WO2001055163A1 - Acides nucleiques, proteines, et anticorps - Google Patents

Acides nucleiques, proteines, et anticorps Download PDF

Info

Publication number
WO2001055163A1
WO2001055163A1 PCT/US2001/001358 US0101358W WO0155163A1 WO 2001055163 A1 WO2001055163 A1 WO 2001055163A1 US 0101358 W US0101358 W US 0101358W WO 0155163 A1 WO0155163 A1 WO 0155163A1
Authority
WO
WIPO (PCT)
Prior art keywords
polypeptide
seq
sequence
polypeptides
polynucleotide
Prior art date
Application number
PCT/US2001/001358
Other languages
English (en)
Other versions
WO2001055163A8 (fr
Inventor
Craig A. Rosen
Steven C. Barash
Steven M. Ruben
Original Assignee
Human Genome Sciences, Inc.
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Human Genome Sciences, Inc. filed Critical Human Genome Sciences, Inc.
Priority to EP01918156A priority Critical patent/EP1254151A1/fr
Priority to AU2001245262A priority patent/AU2001245262A1/en
Publication of WO2001055163A1 publication Critical patent/WO2001055163A1/fr
Publication of WO2001055163A8 publication Critical patent/WO2001055163A8/fr

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N9/00Enzymes; Proenzymes; Compositions thereof; Processes for preparing, activating, inhibiting, separating or purifying enzymes
    • C12N9/14Hydrolases (3)
    • C12N9/48Hydrolases (3) acting on peptide bonds (3.4)
    • C12N9/50Proteinases, e.g. Endopeptidases (3.4.21-3.4.25)
    • C12N9/64Proteinases, e.g. Endopeptidases (3.4.21-3.4.25) derived from animal tissue
    • C12N9/6421Proteinases, e.g. Endopeptidases (3.4.21-3.4.25) derived from animal tissue from mammals
    • C12N9/6424Serine endopeptidases (3.4.21)
    • 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
    • C07K14/46Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates
    • C07K14/47Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates from mammals
    • 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
    • C07K14/705Receptors; Cell surface antigens; Cell surface determinants
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N9/00Enzymes; Proenzymes; Compositions thereof; Processes for preparing, activating, inhibiting, separating or purifying enzymes
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N9/00Enzymes; Proenzymes; Compositions thereof; Processes for preparing, activating, inhibiting, separating or purifying enzymes
    • C12N9/14Hydrolases (3)
    • C12N9/48Hydrolases (3) acting on peptide bonds (3.4)
    • C12N9/50Proteinases, e.g. Endopeptidases (3.4.21-3.4.25)
    • C12N9/64Proteinases, e.g. Endopeptidases (3.4.21-3.4.25) derived from animal tissue
    • C12N9/6421Proteinases, e.g. Endopeptidases (3.4.21-3.4.25) derived from animal tissue from mammals
    • C12N9/6489Metalloendopeptidases (3.4.24)
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12QMEASURING OR TESTING PROCESSES INVOLVING ENZYMES, NUCLEIC ACIDS OR MICROORGANISMS; COMPOSITIONS OR TEST PAPERS THEREFOR; PROCESSES OF PREPARING SUCH COMPOSITIONS; CONDITION-RESPONSIVE CONTROL IN MICROBIOLOGICAL OR ENZYMOLOGICAL PROCESSES
    • C12Q1/00Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions
    • C12Q1/68Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions involving nucleic acids
    • C12Q1/6876Nucleic acid products used in the analysis of nucleic acids, e.g. primers or probes
    • C12Q1/6883Nucleic acid products used in the analysis of nucleic acids, e.g. primers or probes for diseases caused by alterations of genetic material
    • AHUMAN NECESSITIES
    • A01AGRICULTURE; FORESTRY; ANIMAL HUSBANDRY; HUNTING; TRAPPING; FISHING
    • A01KANIMAL HUSBANDRY; AVICULTURE; APICULTURE; PISCICULTURE; FISHING; REARING OR BREEDING ANIMALS, NOT OTHERWISE PROVIDED FOR; NEW BREEDS OF ANIMALS
    • A01K2217/00Genetically modified animals
    • A01K2217/05Animals comprising random inserted nucleic acids (transgenic)
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/505Medicinal preparations containing antigens or antibodies comprising antibodies
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K48/00Medicinal preparations containing genetic material which is inserted into cells of the living body to treat genetic diseases; Gene therapy
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2319/00Fusion polypeptide
    • C07K2319/30Non-immunoglobulin-derived peptide or protein having an immunoglobulin constant or Fc region, or a fragment thereof, attached thereto
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N2500/00Screening for compounds of potential therapeutic value
    • YGENERAL TAGGING OF NEW TECHNOLOGICAL DEVELOPMENTS; GENERAL TAGGING OF CROSS-SECTIONAL TECHNOLOGIES SPANNING OVER SEVERAL SECTIONS OF THE IPC; TECHNICAL SUBJECTS COVERED BY FORMER USPC CROSS-REFERENCE ART COLLECTIONS [XRACs] AND DIGESTS
