting G-CSF mRNA expression in specific tissues. [090] The present application is also directed to nucleic acid molecules having sequences at least 93%, 95%, 96%, 97%, 98%, or 99% identical to a nucleic acid sequence of the Sequence Listing which, encode a polypeptide having G-CSF polypeptide activity, i.e., a polypeptide exhibiting activity either identical to or similar, but not identical, to an activity of the G-CSF polypeptides of the invention, as measured in a particular biological assay. For example, the G-CSF polypeptides of the present invention may either stimulate or inhibit the proliferation of various mammalian cells, as demonstrated below.
[091] Of course, due to the degeneracy of the genetic code, one of ordinary skill in the art will immediately recognize that a large number of the nucleic acid molecules having a sequence at least 93%, 95%, 96%, 97%, 98%, or 99% identical to the nucleic acid sequence of the nucleic acid sequences set forth in the Sequence Listing will encode a polypeptide having G-CSF polypeptide activity. In fact, since multiple degenerate variants of these nucleotide sequences encode the same polypeptide, this will be clear to the skilled artisan even without performing the above described comparison assay. It will be further recognized in the art that a reasonable number of nucleic acid molecules that are not degenerate variants will also encode a polypeptide having G-CSF polypeptide activity, the skilled artisan is fully aware of amino acid substitutions that are either less likely or not likely to significantly affect protein function (e.g., replacing one aliphatic amino acid with a second aliphatic amino acid), as further described below. N-Terminal and C-Terminal Deletion Mutants [092] For instance, for many proteins, including the extracellular domain of a membrane associated protein or the mature form(s) of a secreted protein, it is known in the art that one or more amino acids may be deleted from the N-terminus or C-terminus without substantial loss of biological function. For instance, Ron et al., J. Biol. Chem., 268:2984-2988 (1993), reported modified KGF proteins that had heparin binding activity even if 3, 8, or 27 amino-terminal amino acid residues were missing. [093] However, even if deletion of one or more amino acids from the N-terminus of a protein results in modification or loss of one or more biological functions of the protein, other biological activities may still be retained. Thus, the ability of the shortened protein to induce and/or bind to antibodies which recognize the complete or mature from of the protein generally will be retained when less than the majority of the residues of the complete or mature protein are removed from the N-terminus. Whether a particular polypeptide lacking N-terminal residues of a complete protein retains such immunologic activities can readily be determined by routine methods described herein and otherwise known in the art. Accordingly, the present invention further provides polypeptides having one or more residues deleted from the amino terminus of the amino acid sequences of the G-CSF molecules as shown in the Sequence Listing. [094] Similarly, many examples of biologically functional C-terminal deletion muteins are known. For instance, interferon gamma increases in activity as much as ten fold when 8-10 amino acid residues are deleted from the carboxy terminus of the protein, see, for example, Dobeli et al., /. Biotechnology, 7:199-216 (1988). [095] However, even if deletion of one or more amino acids from the C-terminus of a protein results in modification of loss of one or more biological functions of the protein, other biological activities may still be retained. Thus, the ability of the shortened protein to induce and/or bind to antibodies which recognize the complete or mature form of the protein generally will be retained when less than the majority of the residues of the complete or mature protein are removed from the C-terminus. Whether a particular polypeptide lacking C-terminal residues of a complete protein retains such immunologic activities can readily be determined by routine methods described herein and otherwise known in the art. Other Mutants [096] In addition to terminal deletion forms of the protein discussed above, it also will be recognized by one of ordinary skill in the art that some amino acid sequences of the G-CSF polypeptides can be varied without significant effect of the structure or function of the protein. If such differences in sequence are contemplated, it should be remembered that there will be critical areas on the protein which determine activity. [097] Thus, the invention further includes variations of the G-CSF polypeptides which show substantial G-CSF polypeptide activity or which include regions of the G- CSF proteins such as the protein portions discussed below. Such mutants include deletions, insertions, inversions, repeats, and type substitutions, selected according to general rules known in the art, so as have little effect on activity. For example, guidance concerning how to make phenotypically silent amino acid substitutions is provided in Bowie et al., Science, 247: 1306-1310 (1990), wherein the authors indicate that there are two main approaches for studying the tolerance of an amino acid sequence to change. The first method relies on the process of evolution, in which mutations are either accepted or rejected by natural selection. The second approach uses genetic engineering to introduce amino acid changes at specific positions of a cloned gene and selections, or screens, to identify sequences that maintain functionality.
[098] As the authors state, these studies have revealed that proteins are surprisingly tolerant of amino acid substitutions. The authors further indicate which amino acid changes are likely to be permissive at a certain position of the protein. For example, most buried amino acid residues require nonpolar side chains, whereas few features of surface side chains are generally conserved. Other such phenotypically silent substitutions are described in Bowie, et al., supra, and the references cited therein. Typically seen as conservative substitutions are the replacements, one for another, among the aliphatic amino acids Ala, Val, Leu, and lie; interchange of the hydroxyl residues Ser and Thr, exchange of the acidic residues Asp and Glu, substitution between the amide residues Asn and Gin, exchange of the basic residues Lys and Arg, and replacements between the aromatic residues Phe and Tyr.
[099] Thus, a fragment, derivative, or analog of a polypeptide of the Sequence
Listing or polypeptide encoded by a nucleic acid sequence of the Sequence Listing 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; such a substituted amino acid residue may or may not be one encoded by the genetic code; (ii) one in which one or more of the amino acid residues includes a substituent group; (iii) 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 in which the additional amino acids are fused to the above form of the polypeptide, such as an IgG Fc fusion region peptide, a leader or secretory sequence, a sequence employed to purify the above form of the 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. [0100] Thus, the G-CSF polypeptides of the present invention may include one or more amino acid substitutions, deletions, or additions, either from natural mutations or human manipulation. As indicated, these changes may be of a minor nature, such as conservative amino acid substitutions, that do not significantly affect the folding or activity of the protein. Conservative amino acid substitutions include the aromatic substitutions Phe, Trp, and Tyr; the hydrophobic substitutions Leu, Iso, and Val; the polar substitutions Glu and Asp; the basic substitutions Arg, Lys, and His; the acidic substitutions Asp and Glu; and the small amino acid substations Ala, Ser, Thr, Met, and Gly.
[0101] Amino acids essential for the functions of G-CSF polypeptides can be identified by methods known in the art, such as site-directed mutagenesis or alanine- scanning mutagenesis, see, for example, Cunningham and Wells, Science, 244:1081-1085 (1989). The latter procedure introduces single alanine mutations. The resulting mutant molecules are then tested for biological activity such as receptor binding, or in vitro or in vitro proliferative activity.
[0102] Of special interest are substitutions of charged amino acids with other charged or neutral amino acids which may produce proteins with highly desirable improved characteristics, such as less aggregation. Aggregation may not only reduce activity but also be problematic when preparing pharmaceutical formulations, because, for example, aggregates can be immunogenic, Pinckard et al., Clin. Exp. Immunol, 2:331-340 (1967); Robbins et al., Diabetes, 36:838-845 (1987); Cleland et al., Crit. Rev. Therapeutic Drug Carrier Systems, 10:307-377 (1993).
[0103] Replacing amino acids can also change the selectivity of the binding of a ligand to cell surface receptors. For example, Ostade et al., Nature, 361:266-268 (1993) describes certain mutations resulting in selective binding of TNF-α to only one of the two known types of TNF receptors. Sites that are critical for ligand-receptor binding can also be determined by structural analysis such as crystallization, nuclear magnetic resonance, or photoaffinity labeling, for example, Smith et al., J. Mol. Biol, 224:899-904 (1992) and de Vos et al., Science, 255:306-312 (1992).
[0104] The polypeptides of the present invention can be provided in an isolated form, and can be substantially purified. A recombinantly produced version of the herein described G-CSF polypeptides can be substantially purified, e.g., by the one-step method described in Smith and Johnson, Gene, 67:31-40 (1988). Polypeptides of the invention also can be purified from natural or recombinant sources using anti- G-CSF antibodies of the invention using methods which are well known in the art of protein purification. The polypeptides herein may be purified or isolated in the presence of ions or agents that aid in the refolding of the molecules or aid in dimerizing or trimerizing the molecules as conventional in the art.
[0105] Further polypeptides of the present invention include polypeptides which have at least 93%, 95%, 96%, 97%, 98%, or 99% similarity to those described above. The polypeptides of the invention also contain those which are at least 93%, 94%, or 95%, 96%, 97%, 98%, or 99% identical to a polypeptide encoded by a nucleic acid sequence of the Sequence Listing.
[0106] The percent similarity of two polypeptides can be measured by a similarity score determined by comparing the amino acid sequences of the two polypeptides using the Bestfit program with the default settings for determining similarity. Bestfit uses the local homology algorithm of Smith and Waterman, Advances in Applied Mathematics 2:482-489 (1981) to find the best segment of similarity between two sequences. [0107] As described in detail below, the polypeptides of the present invention can be used to raise polyclonal and monoclonal antibodies, which are useful in assays for detecting G-CSF protein expression, also as described below, or as agonists and/or antagonists capable of enhancing or inhibiting G-CSF protein function. These polypeptides can also be used in a yeast two-hybrid system to capture G-CSF protein binding proteins, which are also candidate agonists and antagonists, according to the present invention. The yeast two hybrid system is described in Fields and Song, Nature, 340:245-246 (1989). Aptamers [0108] Another suitable agent for modulating an activity of a subject polypeptide is an aptamer. Aptamers of the invention include both nucleotide and peptide aptamers. Nucleotide aptamers of the invention include double stranded DNA and single stranded RNA molecules that bind to G-CSF proteins or fragments thereof. Peptide aptamers are peptides or small polypeptides that act as dominant inhibitors of protein function. Peptide aptamers specifically bind to target proteins, blocking their functional ability (Kolonin et al., Proc. Natl. Acad. Sci. 95:14,266-14,271 (1998)). [0109] Due to the highly selective nature of peptide aptamers, they can be used not only to target a specific protein, but also to target specific functions of a given protein (for example, a signaling function). Further, peptide aptamers can be expressed in a controlled fashion by use of promoters which regulate expression in a temporal, spatial, or inducible manner. Peptide aptamers act dominantly, therefore, they can be used to analyze proteins for which loss-of-function mutants are not available. Aptamers of the invention may bind nucleotide cofactors (Latham et al., Nucl. Acids Res. 22:2817-2822 (1994)).
[0110] Peptide aptamers that bind with high affinity and specificity to a target protein can be isolated by a variety of techniques known in the art. Peptide aptamers can be isolated from random peptide libraries by yeast two-hybrid screens (Xu et al., Proc. Natl. Acad. Sci. 94:12,473-12,478. (1997)). They can also be isolated from phage libraries (Hoogenboom et al., Immunotechnology 4:1-20 (1998)) or chemically generated peptides/libraries. Epitope-Bearing Portions [0111] In another aspect, the invention provides a polypeptide comprising an epitope-bearing portion of a polypeptide of the invention. The epitope of this polypeptide portion is an immunogenic or antigenic epitope of a polypeptide of the invention. Immunogenic epitopes are those parts of a protein that elicit an antibody response when the whole protein is provided as the immunogen. On the other hand, a region of a protein molecule to which an antibody can bind is an antigenic epitope. The number of immunogenic epitopes of a protein generally is less than the number of antigenic epitopes. See, for instance, Geysen et al., Proc. Natl. Acad. Sci., USA 81:3998-4002 (1983).
[0112] As to the selection of polypeptides bearing an antigenic epitope (i.e., that contain a region of a protein molecule to which an antibody can bind), it is well known in that art that relatively short synthetic peptides that mimic part of a protein sequence are routinely capable of eliciting an antiserum that reacts with the partially mimicked protein. See, for instance, Sutcliffe et al., Science, 219:660-666 (1983). Peptides capable of eliciting protein-reactive sera are frequently represented in the primary sequence of a protein, can be characterized by a set of simple chemical rules, and are confined neither to immunodominant regions of intact proteins (i.e., immunogenic epitopes) nor to the amino or carboxyl terminals. Antigenic epitope-bearing peptides and polypeptides of the invention are therefore useful for raising antibodies, including monoclonal antibodies, that bind specifically to a polypeptide of the invention. See, for instance, Wilson et al., Cell, 37:767-778 (1984). The epitope-bearing peptides and polypeptides of the invention may be produced by any conventional means. See, for example, Houghten, Proc. Natl. Acad. Sci., USA 82:5131-5135 (1985), and U.S. Pat. No. 4,631,211 (1986). [0113] Epitope-bearing peptides and polypeptides of the invention can be used to induce antibodies according to methods well known in the art. See, for instance, Bittle, et al, J. Gen. Virol, 66:2347-2354 (1985). Immunogenic epitope-bearing peptides of the invention, i.e., those parts of a protein that elicit an antibody response when the whole protein is the immunogen, are identified according to methods known in the art. See, for instance, U.S. Pat. No. 5,194,392 (1990), which describes a general method of detecting or determining the sequence of monomers (amino acids or other compounds) which is a topological equivalent of the epitope (i.e., a "mimotope") which is complementary to a particular paratope (antigen binding site) of an antibody of interest. More generally, U.S. Pat. No. 4,433,092 (1989) describes a method of detecting or determining a sequence of monomers which is a topographical equivalent of a ligand which is complementary to the ligand binding site of a particular receptor of interest. Similarly, U.S. Pat. No. 5,480,971 (1996) discloses linear Cl-C7-alkyl peralkylated oligopeptides, and sets and libraries of such peptides, as well as methods for using such oligopeptide sets and libraries for determining the sequence of a peralkylated oligopeptide that preferentially binds to an acceptor molecule of interest. Thus, non-peptide analogs of the epitope-bearing peptides of the invention also can be made routinely by these methods. Fusion Proteins [0114] As one of skill in the art will appreciate, G-CSF polypeptides of the present invention, and the epitope-bearing fragments thereof described above, can be combined with parts of the constant domain of immunoglobulins, resulting in chimeric polypeptides. These fusion proteins facilitate purification and show an increased half-life in vivo. This has been shown, for example, for chimeric proteins consisting of the first two domains of the human CD4-polypeptide and various domains of the constant regions of the heavy or light chains of mammalian immunoglobulins (Fc), for example, as described in EP A 394,827; Traunecker et al., Nature, 331:84-86 (1988). Fusion proteins that have a disulfide-linked dimeric structure due to the IgG part can also be more efficient in binding and neutralizing other molecules than the monomeric G-CSF protein or protein fragment alone, for example, as described by Fountoulakis et al., J. Biochem., 270:3958-3964 (1995). Suitable chemical moieties for derivatization of a heterologous polypeptide include, for example, polymers, such as water soluble polymers, all or part of human serum albumin, fetuin A; fetuin B; a leucine zipper domain; a tetranectin trimerization domain; mannose binding protein (also known as mannose binding lectin), for example, mannose binding protein 1 ; and an Fc region.
[0115] Additionally, heterologous polypeptides of the present invention and the epitope-bearing fragments thereof described herein can be combined with parts of the constant domain of immunoglobulins (IgG), resulting in chimeric polypeptides. These particular fusion molecules facilitate purification and show an increased half-life in vivo. This has been shown, e.g., for chimeric proteins consisting of the first two domains of the human CD4-polypeptide and various domains of the constant regions of the heavy or light chains of mammalian immunoglobulins, for example, EP A 394,827; Traunecker et al., Nature, 331:84-86 (1988). Fusion molecules that have a disulfide-linked dimeric structure due to the IgG part can also be more efficient in binding and neutralizing other molecules than, for example, a monomeric polypeptide or polypeptide fragment alone, see, for example, Fountoulakis et al., J. Biochem., 270:3958-3964 (1995). [0116] In another described embodiment, a human serum albumin fusion molecule may also be prepared as described herein and as further described in U.S. Patent No. 6,686,179. Peptides that bind albumin are also suitable fusion partners, as are lipids or other molecules that bind albumin.
[0117] Moreover, the polypeptides of the present invention can be fused to marker sequences, such as a peptide that facilitates purification of the fused polypeptide. The marker amino acid sequence may be a hexa-histidine peptide such as the tag provided in a pQE vector (Qiagen, Inc., among others, many of which are commercially available. As described in Gentz et al., Proc. Natl Acad. Sci. USA 86:821-824 (1989), for instance, hexa-histidine provides for convenient purification of the fusion protein. Another peptide tag useful for purification, the chemagglutinin HA tag, corresponds to an epitope derived from the influenza hemagglutinin protein. (Wilson et al., Cell 37:767 (1984)). [0118] Oligomeric proteins may be used as fusion partners to increase molecular weight, thus decreasing kidney filtration. For example, coiled-coil domains, such as the tetramectin coiled-coil domain, the coiled-coil domain of cartilage oligomeric matrix protein, and angiopoietin coiled-coil domains, and leucine zipper domains are suitable fusion partners. Also suitable are collagen and collagen-like domains, including those found in collagens, mannose-binding lectin, lung surfactant proteins A and D, adiponectin, ficolin, conglutinin, macrophage scavenger receptor, and emilin. Dimeric immunoglobulin domains, for example antibody CH3 domains, are also suitable. [0119] Covalent modifications can also increase molecular size. PEGylation, described in more detail above, and hyperglycosylation are examples of suitable covalent modifications.
[0120] Polymers, e.g., water soluble polymers, are useful in the present invention as the polypeptide to which each polymer is attached will not precipitate in an aqueous environment, such as a physiological environment. Polymers employed in the invention will be pharmaceutically acceptable for the preparation of a therapeutic product or composition. One skilled in the art will be able to select the desired polymer based on such considerations as whether the polymer/protein conjugate will be used therapeutically and, if so, the desired dosage, circulation time, and resistance to proteolysis. [0121] Suitable, clinically acceptable, water soluble polymers include, but are not limited to, polyethylene glycol (PEG), polyethylene glycol propionaldehyde, copolymers of ethylene glycol/propylene glycol, monomethoxy-polyethylene glycol, carboxymethylcellulose, dextran, polyvinyl alcohol (PVA), polyvinyl pyrrolidone, poly- 1,3-dioxolane, poly-l,3,6-trioxane, ethylene/maleic anhydride copolymer, poly (β-amino acids) (either homopolymers or random copolymers), poly(n- vinyl pyrrolidone) polyethylene glycol, polypropylene glycol homopolymers (PPG) and other polyakylene oxides, polypropylene oxide/ethylene oxide copolymers, polyoxyethylated polyols (POG) (e.g., glycerol) and other polyoxyethylated polyols, polyoxyethylated sorbitol, or polyoxyethylated glucose, colonic acids or other carbohydrate polymers, Ficoll, or dextran and mixtures thereof.
[0122] As used herein, polyethylene glycol (PEG) is meant to encompass any of the forms that have been used to derivatize other proteins, such as mono-(Cl-ClO) alkoxy- or aryloxy-polyethylene glycol. Polyethylene glycol propionaldehyde may have advantages in manufacturing due to its stability in water.
[0123] Specifically, a modified heterologous polypeptide of the invention may be prepared by attaching polyamino acids or branch point amino acids to the polypeptide. For example, the polyamino acid may be a carrier protein that serves to increase the circulation half life of the polypeptide (i.e., in addition to the advantages achieved via a fusion molecule). For the therapeutic purpose of the present invention, such polyamino acids should ideally be those that have or do not create neutralizing antigenic response, or other adverse responses. Such polyamino acids may be selected from serum album (such as human serum albumin), an additional antibody or portion thereof, for example the Fc region, fetuin A, fetuin B, leucine zipper nuclear factor erythroid derivative-2 (NFE2), neuroretinal leucine zipper, mannose motif (mbpl), tetranectin, or other polyamino acids, e.g. lysines. As described herein, the location of attachment of the polyamino acid may be at the N-terminus, or C-terminus, or other places in between, and also may be connected by a chemical "linker" moiety to the selected molecule. [0124] Polymers used herein, for example water soluble polymers, may be of any molecular weight and may be branched or unbranched. The polymers each typically have an average molecular weight of between about 1 kDa to about 100 kDa (in reference to a polymer, the term "about" indicating that in preparations of a polymer, some molecules will weigh more, some less, than the stated molecular weight). The average molecular weight of each polymer may be between about 5 kDa and about 50 kDa, or between about 12 kDa and about 25 kDa. Generally, the higher the molecular weight or the more branches, the higher the polymeπprotein ratio. Other sizes may also be used, depending on the desired therapeutic profile; for example, the duration of sustained release; the effects, if any, on biological activity; the ease in handling; the degree or lack of antigenicity; and other known effects of a polymer on a modified molecule of the invention. [0125] Polymers employed in the present invention are typically attached to a heterologous polypeptide with consideration of effects on functional or antigenic domains of the polypeptide. h general, chemical derivatization may be performed under any suitable condition used to react a protein with an activated polymer molecule. Activating groups which can be used to link the polymer to the active moieties include the following: sulfone, maleimide, sulfhydryl, thiol, triflate, tresylate, azidirine, oxirane, and 5-pyridyl.
[0126] Polymers of the invention are typically attached to a heterologous polypeptide at the alpha (α) or epsilon (ε) amino groups of amino acids or a reactive thiol group, but it is also contemplated that a polymer group could be attached to any reactive group of the protein that is sufficiently reactive to become attached to a polymer group under suitable reaction conditions. Thus, a polymer may be covalently bound to a heterologous polypeptide via a reactive group, such as a free amino or carboxyl group. The amino acid residues having a free amino group may include lysine residues and the N-terminal amino acid residue. Those having a free carboxyl group may include aspartic acid residues, glutamic acid residues, and the C-terminal amino acid residue. Those having a reactive thiol group include cysteine residues.
[0127] Methods for preparing fusion molecules conjugated with polymers, such as water soluble polymers, will each generally involve (a) reacting a heterologous polypeptide with a polymer under conditions whereby the polypeptide becomes attached to one or more polymers and (b) obtaining the reaction product. Reaction conditions for each conjugation may be selected from any of those known in the art or those subsequently developed, but should be selected to avoid or limit exposure to reaction conditions such as temperatures, solvents, and pH levels that would inactivate the protein to be modified. In general, the optimal reaction conditions for the reactions will be determined case-by-case based on known parameters and the desired result. For example, the larger the ratio of polymer:polypeptide conjugate, the greater the percentage of conjugated product. The optimum ratio (in terms of efficiency of reaction in that there is no excess unreacted polypeptide or polymer) may be determined by factors such as the desired degree of derivatization (e.g., mono-, di-tri- etc.), the molecular weight of the polymer selected, whether the polymer is branched or unbranched and the reaction conditions used. The ratio of polymer (e.g., PEG) to a polypeptide will generally range from 1 : 1 to 100: 1. One or more purified conjugates may be prepared from each mixture by standard purification techniques, including among others, dialysis, salting-out, ultrafiltration, ion-exchange chromatography, gel filtration chromatography, and electrophoresis.
[0128] One may specifically desire an N-terminal chemically modified protein. One may select a polymer by molecular weight, branching, etc., the proportion of polymers to protein (polypeptide or peptide) molecules in the reaction mix, the type of reaction to be performed, and the method of obtaining the selected N-terminal chemically modified protein. The method of obtaining the N-terminal chemically modified protein preparation (i.e., separating this moiety from other monoderivatized moieties if necessary) may be by purification of the N-terminal chemically modified protein material from a population of chemically modified protein molecules.
[0129] Selective N-terminal chemical modification may be accomplished by reductive alkylation which exploits differential reactivity of different types of primary amino groups (lysine versus the N-terminal) available for derivatization in a particular protein. Under the appropriate reaction conditions, substantially selective derivatization of the protein at the N-terminus with a carbonyl group containing polymer is achieved. For example, one may selectively attach a polymer to the N-terminus of the protein by performing the reaction at a pH which allows one to take advantage of the pKa differences between the ε-amino group of the lysine residues and that of the α-amino group of the N-terminal residue of the protein. By such selective derivatization, attachment of a polymer to a protein is controlled: the conjugation with the polymer takes place predominantly at the N-terminus of the protein and no significant modification of other reactive groups, such as the lysine side chain amino groups, occurs. Using reductive alkylation, the polymer may be of the type described above and should have a single reactive aldehyde for coupling to the protein. Polyethylene glycol propionaldehyde, containing a single reactive aldehyde, may also be used. [0130] In one embodiment, the present invention contemplates the chemically derivatized polypeptide to include mono- or poly- (e.g., 2-4) PEG moieties. Pegylation may be carried out by any of the pegylation reactions known in the art. Methods for preparing a pegylated protein product will generally include (a) reacting a polypeptide with polyethylene glycol (such as a reactive ester or aldehyde derivative of PEG) under conditions whereby the protein becomes attached to one or more PEG groups; and (b) obtaining the reaction product(s). In general, the optimal reaction conditions for the reactions will be determined case by case based on known parameters and the desired result.
[0131] There are a number of PEG attachment methods available to those skilled in the art. See, for example, EP 0401 384; Malik et al., Exp. HematoL, 20:1028-1035 (1992); Francis, Focus on Growth Factors, 3(2):4-10 (1992); EP 0 154 316; EP 0401 384; WO 92/16221; WO 95/34326; and the other publications cited herein that relate to pegylation, the disclosures of which are hereby incorporated by reference. Secretory Leader Sequences [0132] In order for some secreted proteins to express and secrete in larger quantities, a secretory leader sequence from another, different, secreted protein is desirable in some embodiments (U.S. 60/647,013). Employing heterologous secretory leader sequences is advantageous in that a resulting mature amino acid sequence, of the secreted polypeptide is not altered as the secretory leader sequence is removed in the ER during the secretion process. Moreover, the addition of a heterologous secretory leader is often required to express and secrete. For example, extracellular domains of Type II single transmembrane proteins (STM), as the secretory leader, which is also a transmembrane spanning domain, must typically be removed so that they may be soluble. Co-Translational and Post-Translational Modifications [0133] The invention encompasses polypeptides which are differentially modified during or after translation, e.g., by glycosylation, acetylation, phosphorylation, amidation, derivatization by known protecting/blocking groups, proteolytic cleavage, linkage to an antibody molecule or other cellular ligand. Any of numerous chemical modifications may be carried out by known techniques, including but not limited to specific chemical cleavage by cyanogen bromide, trypsin, chymotrypsin, papain, V8 protease; NABH4; acetylation; formylation; oxidation; reduction; and/or metabolic synthesis in the presence of tunicamycin. [0134] Additional post-translational modifications encompassed by the invention include, for example, e.g., N-linked or O-linked carbohydrate chains, processing of N- terminal or C-terminal ends), attachment of chemical moieties to the amino acid backbone, chemical modifications of N-linked or O-linked carbohydrate chains, and addition or deletion of an N-terminal methionine residue as a result of prokaryotic host cell expression. The polypeptides may also be modified with a detectable label, such as an enzymatic, fluorescent, isotopic, or affinity label to allow for detection and isolation of the protein. Compositions [0135] In some embodiments, G-CSF compositions are provided in formulation with pharmaceutically acceptable excipients, a wide variety of which are known in the art (Gennaro, Remington: The Science and Practice of Pharmacy with Facts and Comparisons: Drugfacts Plus, 20th ed. (2003); Ansel et al., Pharmaceutical Dosage Forms and Drug Delivery Systems, 7th ed., Lippencott Williams and Wilkins (2004); Kibbe et al., Handbook of Pharmaceutical Excipients, 3r ed., Pharmaceutical Press (2000)). Pharmaceutically acceptable excipients, such as vehicles, adjuvants, carriers or diluents, are readily available to the public. Moreover, pharmaceutically acceptable auxiliary substances, such as pH adjusting and buffering agents, tonicity adjusting agents, stabilizers, wetting agents and the like, are readily available to the public. [0136] Compositions for oral administration can form solutions, suspensions, tablets, pills, granules, capsules, sustained release formulations, oral rinses, or powders. For oral preparations, the agents, polynucleotides, and polypeptides can be used alone or in combination with appropriate additives, for example, with conventional additives, such as lactose, mannitol, corn starch, or potato starch; with binders, such as crystalline cellulose, cellulose derivatives, acacia, corn starch, or gelatins; with disintegrators, such as corn starch, potato starch, or sodium carboxymethylcellulose; with lubricants, such as talc or magnesium stearate; and if desired, with diluents, buffering agents, moistening agents, preservatives, and flavoring agents.
[0137] Suitable excipient vehicles are, for example, water, saline, dextrose, glycerol, ethanol, or the like, and combinations thereof. In addition, if desired, the vehicle can contain minor amounts of auxiliary substances such as wetting or emulsifying agents or pH buffering agents. Actual methods of preparing such dosage forms are known, or will be apparent, to those skilled in the art (Gennaro, 2003). The composition or formulation to be administered will, in any event, contain a quantity of the agent adequate to achieve the desired state in the subject being treated.
[0138] The agents, polynucleotides, and polypeptides can be formulated into preparations for injection by dissolving, suspending or emulsifying them in an aqueous or nonaqueous solvent, such as vegetable or other similar oils, synthetic aliphatic acid glycerides, esters of higher aliphatic acids or propylene glycol; and if desired, with conventional additives such as solubilizers, isotonic agents, suspending agents, emulsifying agents, stabilizers and preservatives. Other formulations for oral or parenteral delivery can also be used, as conventional in the art.
[0139] The antibodies, agents, polynucleotides, and polypeptides can be utilized in aerosol formulation to be administered via inhalation. The compounds of the present invention can be formulated into pressurized acceptable propellants such as dichlorodifluoromethane, propane, nitrogen, and the like. Further, the agent, polynucleotides, or polypeptide composition may be converted to powder form for administration intranasally or by inhalation, as conventional in the art. [0140] Furthermore, the agents can be made into suppositories by mixing with a variety of bases such as emulsifying bases or water-soluble bases. The compounds of the present invention can be administered rectally via a suppository. The suppository can include vehicles such as cocoa butter, carbowaxes and polyethylene glycols, which melt at body temperature, yet are solidified at room temperature.