    • Y02TECHNOLOGIES OR APPLICATIONS FOR MITIGATION OR ADAPTATION AGAINST CLIMATE CHANGE
    • Y02ATECHNOLOGIES FOR ADAPTATION TO CLIMATE CHANGE
    • Y02A50/00TECHNOLOGIES FOR ADAPTATION TO CLIMATE CHANGE in human health protection, e.g. against extreme weather
    • Y02A50/30Against vector-borne diseases, e.g. mosquito-borne, fly-borne, tick-borne or waterborne diseases whose impact is exacerbated by climate change

Definitions

  • the present invention relates to novel proteins. More specifically, isolated nucleic acid molecules are provided encoding novel polypeptides. Novel polypeptides and antibodies that bind to these polypeptides are provided. Also provided are vectors, host cells, and recombinant and synthetic methods for producing human polynucleotides and/or polypeptides, and antibodies. The invention further relates to diagnostic and therapeutic methods useful for diagnosing, treating, preventing and/or prognosing disorders related to these novel polypeptides. The invention further relates to screening methods for identifying agonists and antagonists of polynucleotides and polypeptides of the invention. The present invention further relates to methods and/or compositions for inhibiting or enhancing the production and function of the polypeptides of the present invention.
  • Neoplastic disease is actually a diverse group of diseases sharing one characteristic in common; all neoplastic diseases (cancers) result from the uncontrolled proliferation of cells.
  • the human body is composed of many different cell types, e.g. liver cells, muscle cells, brain cells, etc. Normally, these cells grow and divide to produce more cells only as the body needs them (e.g. to regenerate blood cells or replace epithelial cells lining the stomach). Sometimes, however, cells begin to divide unchecked even though new cells are not needed. These extra cells accumulate and form a mass of tissue, called a tumor.
  • a tumor a mass of tissue
  • Tumors are either benign or malignant. Benign tumors are not cancerous; they can usually be removed, they do not spread to other parts of the body and, they rarely threaten life. Malignant tumors, however, are cancerous. Cells in malignant tumors can invade and damage nearby or distant tissues and organs. The spread of cancerous cells is called metastasis. Malignant (or metastatic) cells can invade adjacent organs by proliferating directly from the primary tumor. Additionally, malignant cells can also metastasize to distant organs by breaking away from the primary tumor, entering the bloodstream or lymphatic system, and settling down in a new organ or tissue to produce a secondary tumor. The origin of secondary tumors is established by comparing cells comprising these tumors to cells in the original (primary) tumor.
  • leukemia In contrast to solid organ cancers (such as cancer in the liver, lung, and brain) cancer can also develop in blood-forming cells. These cancers are referred to as leukemias or lymphomas.
  • Leukemia refers to cancer of blood forming cells such as red blood cells, platelets, and plasma cells.
  • Lymphomas are a subset of leukemias, primarily involving white blood cells, in which the cancerous cells originated in, or are associated with, the lymph system and lymph organs (e.g. T-lymphocytes in the lymph nodes, spleen, or thymus).
  • lymph system e.g. T-lymphocytes in the lymph nodes, spleen, or thymus.
  • the present invention relates to novel proteins. More specifically, isolated nucleic acid molecules are provided encoding novel polypeptides. Novel polypeptides and antibodies that bind to these polypeptides are provided. Also provided are vectors, host cells, and recombinant and synthetic methods for producing human polynucleotides and/or polypeptides, and antibodies. The invention further relates to diagnostic and therapeutic methods useful for diagnosing, treating, preventing and/or prognosing disorders related to these novel polypeptides. The invention further relates to screening methods for identifying agonists and antagonists of polynucleotides and polypeptides of the invention. The present invention further relates to methods and/or compositions for inhibiting or enhancing the production and function of the polypeptides of the present invention.
  • the first column provides the gene number in the application for each clone identifier.
  • the second column provides a unique clone identifier, "Clone ID NO:Z", for a cDNA clone related to each contig sequence disclosed in Table 1A.
  • the third column provides a unique contig identifier, "Contig ID:” for each of the contig sequences disclosed in Table 1 A.