[0141] A polynucleotide, polypeptide, or other modulator, can also be introduced into tissues or host cells by other routes, such as viral infection, microinjection, or vesicle fusion. For example, expression vectors can be used to introduce nucleic acid compositions into a cell as described above. Further, jet injection can be used for intramuscular administration (Furth et al., Anal. Biochem. 205:365-368 (1992)). The DNA can be coated onto gold microparticles, and delivered intradermally by a particle bombardment device, or "gene gun" as described in the literature (Tang et al., Nature 356:152-154 (1992)), where gold microprojectiles are coated with the DNA, then bombarded into skin cells. [0142] Unit dosage forms for oral or rectal administration such as syrups, elixirs, and suspensions can be provided wherein each dosage unit, for example, teaspoonful, tablespoonful, tablet, or suppository, contains a predetermined amount of the composition containing one or more agents. Similarly, unit dosage forms for injection or intravenous administration can comprise the agent(s) in a composition as a solution in sterile water, normal saline or another pharmaceutically acceptable carrier. Chromosome Assays [0143] In certain embodiments relating to chromosomal mapping, a cDNA herein disclosed is used to clone the genomic nucleic acid of the G-CSF. This can be accomplished using a variety of well known techniques and libraries, which generally are commercially available. The genomic DNA then is used for in situ chromosome mapping using techniques well known for this purpose. Therefore, the nucleic acid molecules 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 the 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 is an important first step in correlating those sequences with genes associated with disease. [0144] Briefly, sequences can be mapped to chromosomes by preparing PCR primers 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. [0145] PCR mapping of somatic cell hybrids is a rapid procedure for assigning a particular DNA to a particular chromosome. Using the present 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. [0146] Fluorescence in 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 a cDNA as short as approximately 50 - 60 bases. For a review of this technique, see Verma et al., Human Chromosomes: A Manual of Basic Techniques, Pergamon Press, New York (1988).
[0147] 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 (coinheritance of physically adjacent genes).
[0148] Next, differences can be determined in the cDNA or genomic sequences of affected and unaffected individuals. If a mutation is observed in some or all of the 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 physical 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 genes (assuming 1 megabase mapping resolution and one gene per 20 kb). Identification of Agonists and Antagonists [0149] This invention provides modulators, i.e., polypeptides, polynucleotides, or other agents that increase or decrease the activity of their target. They may act as an agonist or antagonist, and interfere with the binding or activity of polypeptides or polynucleotides. Such modulators or agents include, for example, polypeptide variants, whether agonist or antagonist; antibodies, whether agonist or antagonist; soluble receptors, usually antagonists; small molecule drugs, whether agonist or antagonist; RNAi, usually an antagonist; antisense molecules, usually an antagonist; and ribozymes, usually an antagonist. In some embodiments, an agent is a subject polypeptide, where the subject polypeptide itself is administered to an individual. In some embodiments, an agent is an antibody specific for a subject "target" polypeptide. In some embodiments, an agent is a chemical compound such as a small molecule that may be useful as an orally available drug. Such modulation includes the recruitment of other molecules that directly effect the modulation. For example, an antibody that modulates the activity of a subject polypeptide that is a receptor on a cell surface may bind to the receptor and fix complement, activating the complement cascade and resulting in lysis of the cell. An agent which modulates a biological activity of a subject polypeptide or polynucleotide increases or decreases the activity or binding at least about 10%, at least about 15%, at least about 20%, at least about 25%, at least about 50%, at least about 80%, or at least about 2-fold, at least about 5-fold, or at least about 10-fold or more when compared to a suitable control.
[0150] This invention also provides a method of screening compounds to identify those which modulate the action of the polypeptide of the present invention. An example of such an assay comprises combining a mammalian fibroblast cell and the polypeptide(s) of the present invention, the compound to be screened and 3[H] thymidine under cell culture conditions where the fibroblast cell would normally proliferate. A control assay may be performed in the absence of the compound to be screened and compared to the amount of fibroblast proliferation in the presence of the compound to determine if the compound stimulates proliferation by determining the uptake of 3[H] thymidine in each case. The amount of fibroblast cell proliferation is measured by liquid scintillation chromatography, which measures the incorporation of [H] thymidine. Both agonistic and antagonistic compounds may be identified by this procedure. [0151] In another method, a mammalian cell or membrane preparation expressing a receptor for a polypeptide of the present invention, as described above, is incubated with a labeled polypeptide of the present invention in the presence of the compound. The ability of the compound to enhance or block this interaction could then be measured. Alternatively, the response of a known second messenger system following interaction of a compound to be screened and the G-CSF receptor is measured and the ability of the compound to bind to the receptor and elicit a second messenger response is measured to determine if the compound is a potential agonist or antagonist. Such second messenger systems include, but are not limited to, cAMP, guanylate cyclase, ion channels, and phosphoinositide hydrolysis.
[0152] Examples of antagonistic compounds include antibodies, or in some cases, oligonucleotides, which bind to a receptor of a polypeptide of the present invention but elicit no second messenger response, or which bind to the G-CSF polypeptide itself. Alternatively, a potential antagonist may be a mutant form of the polypeptide which binds to the receptors but elicits no second messenger response, thus effectively blocking the action of the polypeptide.
[0153] Another compound antagonistic to G-CSF genes and gene products 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 methods are based on the binding of a polynucleotide to DNA or RNA. For example, a 5' coding portion of the polynucleotide sequence, which encodes mature polypeptides of the present invention, can be used to design an antisense RNA oligonucleotide of from about 10 to about 40 base pairs in length. A DNA oligonucleotide is designed to be complementary to a region of the gene involved in transcription, for example, a triple helix (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 the polypeptides of the present invention. The antisense RNA oligonucleotide hybridizes to the mRNA in vivo and blocks translation of the mRNA molecule into the polypeptide, as described by Okano, J. Neurochem., 56:560 (1991); Oligodeoxynucleotides as Antisense Inhibitors of Gene Expression, CRC Press, Boca Raton, Fla. (1988). The oligonucleotides described above can also be delivered to cells such that the antisense RNA or DNA is expressed in vivo to inhibit polypeptide production.
[0154] Potential antagonist compounds also include small molecules which bind to and occupy the binding site of the receptors, thereby making the receptor inaccessible to its polypeptide such that normal biological activity is prevented. Examples of small molecules include, but are not limited to, small peptides or peptide-like molecules. Antagonist compounds may be employed to inhibit the cell growth and proliferation effects of the polypeptides of the present invention on neoplastic cells and tissues, i.e. stimulation of angiogenesis of tumors, and, therefore, retard or prevent abnormal cellular growth and proliferation, for example, in tumor formation or growth. [0155] The present invention also provides methods for identifying agents, such as antibodies, which enhance or block the actions of G-CSF molecules on cells. For example these agents may enhance or block interaction of G-CSF -binding molecules, such as receptors. Agents of interest include both agonists and antagonists. The invention provides agonists which increase the natural biological functions of G-CSF or which function in a manner similar to G-CSF. The invention also provides antagonists, which decrease or eliminate the functions of G-CSF.
[0156] One method of identifying G-CSF agonists and antagonists involves biochemical assays following subcellular fractionation. For example, a cellular compartment, such as a membrane or cytosolic preparation may be prepared from a cell that expresses a molecule that binds G-CSF molecules, such as a molecule of a signaling or regulatory pathway modulated by G-CSF molecules. Subcellular fractionation methods are known in the art of cell biology, and can be tailored to produce crude fractions with discrete and defined components, e.g., organelles or organellar membranes. The preparation is incubated with labeled G-CSF molecules in the absence or the presence of a candidate molecule which may be an G-CSF agonist or antagonist. The ability of the candidate molecule to interact with the binding molecule or an G-CSF molecules is reflected in decreased binding of the labeled ligand. Molecules which bind gratuitously, that is, without inducing the effects of G-CSF molecules, are most likely antagonists. Molecules that bind well and elicit effects that are the same as or closely related to G-CSF molecules may potentially prove to be agonists. [0157] The effects of potential agonists and antagonists may by measured, for instance, by determining an activity of one or more components of a second messenger system following interaction of the candidate molecule with a cell or appropriate cell preparation, and comparing the effect with that of G-CSF molecules, or with that of molecules that elicit the same effects as G-CSF. Second messenger systems which may be useful in this regard include, but are not limited to, cAMP, cGMP, ion channel, and phosphoinositide hydrolysis second messenger systems. [0158] A competitive assay for G-CSF antagonists combines a mixture of G-CSF molecules and a potential antagonist with membrane-bound G-CSF receptor molecules. Under appropriate conditions, this assay can also be performed with recombinant G-CSF receptor molecules. G-CSF molecules can be labeled, such as by radioactivity, such that the number of G-CSF molecules bound to a receptor molecule can be determined accurately to assess the effectiveness of the potential antagonist. [0159] Potential antagonists include small organic molecules, polypeptides, and antibodies that bind to a polypeptide of the invention, and thereby inhibit or extinguish its activity. Potential antagonists also may be small organic molecules, polypeptides such as closely related proteins or antibodies that bind the same sites on a binding molecule, such as a receptor molecule, without inducing G-CSF -induced activities, thereby preventing the action of G-CSF molecules by excluding G-CSF molecules from binding. Antagonists of the invention include fragments of the G-CSF molecules having the nucleic acid and amino acid sequences shown in the Sequence Listing. [0160] G-CSF polynucleotides, polypeptides, agonists, and/or antagonists of the invention may be used to treat neutropenia, including febrile neutropenia, sepsis, and other bacterial-mediated infections. They may be administered, for example, to enhance the efficacy of neutrophils or to expand the neutrophil compartment to reduce inflammation mediated by defenders, such as lymphocytes. They can be used to treat inflammatory disease caused by or characterized by an inability to eradicate pathogenic bacteria via the innate immune system, such that inappropriate reactions to self-antigens ensue when the adaptive immune response, such as a lymphocytic response, occurs. Crohn's disease is an example of such an inflammatory disease. In Crohn's disease, the innate immune response fails to remove bacteria that breach the intestinal epithelium. An adaptive immune response, characterized by inflammation resulting from lymphocytes secreting cytokines results.
[0161] Other potential antagonists include antisense molecules. Antisense technology can be used to control gene expression through, e.g., antisense DNA or RNA, or through triple-helix formation. Antisense techniques are discussed, for example, in Okano, J. Neurochem., 56:560 (1991); Oligodeoxynucleotides as Antisense Inhibitors of Gene Expression, CRC Press, Boca Raton, Fla. (1988). Triple helix formation is discussed in, for instance, Lee et al., Nucleic Acids Research, 6:3073 (1979); Cooney et al., Science, 241:456 (1988); and Dervan et al., Science, 251:1360 (1991). The methods are based on the binding of a polynucleotide to a complementary DNA or RNA. For example, the 5' coding portion of a polynucleotide that encodes the mature polypeptide of the present invention may be used to design an antisense RNA oligonucleotide of from about 10 to about 40 base pairs in length. A DNA oligonucleotide is designed to be complementary to a region of the gene involved in transcription, thereby preventing transcription and the subsequent production of G-CSF molecules. The antisense RNA oligonucleotide hybridizes to the mRNA in vivo and blocks translation of the mRNA molecule into an G-CSF polypeptide. 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 G-CSF molecules. Therapeutic Uses of G-CSF and its Agonists and Antagonists [0162] G-CSF polynucleotides, polypeptides, agonists, and/or antagonists of the invention may be used in developing treatments for any disorder mediated (directly or indirectly) by defective G-CSF molecules, or insufficient GSF. G-CSF polypeptides, agonists, and/or antagonists may be administered to a patient (e.g., a mammal, such as human) afflicted with such a disorder. Alternatively, a gene therapy approach may be applied to treat such disorders. Disclosure herein of G-CSF nucleotide sequences permits the detection of defective G-CSF genes, and the replacement thereof with normal G-CSF- encoding genes. Defective genes may be detected in in vitro diagnostic assays, and by comparison of the G-CSF nucleotide sequences disclosed herein with that of an G-CSF gene derived from a patient suspected of harboring a defect in this gene. [0163] The G-CSF molecules of the present invention may be employed to treat lymphoproliferative disease which results in lymphadenopathy. They may also mediate apoptosis by stimulating clonal deletion of T-cells and may, therefore, be employed to treat autoimmune disease to stimulate peripheral tolerance and cytotoxic T-cell mediated apoptosis. The G-CSF molecules may further be employed as a research tool in elucidating the biology of autoimmune disorders, including systemic lupus erythematosus (SLE), Graves' disease, immunoproliferative disease lymphadenopathy (IPL), angioimmunoproliferative lymphadenopathy (AIL), immunoblastic lymphadenopathy (H3L), rheumatoid arthritis, diabetes, and multiple sclerosis. It also finds use in treating allergies and graft versus host disease.
[0164] The G-CSF polynucleotides, polypeptides, and/or agonists or antagonists of the invention may also be used to treat, prevent, diagnose, and/or prognose diseases which include, but are not limited to, autoimmune disorders, immunodeficiency disorders, and graft versus host disease. Specific types of autoimmune diseases that can be treated with the molecules of the invention include, but are not limited to, Th2 lymphocyte related disorders (e.g., atopic dermatitis, atopic asthma, rhinoconjunctivitis, allergic rhinitis, Omenn's syndrome, systemic sclerosis, and graft versus host disease); Th-1 lymphocyte-related disorders (e.g., rheumatoid arthritis, multiple sclerosis, psoriasis, Sjogren's syndrome, Hashimoto's thyroiditis, Grave's disease, primary biliary cirrhosis, Wegener's granulomatosis, and tuberculosis); activated B lymphocyte-related disorders (e.g., systemic lupus erythematosus, Goodpasture's syndrome, rheumatoid arthritis, and type I diabetes).
[0165] Molecules of the invention are useful for killing or inhibiting the multiplication of a cell that produces an infectious disease or for treating an infectious disease. The molecules of the invention can be used accordingly in a variety of settings for the treatment of an infectious disease in an animal. In the context of an infectious disease, the term "treating" includes any or all of preventing the growth, multiplication or replication of the pathogen that causes the infectious disease and ameliorating one or more symptoms of an infectious disease.
[0166] The G-CSF polypeptides of the present invention may be employed to inhibit neoplasia, such as tumor cell growth. They may be responsible for tumor destruction through apoptosis and cytotoxicity to certain cells.
[0167] Diseases associated with increased cell survival, or the inhibition of apoptosis, that may be treated, prevented, diagnosed and/or prognosed with the G-CSF polynucleotides, polypeptides and/or agonists or antagonists of the invention include, but are not limited to, cancers (such as follicular lymphomas, carcinomas with p53 mutations, and hormone-dependent tumors, including, but not limited to colon cancer, cardiac tumors, pancreatic cancer, melanoma, retinoblastoma, glioblastoma, lung cancer, intestinal cancer, testicular cancer, stomach cancer, neuroblastoma, myxoma, myoma, lymphoma, endothelioma, osteoblastoma, osteoclastoma, osteosarcoma, chondrosarcoma, adenoma, breast cancer, prostate cancer, Kaposi's sarcoma, and ovarian cancer); autoimmune disorders (such as, multiple sclerosis, Sjδgren's syndrome, Graves' disease, Hashimoto's thyroiditis, autoimmune diabetes, biliary cirrhosis, Behcet's disease, Crohn's disease, polymyositis, systemic lupus erythematosus, and immune-related glomerulonephritis, autoimmune gastritis, autoimmune thrombocytopenic purpura, and rheumatoid arthritis) and viral infections (such as herpes viruses, pox viruses and adenoviruses), inflammation, graft vs. host disease (acute and/or chronic), acute graft rejection, and chronic graft rejection. G-CSF polynucleotides and/or polypeptides, and their agonists, and/or antagonists may be used to inhibit growth, progression, and/or metastasis of cancers, in particular those listed above or in the paragraph that follows. [0168] Additional diseases or conditions associated with increased cell survival, that may be treated, prevented, diagnosed, and/or prognosed with the G-CSF polynucleotides and/or polypeptides and their agonists and/or antagonists include, but are not limited to, progression and/or metastases of malignancies and related disorders such as leukemia (including acute leukemias, (e.g., acute lymphocytic leukemia and acute myelocytic leukemia (including myeloblastic, promyelocytic, myelomonocytic, monocytic, and erythroleukemia) and chronic leukemias (e.g., chronic myelocytic (granulocytic) leukemia and chronic lymphocytic leukemia)), polycythemia vera, lymphomas (e.g., Hodgkin's disease and non-Hodgkin's disease), multiple myeloma, Waldenstrom's macroglobulinemia, heavy chain diseases, and solid tumors including, but not limited to, sarcomas and carcinomas such as fibrosarcoma, myxosarcoma, liposarcoma, chondrosarcoma, osteogenic sarcoma, chordoma, angiosarcoma, endotheliosarcoma, lymphangiosarcoma, lymphangioendotheliosarcoma, synovioma, mesothelioma, Ewing's tumor, leiomyosarcoma, rhabdomyosarcoma, colon carcinoma, pancreatic cancer, breast cancer, ovarian cancer, prostate cancer, squamous cell carcinoma, basal cell carcinoma, adenocarcinoma, sweat gland carcinoma, sebaceous gland carcinoma, papillary carcinoma, papillary adenocarcinomas, cystadenocarcinoma, medullary carcinoma, bronchogenic carcinoma, renal cell carcinoma, hepatoma, bile duct carcinoma, choriocarcinoma, seminoma, embryonal carcinoma, Wilm's tumor, cervical cancer, testicular tumor, lung carcinoma, small cell lung carcinoma, bladder carcinoma, epithelial carcinoma, glioma, astrocytoma, medulloblastoma, craniopharyngioma, ependymoma, pinealoma, hemangioblastoma, acoustic neuroma, oligodendroglioma, menangioma, melanoma, neuroblastoma, and retinoblastoma.
[0169] Diseases associated with increased apoptosis, that may be treated, prevented, diagnosed and/or prognosed with the G-CSF polynucleotides, polypeptides and/or agonists or antagonists of the invention include, but are not limited to, AIDS, neurodegenerative disorders (such as Alzheimer's disease, Parkinson's disease, amyotrophic lateral sclerosis, retinitis pigmentosa, cerebellar degeneration, and brain tumor or prior associated disease); diabetes, autoimmune disorders (such as, multiple sclerosis, Sjδgren's syndrome, Graves' disease, Hashimoto's thyroiditis, autoimmune diabetes, biliary cirrhosis, Behcet's disease, Crohn's disease, polymyositis, SLE, immune- related glomerulonephritis, autoimmune gastritis, thrombocytopenic purpura, and rheumatoid arthritis) myelodysplastic syndromes (such as aplastic anemia), graft vs. host disease (acute and/or chronic), ischemic injury (such as that caused by myocardial infarction, stroke, and reperfusion injury), liver injury or disease (e.g., hepatitis related liver injury, cirrhosis, ischemia/reperfusion injury, cholestosis (bile duct injury) and liver cancer); toxin-induced liver disease (such as that caused by alcohol), septic shock, ulcerative colitis, cachexia, and anorexia. In some embodiments, G-CSF polynucleotides, polypeptides, agonists, and/or antagonists are used to treat the diseases and disorders listed above.
[0170] Many of the pathologies associated with HIV are mediated by apoptosis, including HlV-induced nephropathy and HIV encephalitis. Thus, in some embodiments, G-CSF polynucleotides, polypeptides, agonists or antagonists of the invention are used to treat AIDS and pathologies associated with AIDS.
[0171] Another embodiment of the present invention is directed to the use of G-CSF polynucleotides, polypeptides, or antagonists to reduce G-CSF-mediated death of T cells in HlV-infected patients. The role of T cell apoptosis in the development of AIDS has been the subject of a number of studies (for example, Meyaard et al., Science, 257:217- 219 (1992); Groux et al., J. Exp. Med., 175:331 (1992); and Oyaizu et al, in Cell Activation and Apoptosis in HIV Infection, Andrieu and Lu, Eds., Plenum Press, New York, pp. 101-114 (1995)). Fas-mediated apoptosis has been implicated in the loss of T cells in HTV positive individuals (Katsikis et al., J. Exp. Med. 181:2029-2036 (1995). It is also likely that T cell apoptosis occurs through multiple mechanisms. [0172] Activated human T cells are induced to undergo programmed cell death (apoptosis) upon triggering through the CD3/T cell receptor complex, a process termed activated-induced cell death (AICD). AICD of CD4 T cells isolated from HiV-infected asymptomatic individuals has been reported (Groux et al., supra). Thus, AICD may play a role in the depletion of CD4+ T cells and the progression to AIDS in HiV-infected individuals. Thus, the present invention provides a method of inhibiting G-CSF- mediated T cell death in HIV patients, comprising administering G-CSF polynucleotides, polypeptides, or antagonists of the invention to the patients. In one embodiment, the patient is asymptomatic when treatment with G-CSF polynucleotides, polypeptides, or antagonists commences. If desired, prior to treatment, peripheral blood T cells may be extracted from an HIV patient, and tested for susceptibility to G-CSF-mediated cell death by procedures known in the art. In one embodiment, a patient's blood or plasma is contacted with G-CSF antagonists, anti-G-CSF antibodies, of the invention ex vivo. The G-CSF antagonists may be bound to a suitable chromatography matrix by procedures known in the art. The patient's blood or plasma flows through a chromatography column containing G-CSF antagonist bound to the matrix, before being returned to the patient. The immobilized G-CSF antagonist binds G-CSF, thus removing G-CSF protein from the patient's blood.
[0173] In additional embodiments, an G-CSF polynucleotide, polypeptide, or antagonist of the invention is administered in combination with other inhibitors of T cell apoptosis. For example, as discussed above, Fas-mediated apoptosis also has been implicated in loss of T cells in HIV individuals (Katsikis et al., J. Exp. Med., 181:2029- 2036 (1995)). Thus, a patient susceptible to both Fas ligand mediated and G-CSF- mediated T cell death may be treated with both an agent that blocks G-CSF/G-CSF receptor interactions and an agent that blocks Fas-ligand/Fas interactions. Suitable agents for blocking binding of Fas-ligand to Fas include, but are not limited to, soluble Fas polypeptides; multimeric forms of soluble Fas polypeptides (e.g., dimers of sFas/Fc); anti-Fas antibodies that bind Fas without transducing the biological signal that results in apoptosis; anti-Fas-ligand antibodies that block binding of Fas-ligand to Fas; and muteins of Fas-ligand that bind Fas but do not transduce the biological signal that results in apoptosis. Preferably, the antibodies employed according to this method are monoclonal antibodies. Examples of suitable agents for blocking Fas-ligand/Fas interactions, including blocking anti-Fas monoclonal antibodies, are described in WO 95/10540, hereby incorporated by reference.
[0174] In another example, agents which block binding of G-CSF to an G-CSF receptor are administered with the G-CSF polynucleotides, polypeptides, or antagonists of the invention. Such agents include, but are not limited to, soluble G-CSF receptor polypeptides; multimeric forms of soluble G-CSF receptor polypeptides; and G-CSF receptor antibodies that bind the G-CSF receptor without transducing the biological signal that results in apoptosis, anti- G-CSF antibodies that block binding of G-CSF to one or more G-CSF receptors, and muteins of G-CSF that bind G-CSF receptors but do not transduce the biological signal that results in apoptosis.
[0175] G-CSF polypeptides of the invention may also be employed to regulate hematopoeisis and, in particular, erythropoiesis. Hematopoeisis is a multi-step cell proliferation and differentiation process which begins with a pool of multipotent stem cells. These cells can proliferate and differentiate into hematopoietic progenitors in reply to different stimuli. The G-CSF polypeptides of the invention, as well as agonists and antagonists thereof, may be used to either stimulate or inhibit development of hematopoietic cells and, in particular, erythropoietic precursor cells. [0176] G-CSF may be used to treat B-cell deficiencies, including those arising in the context of infection, cancer treatment, transplant, immunodeficiency, immune disorder, agammaglobulinemia, hypogammaglobulinemia, defects in B cell development, defects in B cell function, defects in B-cell regulation, and shortened B cell lifespan. IL-7 is one of the factors involved in mediating B cell development (Stoddart et al., Immunol. Rev. 175:47-58 (2000)). Specifically, G-CSF may be used to treat Bruton agammaglobulinemia, an X chromosome-linked agammaglobulinemia conventionally understood as a life-threatening disease that involves a failure in normal development of B lymphocytes and is associated with missense mutations in BTK, a gene encoding a cytoplasmic tyrosine kinase (Bruton agammaglobulinemia tyrosine kinase, EC 2.7.1.112), a member of the Tec family of protein-tyrosine kinases (Ohta et al., Proc. Natl. Acad. Sci. 91:9062-9066 (1994).
[0177] G-CSF may be used to treat platelet deficiencies, such as manifest by thrombocytopenia, for example, immune-mediated thrombocytopenia. It can stimulate the growth and proliferation of lymphocytes, and can promote angiogenesis. G-CSF is useful in the treatment of diabetes. It can modulate the immune response of an organism, can treat allergies, and can be used to treat and/or prevent infection. G-CSF can inhibit tumor growth and can be used to treat cancer.
[0178] Additionally, molecules of the invention may be employed as agents to boost immunoresponsiveness among individuals having a temporary immune deficiency. Conditions resulting in a temporary immune deficiency that may be ameliorated or treated by administering the G-CSF polypeptides or polynucleotides of the invention, or agonists thereof, include, but are not limited to, recovery from viral infections (e.g., influenza), conditions associated with malnutrition, recovery from infectious mononucleosis, or conditions associated with stress, recovery from measles, recovery from blood transfusion, and recovery from surgery.
[0179] In the context of an autoimmune disease, the term "treating" includes any or all of preventing replication of cells associated with an autoimmune disease state including, but not limited to, cells capable of producing an autoimmune antibody, lessening the autoimmune-antibody burden, and ameliorating one or more symptoms of an autoimmune disease.
[0180] G-CSF polynucleotides or polypeptides of the invention, or agonists or antagonists thereof, may be used to diagnose, prognose, treat, or prevent one or more of the following diseases or disorders, or conditions associated therewith: primary immunodeficiencies, Kawasaki syndrome, bone marrow transplant (e.g., recent bone marrow transplant in adults or children), chronic B-cell lymphocytic leukemia, chronic inflammatory demyelinating polyneuropathy, and post-transfusion purpura, anemia (e.g., anemia associated with parvovirus B19, patients with stable multiple myeloma who are at high risk for infection (e.g., recurrent infection), autoimmune hemolytic anemia (e.g., warm-type autoimmune hemolytic anemia), thrombocytopenia (e.g., neonatal thrombocytopenia), and immune-mediated neutropenia), transplantation (e.g., cytomegalovirus (CMV)-negative recipients of CMV-positive organs), epilepsy (e.g., intractable epilepsy), and systemic vasculitic syndromes.
[0181] Autoimmune disorders and conditions associated with these disorders that may be treated, prevented, and/or diagnosed with the G-CSF polynucleotides, polypeptides, and/or antagonist of the invention (e.g., anti- G-CSF antibodies), include, but are not limited to, autoimmune hemolytic anemia, autoimmune neonatal thrombocytopenia, idiopathic thrombocytopenia purpura, autoimmunocytopenia, hemolytic anemia, antiphospholipid syndrome, dermatitis, allergic encephalomyelitis, myocarditis, relapsing polychondritis, rheumatic heart disease, glomerulonephritis (e.g., IgA nephropathy), multiple sclerosis, neuritis, uveitis ophthalmia, polyendocrinopathies, purpura (e.g., Henloch-Scoenlein purpura), Reiter's disease, stiff-man syndrome, autoimmune pulmonary inflammation, Guillain-Barre syndrome, insulin dependent diabetes mellitus, and autoimmune inflammatory eye disease. [0182] Additional autoimmune disorders highly likely to be treated, prevented, and/or diagnosed with the compositions of the invention include, but are not limited to, autoimmune thyroiditis, hypothyroidism (i.e., Hashimoto's thyroiditis) (often characterized, e.g., by cell-mediated and humoral thyroid cytotoxicity), systemic lupus erythematosus (often characterized, e.g., by circulating and locally generated immune complexes), Goodpasture's syndrome (often characterized, e.g., by anti-basement membrane antibodies), pemphigus (often characterized, e.g., by epidermal acantholytic antibodies), receptor autoimmunities such as, for example, (a) Graves' disease (often characterized, e.g., by TSH receptor antibodies), (b) myasthenia gravis (often characterized, e.g., by acetylcholine receptor antibodies), and (c) insulin resistance (often characterized, e.g., by insulin receptor antibodies), autoimmune hemolytic anemia (often characterized, e.g., by phagocytosis of antibody-sensitized red blood cells), autoimmune thrombocytopenic purpura (often characterized, e.g., by phagocytosis of antibody- sensitized platelets.
[0183] Additional autoimmune disorders which may be treated, prevented, and/or diagnosed with the compositions of the invention include, but are not limited to, rheumatoid arthritis (often characterized, e.g., by immune complexes in joints), scleroderma with anti-collagen antibodies (often characterized, e.g., by nucleolar and other nuclear antibodies), mixed connective tissue disease (often characterized, e.g., by antibodies to extractable nuclear antigens (e.g., ribonucleoprotein)), polymyositis/dermatomyositis (often characterized, e.g., by nonhistone anti-nuclear antibodies), pernicious anemia (often characterized, e.g., by antibodies to parietal cells, microsomes, and intrinsic factor), idiopathic Addison's disease (often characterized, e.g., by humoral and cell-mediated adrenal cytotoxicity, infertility (often characterized, e.g., by antispermatozoal antibodies), glomerulonephritis (often characterized, e.g., by glomerular basement membrane antibodies or immune complexes) such as primary glomerulonephritis and IgA nephropathy, bullous pemphigoid (often characterized, e.g., by IgG and complement in the basement membrane), Sjogren's syndrome (often characterized, e.g., by multiple tissue antibodies, and/or a specific nonhistone anti- nuclear antibodies (SS-B)), diabetes mellitus (often characterized, e.g., by cell-mediated and humoral islet cell antibodies), and adrenergic drug resistance (including adrenergic drug resistance with asthma or cystic fibrosis) (often characterized, e.g., by beta- adrenergic receptor antibodies).
[0184] Further autoimmune disorders which may be treated, prevented, and/or diagnosed with the compositions of the invention include, but are not limited to, chronic active hepatitis (often characterized, e.g. by smooth muscle antibodies), primary biliary cirrhosis (often characterized, e.g., by mitochondria! antibodies), other endocrine gland failure (often characterized, e.g., by specific tissue antibodies in some cases), vitiligo (often characterized, e.g., by melanocyte antibodies), vasculitis (often characterized, e.g., by Ig and complement in vessel walls and/or low serum complement), post-myocardial infarction (often characterized, e.g., by myocardial antibodies), cardiotomy syndrome (often characterized, e.g., by myocardial antibodies), urticaria (often characterized, e.g., by IgG and IgM antibodies to IgE), atopic dermatitis (often characterized, e.g., by IgG and IgM antibodies to IgE), asthma (often characterized, e.g., by IgG and IgM antibodies to IgE), inflammatory myopathies, and many other inflammatory, granulomatous, degenerative, and atrophic disorders.
[0185] In an additional embodiment, G-CSF polynucleotides or polypeptides, or antagonists thereof (e.g., anti-G-CSF antibodies) are used to treat or prevent systemic lupus erythematosus and/or diseases, disorders or conditions associated therewith. Lupus-associated diseases, disorders, or conditions that may be treated or prevented with G-CSF polynucleotides or polypeptides, or antagonists of the invention, include, but are not limited to, hematologic disorders (e.g., hemolytic anemia, leukopenia, lymphopenia, and thrombocytopenia), immunologic disorders (e.g., anti-DNA antibodies, and anti-Sm antibodies), rashes, photosensitivity, oral ulcers, arthritis, fever, fatigue, weight loss, serositis (e.g., pleuritus (pleuricy)), renal disorders (e.g., nephritis), neurological disorders (e.g., seizures, peripheral neuropathy, CNS related disorders), gastrointestinal disorders, Raynaud phenomenon, and pericarditis.
[0186] G-CSF polypeptides, agonists, or antagonists of the invention may be used to treat diseases associated with ischemia, e.g., cardiovascular disorders, including peripheral artery disease, such as limb ischemia. They may also include stroke, vascular disease, and fulminant liver failure. In the context of an ischemic disease, the term "treating" includes any or all of preventing the growth, multiplication, or replication of the pathogen that causes the ischemic disease and ameliorating one or more symptoms of an ischemic disease.