  • the fourth column provides the sequence identifier, "SEQ ED NO:X", for each of the contig sequences disclosed in Table 1A.
  • the fifth column “ORF (From-To)" provides the location (i.e., nucleotide position numbers) within the polynucleotide sequence of SEQ ID NO:X that delineate the preferred open reading frame (ORF) that encodes the amino acid sequence shown in the sequence listing and referenced in Table 1A as SEQ ID NO:Y (column 6).
  • Column 7 lists residues comprising predicted epitopes contained in the polypeptides encoded by each of the preferred ORFs (SEQ ID NO:Y).
  • polypeptides of the invention comprise, or alternatively consist of, one, two, three, four, five or more of the predicted epitopes described in Table 1A.
  • Tissue Distribution shows the expression profile of tissue, cells, and/or cell line libraries which express the polynucleotides of the invention.
  • the first number in column 8 represents the tissue/cell source identifier code corresponding to the key provided in Table 4. Expression of these polynucleotides was not observed in the other tissues and/or cell libraries tested.
  • the second number in column 8 represents the number of times a sequence corresponding to the reference polynucleotide sequence (e.g., SEQ ID NO:X) was identified in the tissue/cell source.
  • tissue/cell source identifier codes in which the first two letters are "AR” designate information generated using DNA array technology. Utilizing this technology, cDNAs were amplified by PCR and then transferred, in duplicate, onto the array. Gene expression was assayed through hybridization of first strand cDNA probes to the DNA array. cDNA probes were generated from total RNA extracted from a variety of different tissues and cell lines.
  • Probe synthesis was performed in the presence of 3 P dCTP, using oligo(dT) to prime reverse transcription. After hybridization, high stringency washing conditions were employed to remove nonspecific hybrids from the array. The remaining signal, emanating from each gene target, was measured using a Phosphorimager. Gene expression was reported as Phosphor Stimulating Luminescence (PSL) which reflects the level of phosphor signal generated from the probe hybridized to each of the gene targets represented on the array. A local background signal subtraction was performed before the total signal generated from each array was used to normalize gene expression between the different hybridizations. The value presented after "[array code]:" represents the mean of the duplicate values, following background subtraction and probe normalization.
  • PSL Phosphor Stimulating Luminescence
  • the first column provides a unique clone identifier, "Clone ID NO:Z”, for a cDNA clone related to each contig sequence.
  • the second column provides the sequence identifier, "SEQ ID NO:X”, for each contig sequence.
  • the third column provides a unique contig identifier, "Contig ID:” for each contig sequence.
  • the fourth column provides a BAC identifier "BAC ID NO:A” for the BAC clone referenced in the corresponding row of the table.
  • the fifth column provides the nucleotide sequence identifier, "SEQ ID NO:B" for a fragment of the BAC clone identified in column four of the corresponding row of the table.
  • the sixth column provides the location (i.e., nucleotide position numbers) within the polynucleotide sequence of SEQ ID NO:B which delineate certain polynucleotides of the invention that are also exemplary members of polynucleotide sequences that encode polypeptides of the invention (e.g., polypeptides containing amino acid sequences encoded by the polynucleotide sequences delineated in column six, and fragments and variants thereof).
  • Table 2 summarizes homology and features of some of the polypeptides of the invention.
  • the first column provides a unique clone identifier, "Clone ID NO:Z”, corresponding to a cDNA clone disclosed in Table 1A.
  • the second column provides the unique contig identifier, "Contig ID:” corresponding to contigs in Table 1 A and allowing for correlation with the information in Table 1A.
  • the third column provides the sequence identifier, "SEQ ID NO:X”, for the contig polynucleotide sequence.
  • the fourth column provides the analysis method by which the homology/identity disclosed in the Table was determined.
  • NR non-redundant protein database
  • PFAM protein families
  • polypeptides of the invention comprise, or alternatively consist of, an amino acid sequence encoded by a polynucleotide in SEQ ID NO:X as delineated in columns 8 and 9, or fragments or variants thereof.
  • Table 3 provides polynucleotide sequences that may be disclaimed according to certain embodiments of the invention.
  • the first column provides a unique clone identifier, "Clone ID”, for a cDNA clone related to contig sequences disclosed in Table 1A.
  • the second column provides the sequence identifier, "SEQ ID NO:X”, for contig sequences disclosed in Table 1 A.
  • the third column provides the unique contig identifier, "Contig ID:”, for contigs disclosed in Table 1 A.
  • the fourth column provides a unique integer 'a' where 'a' is any integer between 1 and the final nucleotide minus 15 of SEQ ID NO:X
  • the fifth column provides a unique integer 'b' where 'b' is any integer between 15 and the final nucleotide of SEQ ID NO:X, where both a and b correspond to the positions of nucleotide residues shown in SEQ ID NO:X, and where b is greater than or equal to a + 14.