[0187] Stem cell mobilization to the heart and differentiation into cardiac myocytes is a naturally occurring process. Askari et al., Lancet 362:697-703 (2005), have speculated that up-regulation of this process can help recover myocardial function following infarction. In an embodiment, G-CSF induces therapeutic stem cell homing to injured myocardium. G-CSF has been observed to be up-regulated following myocardial infarction (Askari et al., supra). The invention provides compositions and methods for providing G-CSF to the heart. For example, autologous cells genetically modified with molecules of the Sequence Listing can be transplanted to a subject that can benefit from such cells. Bone marrow stimulation may be performed in conjunction with the transplantation. Cardiac fibroblasts and cardiac myoblasts can be transfected with G-CSF as described by Askari et al., supra, or other means known in the art, and provided in vivo by local administration. A rodent model with a ligation of the left anterior descending artery is suitable for studying and practicing this method. The method is suitable for therapeutic purposes in humans. This method of promoting tissue regeneration may be accomplished by stem cell engraftment. It is applicable to the heart, and other organs, including the brain, pancreas, lung, liver, skin, and bone. G-CSF can be delivered to the tissues by conventional local administration of a molecule described herein or a composition comprising such molecule. G-CSF can also be delivered to the tissues by local administration of a cell comprising a molecule described herein or a composition comprising such molecule. Suitable cells include heart cells, brain cells, pancreatic cells, lung cells, liver cells, skin cells, bone cells, mesenchymal stem cells, progenitor cells, adult stem cells, and embryonic stem cells.
[0188] Cardiovascular disorders include cardiovascular abnormalities, such as arterio-arterial fistula, arteriovenous fistula, cerebral arteriovenous malformations, congenital heart defects, pulmonary atresia and scimitar syndrome. Congenital heart defects include aortic coarctation, cor triatriatum, coronary vessel anomalies, crisscross heart, dextrocardia, patent ductus arteriosus, Ebstein's anomaly, Eisenmenger complex, hypoplastic left heart syndrome, levocardia, tetralogy of Fallot, transposition of great vessels, double outlet right ventricle, tricuspid atresia, persistent truncus arteriosus, and heart septal defects, such as aortopulmonary septal defect, endocardial cushion defects, Lutembacher's syndrome, trilogy of Fallot, and ventricular heart septal defects. [0189] Cardiovascular disorders also include heart disease, such as arrhythmias, carcinoid heart disease, high cardiac output, low cardiac output, cardiac tamponade, endocarditis (including bacterial), heart aneurysm, cardiac arrest, congestive heart failure, congestive cardiomyopathy, paroxysmal dyspnea, cardiac edema, heart hypertrophy, congestive cardiomyopathy, left ventricular hypertrophy, right ventricular hypertrophy, post-infarction heart rupture, ventricular septal rupture, heart valve diseases, myocardial diseases, myocardial ischemia, pericardial effusion, pericarditis (including constrictive and tuberculous), pneumopericardium, postpericardiotomy syndrome, pulmonary heart disease, rheumatic heart disease, ventricular dysfunction, hyperemia, cardiovascular pregnancy complications, scimitar syndrome, cardiovascular syphilis, and cardiovascular tuberculosis.
[0190] Arrhythmias include sinus arrhythmia, atrial fibrillation, atrial flutter, bradycardia, extrasystole, Adams-Stokes syndrome, bundle-branch block, sinoatrial block, long QT syndrome, parasystole, Lown-Ganong-Levine syndrome, Mahaim-type pre-excitation syndrome, Wolff-Parkinson- White syndrome, sick sinus syndrome, tachycardias, and ventricular fibrillation. Tachycardias include paroxysmal tachycardia, supraventricular tachycardia, accelerated idioventricular rhythm, atrioventricular nodal reentry tachycardia, ectopic atrial tachycardia, ectopic junctional tachycardia, sinoatrial nodal reentry tachycardia, sinus tachycardia, Torsades de Pointes, and ventricular tachycardia.
[0191] Heart valve diseases include aortic valve insufficiency, aortic valve stenosis, heart murmurs, aortic valve prolapse, mitral valve prolapse, tricuspid valve prolapse, mitral valve insufficiency, mitral valve stenosis, pulmonary atresia, pulmonary valve insufficiency, pulmonary valve stenosis, tricuspid atresia, tricuspid valve insufficiency, and tricuspid valve stenosis.
[0192] Myocardial diseases include alcoholic cardiomyopathy, congestive cardiomyopathy, hypertrophic cardiomyopathy, aortic subvalvular stenosis, pulmonary subvalvular stenosis, restrictive cardiomyopathy, Chagas cardiomyopathy, endocardial fibroelastosis, endomyocardial fibrosis, Kearns syndrome, myocardial reperfusion injury, and myocarditis.
[0193] Myocardial ischemias include coronary disease, such as angina pectoris, coronary aneurysm, coronary arteriosclerosis, coronary thrombosis, coronary vasospasm, myocardial infarction and myocardial stunning.
[0194] Cardiovascular diseases also include vascular diseases such as aneurysms, angiodysplasia, angiomatosis, bacillary angiomatosis, Hippel-Lindau disease, Klippel-
Trenaunay- Weber syndrome, Sturge-Weber syndrome, angioneurotic edema, aortic diseases, Takayasu's arteritis, aortitis, Leriche's syndrome, arterial occlusive diseases, arteritis, enarteritis, polyarteritis nodosa, cerebrovascular disorders, diabetic angiopathies, diabetic retinopathy, embolisms, thrombosis, erythromelalgia, hemorrhoids, hepatic veno-occlusive disease, hypertension, hypotension, ischemia, peripheral vascular diseases, phlebitis, pulmonary veno-occlusive disease, Raynaud's disease, CREST syndrome, retinal vein occlusion, Scimitar syndrome, superior vena cava syndrome, telangiectasia, ataxia telangiectasia, hereditary hemorrhagic telangiectasia, varicocele, varicose veins, varicose ulcer, vasculitis, and venous insufficiency.
[0195] Aneurysms include dissecting aneurysms, false aneurysms, infected aneurysms, ruptured aneurysms, aortic aneurysms, cerebral aneurysms, coronary aneurysms, heart aneurysms, and iliac aneurysms. [0196] Arterial occlusive diseases include arteriosclerosis, intermittent claudication, carotid stenosis, fibromuscular dysplasias, mesenteric vascular occlusion, Moyamoya disease, renal artery obstruction, retinal artery occlusion, and thromboangitis obliterans. [0197] Cerebrovascular disorders include carotid artery diseases, cerebral amyloid angiopathy, cerebral aneurysm, cerebral anoxia, cerebral arteriosclerosis, cerebral arteriovenous malformation, cerebral artery diseases, cerebral embolism and thrombosis, carotid artery thrombosis, sinus thrombosis, Wallenberg's syndrome, cerebral hemorrhage, epidural hematoma, subdural hematoma, subarachnoid hemorrhage, cerebral infarction, cerebral ischemia (including transient), subclavian steal syndrome, periventricular leukomalacia, vascular headache, cluster headache, migraine, and vertebrobasilar insufficiency.
[0198] Embolisms include air embolisms, amniotic fluid embolisms, cholesterol embolisms, blue toe syndrome, fat embolisms, pulmonary embolisms, and thromboembolisms. Thromboses include coronary thrombosis, hepatic vein thrombosis, retinal vein occlusion, carotid artery thrombosis, sinus thrombosis, Wallenberg's syndrome, and thrombophlebitis.
[0199] Ischemia includes cerebral ischemia, ischemic colitis, compartment syndromes, anterior compartment syndrome, myocardial ischemia, reperfusion injuries, and peripheral limb ischemia. Vasculitis includes aortitis, arteritis, Behcet's syndrome, Churg-Strauss syndrome, mucocutaneous lymph node syndrome, thromboangiitis obliterans, hypersensitivity vasculitis, Schoenlein-Henoch purpura, allergic cutaneous vasculitis, and Wegener's granulomatosis.
[0200] Additionally, ocular disorders associated with neovascularization which can be treated with the G-CSF polynucleotides and polypeptides of the present invention (including G-CSF agonists and G-CSF antagonists) include, but are not limited to neovascular glaucoma, diabetic retinopathy, retinoblastoma, retrolental fibroplasia, uveitis, retinopathy of prematurity, macular degeneration, corneal graft neovascularization, as well as other eye inflammatory diseases, ocular tumors, and diseases associated with choroidal or iris neovascularization. See, e.g., reviews by Waltman et al., Am. J. Ophthal., 85:704-710 (1978) and Gartner et al., Surv. Ophthal., 22:291-312 (1978). [0201] Additionally, disorders which can be treated with the G-CSF polynucleotides and polypeptides of the present invention (including G-CSF agonists and G-CSF antagonists) include, but are not limited to, hemangioma, arthritis, psoriasis, angiofibroma, atherosclerotic plaques, delayed wound healing, granulations, hemophilic joints, hypertrophic scars, nonunion fractures, Osier- Weber syndrome, pyogenic granuloma, scleroderma, trachoma, and vascular adhesions.
[0202] Polynucleotides and/or polypeptides of the invention, and/or agonists and/or antagonists thereof, are useful in the diagnosis and treatment or prevention of a wide range of diseases and/or conditions. Such diseases and conditions include, but are not limited to, cancer (e.g., immune cell related cancers, breast cancer, prostate cancer, ovarian cancer, follicular lymphoma, cancer associated with mutation or alteration of p53, brain tumor, bladder cancer, uterocervical cancer, colon cancer, colorectal cancer, non-small cell carcinoma of the lung, small cell carcinoma of the lung, stomach cancer, etc.), lymphoproliferative disorders (e.g., lymphadenopathy), microbial (e.g., viral, bacterial, etc.) infection (e.g., HIV-1 infection, HIV-2 infection, herpes virus infection (including, but not limited to, HSV-1, HSV-2, CMV, VZV, HHV-6, HHV-7, EBV), adenovirus infection, poxvirus infection, human papillorna virus infection, hepatitis infection (e.g., HAV, HBV, HCV, etc.), Helicobacter pylori infection, invasive Staphylococci, etc.), parasitic infection, nephritis, bone disease (e.g., osteoporosis), atherosclerosis, pain, cardiovascular disorders (e.g., neovascularization, hypovascularization or reduced circulation (e.g., ischemic disease (e.g., myocardial infarction, stroke, etc.)), AIDS, allergy, inflammation, neurodegenerative disease (e.g., Alzheimer's disease, Parkinson's disease, amyotrophic lateral sclerosis, pigmentary retinitis, cerebellar degeneration, etc.), graft rejection (acute and chronic), graft vs. host disease, diseases due to osteomyelodysplasia (e.g., aplastic anemia, etc.), joint tissue destruction in rheumatism, liver disease (e.g., acute and chronic hepatitis, liver injury, and cirrhosis), autoimmune disease (e.g., multiple sclerosis, rheumatoid arthritis, systemic lupus erythematosus, immune complex glomerulonephritis, autoimmune diabetes, autoimmune thrombocytopenic purpura, Grave's disease, Hashimoto's thyroiditis, etc.), cardiomyopathy (e.g., dilated cardiomyopathy), diabetes, diabetic complications (e.g., diabetic nephropathy, diabetic neuropathy, diabetic retinopathy), influenza, asthma, psoriasis, glomerulonephritis, septic shock, and ulcerative colitis. [0203] Polynucleotides and/or polypeptides of the invention and/or agonists and/or antagonists thereof are useful in promoting angiogenesis, wound healing (e.g., wounds, burns, and bone fractures).
[0204] Polynucleotides and/or polypeptides of the invention and/or agonists and/or antagonists thereof are also useful as an adjuvant to enhance immune responsiveness to specific antigen and/or anti- viral immune responses.
[0205] More generally, polynucleotides and/or polypeptides of the invention and/or agonists and/or antagonists thereof are useful in regulating (i.e., elevating or reducing) the immune response. For example, polynucleotides and/or polypeptides of the invention may be useful in preparation or recovery from surgery, trauma, radiation therapy, chemotherapy, and transplantation, or may be used to boost immune response and/or recovery in the elderly and immunocompromised individuals. Alternatively, polynucleotides and/or polypeptides of the invention and/or agonists and/or antagonists thereof are useful as immunosuppressive agents, for example in the treatment or prevention of autoimmune disorders. In specific embodiments, polynucleotides and/or polypeptides of the invention are used to treat or prevent chronic inflammatory, allergic or autoimmune conditions, such as those described herein or otherwise known in the art. [0206] The uses of the G-CSF polypeptides, include, but are not limited to, the treatment or prevention of viral hepatitis, herpes viral infections, allergic reactions, adult respiratory distress syndrome, neoplasia, anaphylaxis, allergic asthma, allergen rhinitis, drug allergies (e.g., to penicillin, cephalosporins), primary central nervous system lymphoma (PCNSL), glioblastoma, chronic lymphocytic leukemia (CLL), lymphadenopathy, autoimmune disease, graft versus host disease, rheumatoid arthritis, osteoarthritis, Graves' disease, acute lymphoblastic leukemia (ALL), lymphomas (Hodgkin's disease and non-Hodgkin's lymphoma (NHL)), ophthalmopathy, uveoretinitis, the autoimmune phase of Type 1 diabetes, myasthenia gravis, glomerulonephritis, autoimmune hepatological disorder, autoimmune inflammatory bowel disease, and Crohn's disease. In addition, the G-CSF polypeptides of the present invention may be employed to inhibit neoplasia, such as tumor cell growth. The combination of G-CSF protein with immunotherapeutic agents such as IL-2 or IL-12 may result in synergistic or additive effects that would be useful for the treatment of established cancers. Antibodies [0207] G-CSF-protein specific antibodies for use in the present invention can be raised against the intact G-CSF protein or an antigenic polypeptide fragment thereof, for example a polypeptide fragment of approximately six or more amino acids. The protein or fragment may be presented with or without a carrier protein, such as an albumin, to an animal system (such as rabbit or mouse); in general the polypeptide fragments are sufficiently immunogenic to produce a satisfactory immune response without a carrier if they are at least about 25 amino acids in length.
[0208] Antibodies of the invention include polyclonal and monoclonal antibody preparations, as well as preparations including hybrid antibodies, altered antibodies, chimeric antibodies and, humanized antibodies, as well as: hybrid (chimeric) antibody molecules (see, for example, Winter et al., Nature 349:293-299 (1991)); and U.S. Patent No. 4,816,567); F(ab')2 and F(ab) fragments; Fv molecules (noncovalent heterodimers, see, for example, Inbar et al., Proc. Natl. Acad. Sci. 69:2659-2662 (1972)); and Ehrlich et al. (1980) Biochem 19:4091-4096); single-chain Fv molecules (sFv) (see, e.g., Huston et al., Proc. Natl. Acad. Sci. 85:5879-5883 (1980)); dimeric and trimeric antibody fragment constructs; minibodies (see, e.g., Pack et al., Biochem. 31:1579-1584 (1992); Cumber et al., J. Immunology 149B: 120-126 (1992)); humanized antibody molecules (see, e.g., Riechmann et al., Nature 332:323-327 (1988); Verhoeyan et al., Science 239:1534-1536 (1988)); and any functional fragments obtained from such molecules, wherein such fragments retain specific binding.
[0209] The invention also provides monoclonal antibodies and G-CSF protein binding fragments thereof. Methods of making monoclonal and polyclonal antibodies are known in the art. Monoclonal antibodies are generally antibodies having a homogeneous antibody population. The term is not limited regarding the species or source of the antibody, nor is it intended to be limited by the manner in which it is made. The term encompasses whole immunoglobulins. Monoclonal antibodies of the invention can be prepared using hybridoma technology, for example, Kohler et al., Nature, 256:495 (1975); Kohler et al., Eur. J. Immunol, 6:511 (1976); Kohler et. al., Eur. J. Immunol, 6:292 (1976); Hammerling et al., in: Monoclonal Antibodies and T-Cell Hybridomas, Elsevier, N.Y., (1981) pp. 563-681). In general, such procedures involve immunizing an animal (e.g., a mouse) with an G-CSF protein antigen or with an G-CSF protein- expressing cell. Suitable cells can be recognized by their capacity to bind anti- G-CSF protein antibody. Such cells may be cultured in any suitable tissue culture medium; e.g., in Earle's modified Eagle's medium supplemented with 10% fetal bovine serum (inactivated at about 56°C), and supplemented with about 10 grams/liter of nonessential amino acids, about 1,000 U/ml of penicillin, and about 100 μg/ml of streptomycin. The splenocytes of such mice are extracted and fused with a suitable myeloma cell line. Any suitable myeloma cell line may be employed in accordance with the present invention; e.g., the parent myeloma cell line (SP20), available from the American Type Culture Collection, Manassas, VA. After fusion, the resulting hybridoma cells are selectively maintained in HAT medium, and then cloned by limiting dilution as described by Wands et al., Gastroenterology, 80:225-232 (1981).
[0210] Polyclonal antibodies are generated by immunizing a suitable animal, such as a mouse, rat, rabbit, sheep or goat, with an antigen of interest, such as a stem cell transformed with a gene encoding an antigen. In order to enhance immunogenicity, the antigen can be linked to a carrier prior to immunization. Suitable carriers are typically large, slowly metabolized macromolecules such as proteins, polysaccharides, polylactic acids, polyglycolic acids, polymeric amino acids, amino acid copolymers, lipid aggregates (such as oil droplets or liposomes), and inactive virus particles. Such carriers are well known to those of ordinary skill in the art. Furthermore, the antigen may be conjugated to a bacterial toxoid, such as toxoid from diphtheria, tetanus, cholera, etc., in order to enhance the immunogenicity thereof.
[0211] In addition, techniques developed for the production of chimeric antibodies (Morrison et al., Proc. Natl. Acad. Sci., 81:851-855 (1984); Neuberger et al., Nature, 312:604-608 (1984); Takeda et al., Nature, 314:452-454 (1985)) by splicing genes from a mouse antibody molecule of appropriate antigen specificity together with genes from a human antibody molecule of appropriate biological activity can be used. Chimeric antibodies, i.e., antibodies in which different portions are derived from different animal species, such as those having a variable region derived from a murine monoclonal antibody and a human immunoglobulin constant region, for example, humanized antibodies, and insertion/deletions relating to cdr and framework regions, re suitable for use in the invention.
[0212] The invention also includes humanized antibodies, i.e., those with mostly human immunoglobulin sequences. Humanized antibodies of the invention generally refer to non-human immunoglobulins that have been modified to incorporate portions of human sequences. A humanized antibody may include a human antibody that contains entirely human immunoglobulin sequences.
[0213] The antibodies of the invention may be prepared by any of a variety of methods. For example, cells expressing the G-CSF protein or an antigenic fragment thereof can be administered to an animal in order to induce the production of sera containing polyclonal antibodies. A preparation of G-CSF protein can be prepared and purified to render it substantially free of natural contaminants, and the preparation introduced into an animal in order to produce polyclonal antisera with specific binding activity.
[0214] Antibodies of the invention specifically bind to their respective antigen(s); they may display high avidity and/or high affinity to a specific polypeptide, or more accurately, to an epitope of an antigen. Antibodies of the invention may bind to one epitope, or to more than one epitope. They may display different affinities and/or avidities to different epitopes on one or more molecules. When an antibody binds more strongly to one epitope than to another, adjusting the binding conditions can, in some instances, result in antibody binding almost exclusively to the specific epitope and not to any other epitopes on the same polypeptide, and not to a polypeptide that does not comprise the epitope. G-CSF Protein Antigen [0215] Alternatively, antibodies capable of binding to the G-CSF protein antigen may be produced in a two-step procedure through the use of anti-idiotypic antibodies. Such a method makes use of the fact that antibodies are themselves antigens, and that, therefore, it is possible to obtain an antibody which binds to a second antibody. In accordance with this method, G-CSF -protein specific antibodies are used to immunize an animal, e.g., a mouse. The splenocytes of such an animal are then used to produce hybridoma cells, and the hybridoma cells are screened to identify clones which produce an antibody whose ability to bind to the G-CSF protein-specific antibody can be blocked by the G-CSF protein antigen. Such antibodies comprise anti-idiotypic antibodies to the G-CSF protein-specific antibody and can be used to immunize an animal to induce formation of further G-CSF protein-specific antibodies.
[0216] It will be appreciated that Fab and F(ab')2 and other fragments of the antibodies of the present invention may be used according to the methods disclosed herein. Such fragments are typically produced by proteolytic cleavage, using enzymes such as papain (to produce Fab fragments) or pepsin (to produce F(ab')2 fragments). Alternatively, G-CSF protein-binding fragments can be produced through the application of recombinant DNA technology or through synthetic chemistry. Humanized chimeric monoclonal antibodies are suitable for in vivo use of anti-G-CSF in humans. Such humanized antibodies can be produced using genetic constructs derived from hybridoma cells producing the monoclonal antibodies described above. Methods for producing chimeric antibodies are known in the art. See, for review, Morrison, Science, 229:1202 (1985); Oi et al., BioTechniques, 4:214 (1986); Cabilly et al., U.S. Pat. No. 4,816,567; Taniguchi et al., EP 171496; Morrison et al., EP 173494; Neuberger et al., WO 8601533; Robinson et al., WO 8702671; Boulianne et al., Nature, 312:643 (1984); Neuberger et al., Nature, 314:268 (1985). Diagnosis [0217] This invention is also related to the use of the genes of the present invention as part of a diagnostic assay for detecting diseases or susceptibility to diseases related to the presence of mutations in the nucleic acid sequences encoding the polypeptide of the present invention. Individuals carrying mutations in a gene of the present invention may be detected at the DNA level by a variety of techniques. Nucleic acids for diagnosis may be obtained from a patient's cells, such 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 PCR, for example, as described by 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 a polypeptide of the present invention can be used to identify and analyze mutations. For example, deletions and insertions can be detected by a change in size of the amplified product in comparison to the normal genotype. Point mutations can be identified by hybridizing amplified DNA to radiolabeled RNA or alternatively, radiolabeled antisense DNA sequences. Perfectly matched sequences can be distinguished from mismatched duplexes by RNase A digestion or by differences in melting temperatures. [0218] Genetic testing based on DNA sequence differences may be achieved by detecting alterations in electrophoretic mobility of DNA fragments in gels run with or without denaturing agents. Small sequence deletions and insertions can be visualized by high resolution gel electrophoresis. DNA fragments of different sequences may be distinguished on denaturing formamide 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, for example, as described by Myers et al., Science, 230:1242 (1985).
[0219] Sequence changes at specific locations may also be revealed by nuclease protection assays, such as RNase and SI protection or the chemical cleavage method as shown in Cotton et al., Proc. Natl. Acad. Set, USA, 85:4397-4401 (1985). 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. [0220] The present invention also relates to a diagnostic assay for detecting altered levels of G-CSF proteins in various tissues. An over-expression of these proteins compared to normal control tissue samples may detect the presence of abnormal cellular proliferation, for example, a tumor. Assays used to detect protein levels in a host-derived sample are well-known to those of skill in the art and include radioimmunoassays, competitive-binding assays, Western Blot analysis, ELISA assays, "sandwich" assays, and other assays for the expression levels of the genes encoding the G-CSF proteins known in the art. Expression can be assayed by qualitatively or quantitatively measuring or estimating the level of G-CSF protein, or the level of mRNA encoding G-CSF protein, in a biological sample. Assays may be performed directly, for example, by determining or estimating absolute protein level or mRNA level, or relatively, by comparing the G- CSF protein or mRNA to a second biological sample. In performing these assays, the G- CSF protein or mRNA level in the first biological sample is measured or estimated and compared to a standard G-CSF protein level or mRNA level; suitable standards include second biological samples obtained from an individual not having the disorder of interest. Standards may be obtained by averaging levels of G-CSF in a population of individuals not having a disorder related to G-CSF expression. As will be appreciated in the art, once a standard G-CSF protein level or mRNA level is known, it can be used repeatedly as a standard for comparison.
[0221] An ELISA assay, for example, as described by Coligan, et al., Current Protocols in Immunology, 1(2), Chap. 6, (1991), utilizes an antibody prepared with specificity to a polypeptide antigen of the present invention. In addition, a reporter antibody is prepared against the monoclonal antibody. To the reporter antibody is attached a detectable reagent such as a radioactive tag, a fluorescent tag, or an enzymatic tag, e.g., a horseradish peroxidase. A sample is removed from a host and incubated on a solid support, e.g. a polystyrene dish, that binds the proteins in the sample. Any free protein binding sites on the dish are then covered by incubating with a non-specific protein, e.g., bovine serum albumin. Next, the specific antibody, e.g., a monoclonal antibody, is incubated in the dish, during which time the antibody attaches to any polypeptides of the present invention attached to the polystyrene dish. All unbound monoclonal antibody is washed out with buffer. The reporter antibody, i.e., one linked to horseradish peroxidase is placed in the dish, resulting in the binding of the reporter antibody to any antibody bound to the protein of interest; unattached reporter antibody is then removed. Substrate, e.g., peroxidase, is then added to the dish, and the amount of signal produced color, e.g., developed in a given time period provides a measurement of the amount of a polypeptide of the present invention present in a given volume of patient sample when compared against a standard.
[0222] A competition assay may be employed wherein antibodies specific to a polypeptide of the present invention are attached to a solid support, and labeled G-CSF, along with a sample derived from the host, are passed over the solid support. The label can be detected and quantified, for example, by liquid scintillation chromatography, and the measurement can be correlated to the quantity of the polypeptide of interest present in the sample. A "sandwich" assay, similar to an ELISA assay, may be employed, wherein a polypeptide of the present invention is passed over a solid support and binds to antibody modules attached to the solid support. A second antibody is then bound to the polypeptide of interest. A third antibody, which is labeled and specific to the second antibody is then passed over the solid support and binds to the second antibody. The amount of antibody binding can be quantified; it correlates with the amount of the polypeptide of interest.
[0223] Biological samples of the invention can include any biological sample obtained from a subject, body fluid, cell line, tissue culture, or other source which contains G-CSF protein or mRNA. As indicated, biological samples include body fluids (such as sera, plasma, urine, synovial fluid, and spinal fluid) which contain free G-CSF protein, ovarian or renal system tissue, and other tissue sources found to express complete or mature G-CSF polypeptide or an G-CSF receptor. Methods for obtaining tissue biopsies and body fluids from mammals are well known in the art. Where the biological sample is to include mRNA, a tissue biopsy may provide the source. [0224] Total cellular RNA can be isolated from a biological sample using any suitable technique such as the single-step guanidinium-thiocyanate-phenol-chloroform method described in Chomczynski and Sacchi, Anal. Biochem., 162:156-159 (1987). Levels of mRNA encoding the G-CSF protein are then assayed using any appropriate method. These include Northern blot analysis, S 1 nuclease mapping, PCR, reverse transcription in combination with PCR (RT-PCR), and reverse transcription in combination with the ligase chain reaction (RT-LCR).
[0225] Assaying G-CSF protein levels in a biological sample can be performed using antibody-based techniques. For example, G-CSF protein expression in tissues can be studied with classical immunohistological methods, for example, Jalkanen, M., et al., J. Cell. Biol, 101:976-985 (1985); Jalkanen, M., et al., J. Cell. Biol, 105:3087-3096 (1987). Other antibody-based methods useful for detecting G-CSF protein gene expression include immunoassays, such as the enzyme linked immunosorbent assay (ELISA) and the radioimmunoassay (RIA). Suitable antibody assay labels are known in the art and include enzyme labels, such as glucose oxidase, radioisotopes, and fluorescent labels, such as fluorescein and rhodamine, and biotin.
[0226] In addition to assaying G-CSF protein levels in a biological sample obtained from an individual, G-CSF protein can also be detected in vivo by imaging. Antibody labels or markers for in vivo imaging of G-CSF protein include those detectable by X- radiography, NMR, or ESR. For X-radiography, suitable labels include radioisotopes such as barium or cesium, which emit detectable radiation but are not overtly harmful to a subject. Suitable markers for NMR and ESR include those with a detectable characteristic spin, such as deuterium, which may be incorporated into the antibody by labeling of nutrients for the relevant hybridoma.
[0227] An G-CSF protein-specific antibody or antibody fragment which has been labeled with an appropriate detectable imaging moiety, such as a radioisotope, a radio- opaque substance, or a material detectable by nuclear magnetic resonance, is introduced, for example, parenterally, subcutaneously or intraperitoneally, into the subject to be examined for an immune system disorder. It will be understood in the art that the size of the subject and the imaging system used will determine the quantity of imaging moiety needed to produce diagnostic images. The labeled antibody or antibody fragment will then preferentially accumulate at the location of cells which contain G-CSF protein. In vivo tumor imaging is described in Burchiel et al., ed., Chapter 13, Tumor Imaging: The Radiochemical Detection of Cancer, Masson Publishing Inc. (1982). Formulations [0228] The G-CSF polypeptide compositions will be formulated and dosed in a fashion consistent with good medical practice, taking into account the clinical condition of the individual subject, the site of delivery of the G-CSF polypeptide composition, the method of administration, the scheduling of administration, and other factors known to practitioners. The effective amount of G-CSF polypeptide for purposes herein is thus determined by such considerations.
[0229] The polypeptides, agonists, and antagonists of the present invention may be employed in combination with a suitable pharmaceutical carrier to comprise a pharmaceutical composition for parenteral administration. Such compositions comprise a therapeutically effective amount of the polypeptide, agonist, or antagonist and a pharmaceutically 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. [0230] 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(s) can be a notice in the form prescribed by a governmental agency regulating the manufacture, use or sale of pharmaceuticals or biological products, which notice reflects approval by the agency of manufacture, use or sale for human administration. In addition, the polypeptides, agonists and antagonists of the present invention may be employed in conjunction with other therapeutic compounds.
[0231] In pharmaceutical dosage forms, the compositions of the invention can be administered in the form of their pharmaceutically acceptable salts, or they can also be used alone or in appropriate association, as well as in combination, with other pharmaceutically active compounds. The subject compositions are formulated in accordance to the mode of potential administration. Administration of the agents can be achieved in various ways, including oral, buccal, nasal, rectal, parenteral, intraperitoneal, intradermal, transdermal, subcutaneous, intravenous, intra-arterial, intracardiac, intraventricular, intracranial, intratracheal, and intrathecal administration, etc., or otherwise by implantation or inhalation. Thus, the subject compositions can be formulated into preparations in solid, semi-solid, liquid or gaseous forms, such as tablets, capsules, powders, granules, ointments, solutions, suppositories, enemas, injections, inhalants and aerosols. The following methods and excipients are merely exemplary and are in no way limiting. In general, they are administered in an amount of at least about 10 micrograms/kg body weight and in most cases they will be administered in an amount not in excess of about 8 milligrams/kg body weight per day.
[0232] The polypeptides of the invention, and agonist and antagonist compounds which are polypeptides, may also be employed in accordance with the present invention by expression of such polypeptides in vivo, i.e., gene therapy. Thus, for example, cells may be engineered with a polynucleotide (DNA or RNA) encoding for the polypeptide ex vivo; the engineered cells are then provided to a patient. 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 for the polypeptide of the present invention. [0233] Similarly, cells may be engineered in vivo for expressing the polypeptide in vivo, for example, by procedures known in the art. As known in the art, a cell producing a retroviral particle containing RNA encoding the polypeptide of the present invention may be administered to a patient for the purpose of engineering cells in vivo and expressing the polypeptide in vivo. These and other methods for administering a polypeptide of the present invention by similar methods 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 retroviral particle, for example, an adenovirus, which may be used to engineer cells in vivo after combination with a suitable delivery vehicle.
[0234] Retroviruses from which the retroviral plasmid vectors hereinabove mentioned may be derived include, but are not limited to, Moloney Murine Leukemia virus, spleen necrosis virus, retroviruses such as Rous sarcoma virus, Harvey sarcoma virus, avian leukosis virus, gibbon ape leukemia virus, human immunodeficiency virus, adenovirus, myeloproliferative sarcoma virus, and mammary tumor virus. In one embodiment, the retroviral plasmid vector is derived from Moloney murine leukemia virus.
[0235] Promoters suitable for these formulations include the viral vectors listed above, as well as include the respiratory syncytial virus (RSV) promoter; inducible promoters, such as the MMT promoter, the metallothionein promoter; heat shock promoters; the albumin promoter; the ApoAl promoter; human globin promoters; viral thymidine kinase promoters, such as the Herpes Simplex thymidine kinase promoter; retroviral LTRs (including the modified retroviral LTRs hereinabove described); the beta- actin promoter; and human growth hormone promoters. The promoter also may be the native promoter which controls the gene encoding the polypeptide. The selection of a suitable promoter will be apparent to those skilled in the art from the teachings contained herein.
[0236] A retroviral plasmid vector can be employed to transduce packaging cell lines to form producer cell lines. Examples of packaging cells which may be transfected include, but are not limited to, the PE501, PA317, -2, -AM, PA12, T19-14X, VT-19-17- H2, CRE, CRIP, GP+E-86, GP+envAml2, and DAN cell lines as described in Miller, Human Gene Therapy, 1:5-14 (1990). The vector may transduce the packaging cells through any means known in the art. Such means include, but are not limited to, electroporation, the use of liposomes, and CaPO4 precipitation. In one alternative, the retroviral plasmid vector may be encapsulated into a liposome, or coupled to a lipid, and then administered to a host.