  • the uniquely defined integers can be substituted into the general formula of a-b, and used to describe polynucleotides which may be preferably excluded from the invention.
  • preferably excluded from the invention are at least one, two, three, four, five, ten, or more of the polynucleotide sequence(s) having the accession number(s) disclosed in the sixth column of this Table (including for example, published sequence in connection with a particular BAC clone).
  • preferably excluded from the invention are the specific polynucleotide sequence(s) contained in the clones co ⁇ esponding to at least one, two, three, four, five, ten, or more of the available material having the accession numbers identified in the sixth column of this Table (including for example, the actual sequence contained in an identified BAC clone).
  • Table 4 provides a key to the tissue/cell source identifier code disclosed in Table
  • tissue/cell source identifier code disclosed in Table 1A, Column 8.
  • Columns 2-5 provide a description of the tissue or cell source. Codes co ⁇ esponding to diseased tissues are indicated in column 6 with the word "disease". The use of the word "disease" in column 6 is non-limiting.
  • the tissue or cell source may be specific (e.g. a neoplasm), or may be disease-associated (e.g., a tissue sample from a normal portion of a diseased organ).
  • tissues and/or cells lacking the "disease" designation may still be derived from sources directly or indirectly involved in a disease state or disorder, and therefore may have a further utility in that disease state or disorder.
  • Table 1A column 10.
  • OMIM reference identification numbers (Column 1) were derived from Online Mendelian Inheritance in Man (Online Mendelian Inheritance in Man, OMIM. McKusick-Nathans Institute for Genetic Medicine, Johns Hopkins University (Baltimore, MD) and National Center for Biotechnology Information, National Library of Medicine, (Bethesda, MD) 2000. World Wide Web URL: http://www.ncbi.nlm.nih.gov/omim/).
  • Column 2 provides diseases associated with the cytologic band disclosed in Table 1A, column 9, as determined using the Morbid Map database.
  • Table 6 summarizes ATCC Deposits, Deposit dates, and ATCC designation numbers of deposits made with the ATCC in connection with the present application.
  • Table 7 shows the cDNA libraries sequenced, and ATCC designation numbers and vector information relating to these cDNA libraries.
  • Table 8 provides a physical characterization of clones encompassed by the invention.
  • the first column provides the unique clone identifier, "Clone ID NO:Z", for certain cDNA clones of the invention, as described in Table 1A.
  • the second column provides the size of the cDNA insert contained in the co ⁇ esponding cDNA clone. Definitions
  • isolated refers to material removed from its original environment (e.g., the natural environment if it is naturally occurring), and thus is altered “by the hand of man” from its natural state.
  • an isolated polynucleotide could be part of a vector or a composition of matter, or could be contained within a cell, and still be “isolated” because that vector, composition of matter, or particular cell is not the original environment of the polynucleotide.
  • isolated does not refer to genomic or cDNA libraries, whole cell total or mRNA preparations, genomic DNA preparations (including those separated by electrophoresis and transfe ⁇ ed onto blots), sheared whole cell genomic DNA preparations or other compositions where the art demonstrates no distinguishing features of the polynucleotide/sequences of the present invention.
  • a "polynucleotide” refers to a molecule having a nucleic acid sequence encoding SEQ ID NO.Y or a fragment or variant thereof; a nucleic acid sequence contained in SEQ ID NO:X (as described in column 3 of Table 1A) or the complement thereof; a cDNA sequence contained in Clone ID NO:Z (as described in column 2 of Table 1 A and contained within a library deposited with the ATCC); a nucleotide sequence encoding the polypeptide encoded by a nucleotide sequence in SEQ D NO:B as defined in column 6 of Table IB or a fragment or variant thereof; or a nucleotide coding sequence in SEQ ID NO:B as defined in column 6 of Table IB or the complement thereof.
  • the polynucleotide can contain the nucleotide sequence of the full length cDNA sequence, including the 5' and 3' untranslated sequences, the coding region, as well as fragments, epitopes, domains, and variants of the nucleic acid sequence.
  • a "polypeptide” refers to a molecule having an amino acid sequence encoded by a polynucleotide of the invention as broadly defined (obviously excluding poly-Phenylalanine or poly-Lysine peptide sequences which result from translation of a polyA tail of a sequence co ⁇ esponding to a cDNA).
  • SEQ ID NO:X was often generated by overlapping sequences contained in multiple clones (contig analysis).