[0237] The producer cell line generates infectious retroviral vector particles which include the nucleic acid sequence(s) encoding the polypeptides. Such retroviral vector particles then may be employed to transduce eukaryotic cells, either in vitro or in vivo. The transduced eukaryotic cells will express the nucleic acid sequence(s) encoding the polypeptide. Eukaryotic cells which may be transduced include, but are not limited to, embryonic stem cells, embryonic carcinoma cells, as well as hematopoietic stem cells, hepatocytes, fibroblasts, myoblasts, keratinocytes, endothelial cells, and bronchial epithelial cells. G-CSF "Knock-outs" and Homologous Recombination [0238] Endogenous gene expression can be reduced by inactivating or "knocking out" a gene of interest and/or its promoter using targeted homologous recombination, (e.g., see Smithies et al., Nature, 317:230-234 (1985); Thomas & Capecchi, Cell, 51:503- 512 (1987); Thompson et al., Cell, 5:313-321 (1989); each of which is incorporated by reference herein in its entirety). For example, a mutant, non-functional polynucleotide of the invention (or a completely unrelated DNA sequence) flanked by DNA homologous to the endogenous polynucleotide sequence (either the coding regions or regulatory regions of the gene) can be used, with or without a selectable marker and/or a negative selectable marker, to transfect cells that express polypeptides of the invention in vivo. In another embodiment, techniques known in the art are used to generate knockouts in cells that contain, but do not express, the gene of interest, insertion of the DNA construct, via targeted homologous recombination, results in inactivation of the targeted gene. Such approaches are particularly suited in research and agricultural fields where modifications to embryonic stem cells can be used to generate animal offspring with an inactive targeted gene (see, e.g., Thomas & Capecchi (1987) supra; Thompson (1989), supra). However, this approach can be routinely adapted for use in humans provided the recombinant DNA constructs are directly administered or targeted to the required site in vivo using appropriate viral vectors that will be apparent to those of skill in the art. [0239] In further embodiments of the invention, cells that are genetically engineered to express the polypeptides of the invention, or alternatively, that are genetically engineered not to express the polypeptides of the invention (e.g., knockouts) are administered to a patient in vivo. Such cells may be obtained from the patient (i.e., animal, including human) or an MHC compatible donor and can include, but are not limited to fibroblasts, bone marrow cells, blood cells (e.g., lymphocytes), adipocytes, muscle cells, endothelial cells, etc. The cells are genetically engineered in vitro using recombinant DNA techniques to introduce the coding sequence of polypeptides of the invention into the cells, or alternatively, to disrupt the coding sequence and/or endogenous regulatory sequence associated with the polypeptides of the invention, e.g., by transduction (using viral vectors, and/or vectors that integrate the transgene into the cell genome) or transfection procedures, including, but not limited to, the use of plasmids, cosmids, YACs, naked DNA, electroporation, liposomes, etc. The coding sequence of the polypeptides of the invention can be placed under the control of a strong constitutive or inducible promoter or promoter/enhancer to achieve expression, and secretion, of the polypeptides of the invention. The engineered cells which express and secrete the polypeptides of the invention can be introduced into the patient systemically, e.g., in the circulation, or intraperitoneally. Alternatively, the cells can be incorporated into a matrix and implanted in the body, e.g., genetically engineered fibroblasts can be implanted as part of a skin graft; genetically engineered endothelial cells can be implanted as part of a lymphatic or vascular graft. (See, for example, Anderson et al. U.S. Pat. No. 5,399,349; and Mulligan & Wilson, U.S. Pat. No. 5,460,959, each of which is incorporated by reference herein in its entirety).
[0240] When the cells to be administered are non-autologous or non-MHC compatible cells, they can be administered using well known techniques which prevent the development of a host immune response against the introduced cells. For example, the cells may be introduced in an encapsulated form which, while allowing for an exchange of components with the immediate extracellular environment, does not allow the introduced cells to be recognized by the host immune system. Transgenic Non-Human Animals [0241] The polypeptides of the invention can also be expressed in transgenic animals. Animals of any species, including, but not limited to, mice, rats, rabbits, hamsters, guinea pigs, pigs, micro-pigs, goats, sheep, cows, and non-human primates, e.g., baboons, monkeys, and chimpanzees may be used to generate transgenic animals. In a specific embodiment, techniques described herein or otherwise known in the art, are used to express polypeptides of the invention in humans, as part of a gene therapy protocol.
[0242] Any technique known in the art may be used to introduce the transgene (i.e., polynucleotides of the invention) into animals to produce a founder lines of transgenic animals. Such techniques include, but are not limited to, pronuclear microinjection (Paterson et al., Appl Microbiol. Biotechnol. 40:691-698 (1994); Carver et al., Biotechnology (NY) 11: 1263-1270 (1993); Wright et al., Biotechnology (NY) 9:830-834 (1991); and Hoppe et al., U.S. Pat. No. 4,873,191 (1989)); retrovirus mediated gene transfer into germ lines (Van der Putten et al., Proc. Natl. Acad. Set, USA 82:6148-6152 (1985)), blastocysts or embryos; gene targeting in embryonic stem cells (Thompson et al., Cell 56:313-321 (1989)); electroporation of cells or embryos (Lo, Mol Cell. Biol. 3:1803- 1814 (1983)); introduction of the polynucleotides of the invention using a gene gun (see, e.g., Ulmer et al., Science 259:1745 (1993); introducing nucleic acid constructs into embryonic pluripotent stem cells and transferring the stem cells back into the blastocyst; and sperm-mediated gene transfer (Lavitrano et al., Cell 57:717-723 (1989); etc. For a review of such techniques, see Gordon, Intl. Rev. Cytol. 115:171-229 (1989), which is incorporated by reference herein in its entirety. Further, the contents of each of the documents recited in this paragraph is herein incorporated by reference in its entirety. See also, U.S. Pat. No. 5,464,764 (Capecchi et al., Positive-Negative Selection Methods and Vectors); U.S. Pat. No. 5,631,153 (Capecchi et al., Cells and Non-Human Organisms Containing Predetermined Genomic Modifications and Positive-Negative Selection Methods and Vectors for Making Same); U.S. Pat. No. 4,736,866 (Leder et al., Transgenic Non-Human Animals); and U.S. Pat. No. 4,873,191 (Wagner et al., Genetic Transformation of Zygotes); each of which is hereby incorporated by reference in its entirety. Any technique known in the art may be used to produce transgenic clones containing polynucleotides of the invention, for example, nuclear transfer into enucleated oocytes of nuclei from cultured embryonic, fetal, or adult cells induced to quiescence (Campbell et al., Nature 380:64-66 (1996); Wilmut et al., Nature 385:810-813 (1997)), each of which is herein incorporated by reference in its entirety).
[0243] The present invention provides for transgenic animals that carry the transgene in all their cells, as well as animals which carry the transgene in some, but not all their cells, i.e., mosaic or chimeric animals. The transgene may be integrated as a single transgene or as multiple copies such as in concatamers, e.g., head-to-head tandems or head-to-tail tandems. The transgene may also be selectively introduced into and activated in a particular cell type by following, for example, the teaching of Lasko et al. (Proc. Natl. Acad. Sci. USA 89:6232-6236 (1992)). The regulatory sequences required for such a cell-type specific activation will depend upon the particular cell type of interest, and will be apparent to those of skill in the art. It may be desired that the polynucleotide transgene be integrated into the chromosomal site of the endogenous gene, gene targeting is then suitable. Briefly, when such a technique is to be utilized, vectors containing some nucleotide sequences homologous to the endogenous gene are designed for the purpose of integrating, via homologous recombination with chromosomal sequences, into and disrupting the function of the nucleotide sequence of the endogenous gene. The transgene may also be selectively introduced into a particular cell type, thus inactivating the endogenous gene in only that cell type, by following, for example, the teaching of Gu et al. (Science 265:103-106 (1994)). The regulatory sequences required for such a cell-type specific inactivation will depend upon the particular cell type of interest, and will be apparent to those of skill in the art. [0244] Once transgenic animals have been generated, the expression of the recombinant gene may be assayed utilizing standard techniques. Initial screening may be accomplished by Southern blot analysis or PCR techniques to analyze animal tissues to verify that integration of the transgene has taken place. The level of mRNA expression of the transgene in the tissues of the transgenic animals may also be assessed using techniques which include, but are not limited to, Northern blot analysis of tissue samples obtained from the animal, in situ hybridization analysis, and reverse transcriptase-PCR (rt-PCR). Samples of transgenic gene-expressing tissue may also be evaluated immunocytochemically or immunohistochemically using antibodies specific for the transgene product.
[0245] Once the founder animals are produced, they may be bred, inbred, outbred, or crossbred to produce colonies of the particular animal. Examples of such breeding strategies include, but are not limited to outbreeding of founder animals with more than one integration site in order to establish separate lines; inbreeding of separate lines in order to produce compound transgenics that express the transgene at higher levels because of the effects of additive expression of each transgene; crossing of heterozygous transgenic animals to produce animals homozygous for a given integration site in order to both augment expression and eliminate the need for screening of animals by DNA analysis; crossing of separate homozygous lines to produce compound heterozygous or homozygous lines; and breeding to place the transgene on a distinct background that is appropriate for an experimental model of interest.
[0246] Transgenic and "knock-out" animals of the invention have uses which include, but are not limited to, animal model systems useful in elaborating the biological function of G-CSF polypeptides, studying conditions and/or disorders associated with aberrant G-CSF expression, and in screening for compounds effective in ameliorating such conditions and/or disorders. Kits [0247] The present invention provides kits that can be used in the above methods. In one embodiment, a kit comprises an antibody of the invention, e.g., a purified antibody, in one or more containers. In a specific embodiment, the kits of the present invention contain a substantially isolated polypeptide comprising an epitope which is specifically immunoreactive with an antibody included in the kit. The kits of the present invention may also comprise a control antibody which does not react with the polypeptide of interest.
[0248] In another embodiment, the kits of the present invention comprise a means for detecting the binding of an antibody to a polypeptide of interest (e.g., the antibody may be conjugated to a detectable substrate such as a fluorescent compound, an enzymatic substrate, a radioactive compound or a luminescent compound, or a second antibody which recognizes the first antibody may be conjugated to a detectable substrate).
[0249] In another embodiment of the present invention, the kit is a diagnostic kit for use in screening serum containing antibodies specific against proliferative and/or cancerous polynucleotides and polypeptides. Such a kit may include a control antibody that does not react with the polypeptide of interest. Such a kit may include a substantially isolated polypeptide antigen comprising an epitope which is specifically immunoreactive with at least one anti-polypeptide antigen antibody. Further, such a kit includes means for detecting the binding of said antibody to the antigen (e.g., the antibody may be conjugated to a fluorescent compound such as fluorescein or rhodamine which can be detected by flow cytometry). In embodiments, the kit may include a recombinantly produced or chemically synthesized polypeptide antigen. The polypeptide antigen of the kit may also be attached to a solid support.
[0250] In a further embodiment, the detecting means of the above-described kit includes a solid support to which said polypeptide antigen is attached. Such a kit may also include a non-attached reporter-labeled anti-human antibody. In this embodiment, binding of the antibody to the polypeptide antigen can be detected by binding of the said reporter-labeled antibody.
[0251] In an additional embodiment, the invention includes a diagnostic kit for use in screening serum containing antigens of the polypeptide of the invention. The diagnostic kit includes a substantially isolated antibody specifically immunoreactive with polypeptide or polynucleotide antigens, and means for detecting the binding of the polynucleotide or polypeptide antigen to the antibody. In one embodiment, the antibody is attached to a solid support. In an embodiment, the antibody is a monoclonal antibody. The detecting means of the kit may include a second, labeled monoclonal antibody. Alternatively, or in addition, the detecting means may include a labeled, competing antigen.
[0252] In one diagnostic configuration, test serum is reacted with a solid phase reagent having a surface-bound antigen obtained by the methods of the present invention. After binding with specific antigen antibody to the reagent and removing unbound serum components by washing, the reagent is reacted with reporter-labeled anti-human antibody to bind reporter to the reagent in proportion to the amount of bound anti-antigen antibody on the solid support. The reagent is again washed to remove unbound labeled antibody, and the amount of reporter associated with the reagent is determined. Typically, the reporter is an enzyme which is detected by incubating the solid phase in the presence of a suitable fluorometric, luminescent or colorimetric substrate (Sigma, St. Louis, Mo.). [0253] The solid surface reagent in the above assay is prepared by known techniques for attaching protein material to solid support material, such as polymeric beads, dip sticks, 96-well plates and/or filter material. These attachment methods generally include non-specific adsorption of the protein to the support or covalent attachment of the protein, typically through a free amine group, to a chemically reactive group on the solid support, such as an activated carboxyl, hydroxyl, or aldehyde group. Alternatively, streptavidin coated plates can be used in conjunction with biotinylated antigen(s). [0254] All publications, patents and patent applications cited herein, whether supra or infra, are hereby incoiporated by reference in their entirety.
[0255] As used in this specification and the appended claims, the singular forms "a," "an," and "the" include plural referents unless the content clearly dictates otherwise. Thus, for example, reference to "an antigen" includes a mixture of two or more antigens, a reference to "a subject polypeptide" includes a plurality of such polypeptides, and reference to "the agent" includes reference to one or more agents and equivalents thereof known to those skilled in the art, and so forth.
[0256] It will be clear that the invention may be practiced otherwise than as particularly described in the foregoing description and examples. Numerous modifications and variations of the present invention are possible in light of the above teachings and, therefore, are within the scope of the appended claims. [0257] It is to be understood that both the foregoing general description and the following detailed description are exemplary and explanatory only and are not restrictive of the invention, as claimed. Moreover, it must be understood that the invention is not limited to the particular embodiments described, as such may, of course, vary. Further, the terminology used to describe particular embodiments is not intended to be limiting, since the scope of the present invention will be limited only by its claim. [0258] Unless defined otherwise, the meanings of all technical and scientific terms used herein are those commonly understood by one of ordinary skill in the art to which this invention belongs. One of ordinary skill in the art will also appreciate that any methods and materials similar or equivalent to those described herein can also be used to practice or test the invention.
[0259] Further, all numbers expressing quantities of ingredients, reaction conditions, % purity, polypeptide and polynucleotide lengths, and so forth, used in the specification and claims, are modified by the term "about," unless otherwise indicated. Accordingly, the numerical parameters set forth in the specification and claims are approximations that may vary depending upon the desired properties of the present invention. At the very least, and not as an attempt to limit the application of the doctrine of equivalents to the scope of the claims, each numerical parameter should at least be construed in light of the number of reported significant digits, applying ordinary rounding techniques. [0260] Nonetheless, the numerical values set forth in the specific examples are reported as precisely as possible. Any numerical value, however, inherently contains certain errors from the standard deviation of its experimental measurement. [0261] Patents and publications cited are incorporated by reference herein in their entireties; references cited in such publications are also incorporated by reference in their entireties.
Tables
Table 1. SEQ. ID. NOS.:l-23
Figure imgf000082_0001
Figure imgf000083_0001
Figure imgf000084_0001
Table 3. Characteristics of G-CSF Sequences
Table 4. Comparison of CLN00489695_5pvl with Known Sequences
Figure imgf000086_0001
Table 5. Comparison of CLN00493523_5pvl with Known Sequences
Figure imgf000087_0001

Claims

91 Claims 1. An isolated first nucleic acid molecule comprising a first polynucleotide sequence chosen from (A) SEQ ID NOS.: 1-2 and/or 19-20, (B) a polynucleotide sequence encoding a polypeptide of SEQ ID NOS.:9 and/or 10, and (C) biologically active fragments of any of these, wherein the fragments comprise a polynucleotide sequence or an amino acid sequence encoded by a polynucleotide sequence comprising the splice sites utilized by CLN00489695 and/or CLN00493523.
2. The first nucleic acid molecule of claim 1 , wherein the nucleic acid molecule is chosen from a cDNA molecule, a genomic DNA molecule, a cRNA molecule, a siRNA molecule, a RNAi molecule, and a mRNA molecule.
3. A double-stranded isolated nucleic acid molecule comprising the first nucleic acid molecule of claim 1 and its complement.
4. A second nucleic acid molecule comprising a second polynucleotide sequence complementary to the first nucleic acid molecule of claim 1.
5. The second nucleic acid molecule of claim 4, chosen from a RNAi molecule, an anti-sense molecule, and a ribozyme.
6. An isolated polypeptide comprising an amino acid sequence chosen from SEQ ID NOS.:9-10 and biologically active fragments of either of these.
7. The polypeptide of claim 6, wherein the polypeptide is present in a cell culture.
8. The polypeptide of claim 7, wherein the cell culture is chosen from a bacterial cell culture, a mammalian cell culture, and an insect cell culture.
9. An isolated polypeptide encoded by the first nucleic acid molecule of claim 1.
10. A vector comprising the nucleic acid molecule of claim 1 and a promoter that regulates the expression of the nucleic acid molecule.
11. The vector of claim 10, wherein the promoter is chosen from one that is naturally contiguous to the nucleic acid molecule and one that is not naturally contiguous to the nucleic acid molecule.
12. The vector of claim 11, wherein the promoter is chosen from an inducible promoter, a conditionally-active promoter, a constitutive promoter, and a tissue-specific promoter. 92
13. A recombinant host cell comprising the nucleic acid molecule of claim 1, the polypeptide of claim 6 or claim 9, and/or the vector of claim 10.
14. The host cell of claim 13, wherein the cell is chosen from a prokaryotic cell and a eukaryotic cell.
15. The host cell of claim 14, wherein the eukaryotic cell is chosen from a human cell, a non-human mammalian cell, an insect cell, a fish cell, a plant cell, and a fungal cell.
16. A non-human animal injected with a nucleic acid molecule of claim 1.
17. A non-human animal transformed with a nucleic acid molecule of claim 1.
18. A non-human animal injected with a polypeptide of claim 6 or claim 9.
19. A nucleic acid composition comprising the nucleic acid molecule of claim 1 and a carrier.
20. A polypeptide composition comprising the polypeptide molecule of claim 6 or claim 9 and a carrier.
21. A vector composition comprising the vector of claim 10 and a carrier.
22. A host cell composition comprising the host cell of claim 13 and a carrier.
23. The composition of any of claims 19-22 wherein the carrier is a pharmaceutically acceptable carrier or excipient.
24. A method of producing a recombinant host cell comprising: (a) providing a composition comprising a vector that comprises the nucleic acid molecule of claim 1; and (b) allowing a host cell to come into contact with the vector to form a recombinant host cell.
25. A method of producing a polypeptide comprising: (a) providing a composition comprising the recombinant host cell of claim 13; and (b) culturing the recombinant host cell to produce the polypeptide.
26. A method of producing a polypeptide comprising: (a) providing the nucleic acid of claim 1 ; and (b) expressing the nucleic acid molecule in a cell free expression system to produce the polypeptide. 93
27. The method of claim 26, wherein the cell free expression system is chosen from a wheat germ lysate expression system, a rabbit reticulocyte expression system, and an E. coli lysate expression system.
28. A diagnostic kit comprising a composition comprising a nucleic acid molecule complementary to the nucleic acid molecule of claim 1 and a vehicle.
29. A diagnostic kit comprising an antibody that specifically binds to the polypeptide of claim 6 or claim 9 or a biologically active fragment of either of these.
30. A diagnostic kit comprising the polypeptide of claim 6 or claim 9 or a biologically active fragment of either of these.
31. The diagnostic kit of claim 28, further comprising instructions for use.
32. The diagnostic kit of claim 29, further comprising instructions for use.
33. The diagnostic kit of claim 30, further comprising instructions for use.
34. A method of determining the presence of the nucleic acid molecule of claim 1 in a sample comprising: (a) providing the nucleic acid molecule of claim 1 ; (b) allowing the nucleic acid molecule of claim 1 to interact with the sample under conditions that allow for specific binding; and (c) determining whether specific binding has occurred.
35. A method of determining the presence of an antibody specific to apolypeptide of claim 6 or claim 9 in a sample comprising: (a) providing a composition comprising the polypeptide; (b) allowing the polypeptide to interact with the sample under conditions hat allow for specific binding; and (c) determining whether interaction has occurred between the polypeptide and the antibody.
36. An antibody that specifically binds to or interferes with activity of the polypeptide of claim 6 or claim 9.
37. The antibody of claim 35, wherein the antibody is a chosen from a polyclonal antibody, a monoclonal antibody, a single chain antibody, and a biologically active fragment of any of these.
38. The antibody of claim 37, wherein the antibody is a fragment chosen from an antigen binding fragment, an Fc fragment, a cdr fragment, and a framework fragment. 94
39. A method for treating or preventing neutropenia in a subject comprising: (a) providing a composition comprising a polypeptide chosen from SEQ ID NOS.:9-10 and biologically active fragments of either of these, and a carrier; and (b) administering the composition to the subject.
40. The method of claim 39 wherein the neutropenia is induced by chemotherapy or radiotherapy.
41. A method for mobilizing peripheral blood stem cells in a subject comprising: (a) providing a composition comprising a polypeptide chosen from SEQ ID NOS.:9-10 and biologically active fragments of either of these, and a carrier; and (b) administering the composition to the subject.
42. A method for mobilizing bone marrow stem cells in a subject comprising: (a) providing a composition comprising a polypeptide chosen from SEQ ID NOS.:9-10 and biologically active fragments of either of these, and a carrier; and (b) administering the composition to the subject.
43. A method for treating or preventing an infection in a subject comprising: (a) providing a composition comprising a polypeptide chosen from SEQ ID NOS.-.9-10 and biologically active fragments of either of these, and a carrier; and (b) administering the composition to the subject.
44. The method of claim 43, wherein the infection is a bacterial, fungal, parasitic, or viral infection.
45. The method of claim 43, further comprising administering granulocyte transfusion therapy.
46. A method for treating or preventing septic shock in a subject comprising: (a) providing a composition comprising a polypeptide chosen from SEQ ID NOS.:9-10 and biologically active fragments of either of these, and a carrier; and (b) administering the composition to the subject. 95 47. The method of any of 39-46, wherein the polypeptide further comprises at least one fusion partner. 48. The method of claim 47, wherein the fusion partner is chosen from a polymer, a polypeptide, a succinyl group, serum albumin, fetuin, a leucine zipper domain, a tetranectin trimerization domain, an Fc region, and a trimerization domain from a mannose binding protein. 49. The method of claim 48, wherein the polymer is a polyethylene glycol moiety. 50. The method of claim 49, wherein the polyethylene glycol moiety is attached through an amino group of an amino acid of the polypeptide. 51. The method of claim 49, wherein the polyethylene glycol moiety is either a branched or a linear chain polymer. 52. A method for treating or preventing an inflammatory disease in a subject comprising: (a) providing a composition comprising a polypeptide chosen from SEQ ID NOS.:9-10 and biologically active fragments of either of these, and a carrier; and (b) administering the composition to the subject. 53. The method of claim 52, wherein the inflammatory disease is chosen from rheumatoid arthritis, myocarditis, inflammatory bowel disease, Crohn's disease, psoriasis, multiple sclerosis, systemic lupus erythematosus, ocular inflammation, intraocular inflammation, surgery, injury related inflammation, osteoarthritis, and central nervous system inflammation. 54. The method of claim 53, wherein the ocular inflammation is chosen from iritis, scleritis, orbital pseudotumor, corneal melt, ocular cicatricial pemphigoid, and Sjδgren's syndrome. 55. The method of any of claims 52-54, wherein the polypeptide further comprises at least one fusion partner. 56. The method of claim 55, wherein the fusion partner is chosen from a polymer, a polypeptide, a succinyl group, and serum albumin. 57. The method of claim 56, wherein the polymer is a polyethylene glycol moiety. 58. The method of claim 57, wherein the polyethylene glycol moiety is attached through an amino group of an amino acid of the polypeptide. 96 59. The method of claim 57, wherein the polyethylene glycol moiety is either a branched or a linear chain polymer. 60. The composition of claim 20, wherein the polypeptide further comprises at least one fusion partner. 61. The composition of claim 60, wherein the fusion partner is chosen from a polymer, a polypeptide, a succinyl group, serum albumin, fetuin, a leucine zipper domain, a tetranectin trimerization domain, an Fc region, and a trimerization domain from a mannose binding protein. 62. The composition of claim 61, wherein the polymer is a polyethylene glycol moiety. 63. The composition of claim 62, wherein the polyethylene glycol moiety is attached through an amino group of an amino acid of the polypeptide. 64. The composition of claim 62, wherein the polyethylene glycol moiety is either a branched or linear chain polymer. 65. A method for treating or preventing an ischemic disease in a subject comprising: (a) providing a composition comprising a polypeptide chosen from SEQ ID NOS.:9-10 and biologically active fragments of either of these, and a carrier; and (b) administering the composition to the subject. 66. The method of claim 65, wherein the ischemic disease is chosen from stroke, myocardial infarction, and fulminant liver failure. 67. A method for treating or preventing an immune disorder or disease in a subject comprising: (a) providing a composition comprising a polypeptide chosen from SEQ ID NOS.:9-10 and biologically active fragments of either of these, and a carrier; and . (b) administering the composition to the subject. 68. The method of claim 67, wherein the immune disease is a B-lymphocytic disease. 69. A method for suppressing immune activity in a subject comprising: 97 (a) providing a composition comprising a polypeptide chosen from SEQ ID NOS.:9-10 and biologically active fragments of either of these, and a carrier; and (b) administering the composition to the subject. 70. The method of claim 69, wherein the immune disease is a B-Iymphocytic disease. 71. A method for modulator screening comprising: (a) providing a polypeptide chosen from claims 6, 9, 25, 26, and biologically active fragments thereof; and (b) using the polypeptide of (a) as a target in an assay that detects the modulator. 72. The method of claim 71, wherein the modulator is a small molecule drug. 73. The method of claim 71, wherein the modulator is an antibody. 74. The method of any of claims 38-46, 52-54, or 60-65, wherein the composition is administered to the subject locally or systemically. 75. The method of any of claims 38-46, 52-54, or 60-65, wherein the polypeptide is encoded by a nucleic acid molecule comprising a nucleotide sequence chosen from SEQ ID NOS. : 1-2, 19-20, and biologically active fragments thereof. 76. The method of any of claims 65-70, wherein the polypeptide further comprises at least one fusion partner. 77. The method of claim 76, wherein the fusion partner is chosen from a polymer, a polypeptide, a succinyl group, serum albumin, fetuin, a leucine zipper domain, a tetramectin trimerization domain, an Fc region, and a trimerization domain from a mannose protein. 78. The method of claim 77, where in the polymer is a polyethylene glycol moiety. 79. The method of claim 78, wherein the polyethylene glycol moiety is attached through an amino group of an amino acid of the polypeptide. 80. The method of claim 78, wherein the polyethylene glycol moiety is either a branched or linear chain polymer. 98 81. The polypeptide of any of claims 6, 9, 25, or 26, wherein at least one cysteiήe residue is replaced by an amino acid chosen from a polar amino acid, a basic amino acid, an acidic amino acid, and a neutral amino acid. 82. The polypeptide of claim 81, wherein the neutral amino acid is chosen from serine and alanine. 83. The polypeptide of claim 82, wherein the neutral amino acid is serine. 84. The polypeptide of claim 81, wherein the polypeptide further comprises at least one fusion partner. 85. The method of claim 84, wherein the fusion partner is chosen from a polymer, a polypeptide, a succinyl group, serum albumin, fetuin, a leucine zipper domain, a tetramectin trimerization domain, an Fc region, and a trimerization domain from a mannose binding protein. 86. The method of claim 85, wherein the polymer is a polyethylene glycol moiety. 87. The method of claim 86, wherein the polyethylene glycol moiety is attached through an amino group of an amino acid of the polypeptide. 88. The method of claim 86, wherein the polyethylene glycol moiety is either a branched or linear chain polymer. 89. A polypeptide composition comprising a polypeptide of any of claims 6, 9, 25, or 26 and a growth factor. 90. The polypeptide composition of claim 89, wherein the growth factor is chosen from erythropoetin, GM-CSF, and M-CSF.
PCT/US2005/019491 2004-06-04 2005-06-03 Novel g-csf polypeptides, polynucleotides, modulators thereof, and methods of use WO2005121174A2 (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US57731604P 2004-06-04 2004-06-04
US60/577,316 2004-06-04

Publications (2)

Publication Number Publication Date
WO2005121174A2 true WO2005121174A2 (en) 2005-12-22
WO2005121174A3 WO2005121174A3 (en) 2006-05-04

Family

ID=35445873

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2005/019491 WO2005121174A2 (en) 2004-06-04 2005-06-03 Novel g-csf polypeptides, polynucleotides, modulators thereof, and methods of use

Country Status (1)

Country Link
WO (1) WO2005121174A2 (en)

Cited By (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US7790174B2 (en) * 2004-12-23 2010-09-07 Merck Serono Sa G-CSF polypeptides and uses thereof
WO2011032204A1 (en) * 2009-09-15 2011-03-24 Csl Limited Treatment of neurological conditions
EP3757117A1 (en) * 2015-11-16 2020-12-30 Ubiprotein, Corp. A method for extending half-life of a protein

Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5665863A (en) * 1992-01-31 1997-09-09 Rhone-Poulenc Rorer S.A. Polypeptides having granulocyte colony stimulating activity, their preparation and pharmaceutical compositions containing them
US20030082679A1 (en) * 2001-10-01 2003-05-01 Sun Lee-Hwei K. Fc fusion proteins of human granulocyte colony-stimulating factor with increased biological activities
WO2003081238A2 (en) * 2002-03-22 2003-10-02 Ludwig Maximilians Universität Cytocapacity test
WO2003093427A2 (en) * 2002-04-30 2003-11-13 Incyte Corporation Molecules for disease detection and treatment

Patent Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5665863A (en) * 1992-01-31 1997-09-09 Rhone-Poulenc Rorer S.A. Polypeptides having granulocyte colony stimulating activity, their preparation and pharmaceutical compositions containing them
US20030082679A1 (en) * 2001-10-01 2003-05-01 Sun Lee-Hwei K. Fc fusion proteins of human granulocyte colony-stimulating factor with increased biological activities
WO2003081238A2 (en) * 2002-03-22 2003-10-02 Ludwig Maximilians Universität Cytocapacity test
WO2003093427A2 (en) * 2002-04-30 2003-11-13 Incyte Corporation Molecules for disease detection and treatment

Cited By (10)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US7790174B2 (en) * 2004-12-23 2010-09-07 Merck Serono Sa G-CSF polypeptides and uses thereof
US7863016B2 (en) 2004-12-23 2011-01-04 Merck Serono S.A. G-CSF polypeptides and uses thereof
US7863018B2 (en) 2004-12-23 2011-01-04 Merck Serono Sa G-CSF polypeptides and uses thereof
WO2011032204A1 (en) * 2009-09-15 2011-03-24 Csl Limited Treatment of neurological conditions
CN102630167A (en) * 2009-09-15 2012-08-08 Csl有限公司 Treatment of neurological conditions
AU2010295223B2 (en) * 2009-09-15 2015-05-07 Csl Limited Treatment of neurological conditions
US9352038B2 (en) 2009-09-15 2016-05-31 Csl Limited Treatment of neurological conditions
US9364534B2 (en) 2009-09-15 2016-06-14 Csl Limited Treatment of neurological conditions
KR101750426B1 (en) * 2009-09-15 2017-06-23 씨에스엘 리미티드 Treatment of neurological conditions
EP3757117A1 (en) * 2015-11-16 2020-12-30 Ubiprotein, Corp. A method for extending half-life of a protein

Also Published As

Publication number Publication date
WO2005121174A3 (en) 2006-05-04

Similar Documents

Publication Publication Date Title
US7776564B2 (en) Stromal cell-derived factor-1 polypeptides, polynucleotides, modulators thereof and methods of use
CA2574654C (en) Compositions and methods of use for mgd-csf in disease treatment
US20080242603A1 (en) Novel Apo2L and IL-24 Polypeptides, Polynucleotides, and Methods of Their Use
JP5191619B2 (en) Mammalian cytokine-like polypeptide-10
JP2013188214A (en) Mu-1, member of cytokine receptor family
JP2001524814A (en) 28 human secreted proteins
JP2002533058A (en) 97 human secreted proteins
JP2001508309A (en) Member of the hematopoietin receptor superfamily
WO2005105840A2 (en) Cd40 variants and uses thereof
WO2005121174A2 (en) Novel g-csf polypeptides, polynucleotides, modulators thereof, and methods of use
ES2352339T3 (en) DEFENSE PROTEINS.
JP4211966B2 (en) Compositions and methods for treating immune related diseases
JP2002504361A (en) 36 human secreted proteins
WO2001031031A1 (en) Novel polypeptide---human helper t cell growth factor 15 and polynucleotide encoding it
JP2002511268A (en) U4, a member of the hematopoietin receptor superfamily

Legal Events

Date Code Title Description
AK Designated states

Kind code of ref document: A2

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

AL Designated countries for regional patents

Kind code of ref document: A2

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

121 Ep: the epo has been informed by wipo that ep was designated in this application
NENP Non-entry into the national phase in:

Ref country code: DE

WWW Wipo information: withdrawn in national office

Country of ref document: DE

122 Ep: pct application non-entry in european phase