  • a representative clone containing all or most of the sequence for SEQ ID NO:X is deposited at Human Genome Sciences, Inc. (HGS) in a catalogued and archived library.
  • HGS Human Genome Sciences, Inc.
  • each clone is identified by a cDNA Clone ID (identifier generally refe ⁇ ed to herein as Clone ID NO:Z).
  • Clone ID NO:Z identifier generally refe ⁇ ed to herein as Clone ID NO:Z.
  • Each Clone ID is unique to an individual clone and the Clone ID is all the information needed to retrieve a given clone from the HGS library.
  • ATCC American Type Culture Collection
  • Library names contain four characters, for example, "HTWE.”
  • the name of a cDNA clone (Clone ID) isolated from that library begins with the same four characters, for example "HTWEP07".
  • Table 1A co ⁇ elates the Clone ID names with SEQ ED NO:X.
  • SEQ ID NO:X the co ⁇ esponding Clone ED, which library it came from and which ATCC deposit the library is contained in.
  • the ATCC is located at 10801 University Boulevard, Manassas, Virginia 20110-2209, USA.
  • the ATCC deposits were made pursuant to the terms of the Budapest Treaty on the international recognition of the deposit of microorganisms for the purposes of patent procedure.
  • the polynucleotides of the invention are at least 15, at least 30, at least 50, at least 100, at least 125, at least 500, or at least 1000 continuous nucleotides but are less than or equal to 300 kb, 200 kb, 100 kb, 50 kb, 15 kb, 10 kb, 7.5kb, 5 kb, 2.5 kb, 2.0 kb, or 1 kb, in length.
  • polynucleotides of the invention comprise a portion of the coding sequences, as disclosed herein, but do not comprise all or a portion of any intron.
  • the polynucleotides comprising coding sequences do not contain coding sequences of a genomic flanking gene (i.e., 5' or 3' to the gene of interest in the genome). In other embodiments, the polynucleotides of the invention do not contain the coding sequence of more than 1000, 500, 250, 100, 50, 25, 20, 15, 10, 5, 4, 3, 2, or 1 genomic flanking gene(s).
  • a "polynucleotide” of the present invention also includes those polynucleotides capable of hybridizing, under stringent hybridization conditions, to sequences contained in SEQ ED NO:X, or the complement thereof (e.g., the complement of any one, two, three, four, or more of the polynucleotide fragments described herein), the polynucleotide sequence delineated in columns 8 and 9 of Table 2 or the complement thereof, and/or cDNA sequences contained in Clone ED NO:Z (e.g., the complement of any one, two, three, four, or more of the polynucleotide fragments, or the cDNA clone within the pool of cDNA clones deposited with the ATCC, described herein), and/or the polynucleotide sequence delineated in column 6 of Table IB or the complement thereof.
  • “Stringent hybridization conditions” refers to an overnight incubation at 42 degree C in a solution comprising 50% formamide, 5x SSC (750 mM NaCl, 75 mM trisodium citrate), 50 mM sodium phosphate (pH 7.6), 5x Denhardt's solution, 10% dextran sulfate, and 20 ⁇ g/ml denatured, sheared salmon sperm DNA, followed by washing the filters in O.lx SSC at about 65 degree C.
  • nucleic acid molecules that hybridize to the polynucleotides of the present invention at lower stringency hybridization conditions. Changes in the stringency of hybridization and signal detection are primarily accomplished through the manipulation of formamide concentration (lower percentages of formamide result in lowered stringency); salt conditions, or temperature.
  • washes performed following stringent hybridization can be done at higher salt concentrations (e.g. 5X SSC).
  • blocking reagents include Denhardt's reagent, BLOTTO, heparin, denatured salmon sperm DNA, and commercially available proprietary formulations.
  • the inclusion of specific blocking reagents may require modification of the hybridization conditions described above, due to problems with compatibility.
  • polynucleotide which hybridizes only to polyA+ sequences (such as any 3' terminal polyA+ tract of a cDNA shown in the sequence listing), or to a complementary stretch of T (or U) residues, would not be included in the definition of "polynucleotide,” since such a polynucleotide would hybridize to any nucleic acid molecule containing a poly (A) stretch or the complement thereof (e.g., practically any double-stranded cDNA clone generated using oligo dT as a primer).
  • the polynucleotide of the present invention can be composed of any polyribonucleotide or polydeoxribonucleotide, which may be unmodified RNA or DNA or modified RNA or DNA.
  • polynucleotides can be composed of single- and double-stranded DNA, DNA that is a mixture of single- and double-stranded regions, single- and double-stranded RNA, and RNA that is mixture of single- and double-stranded regions, hybrid molecules comprising DNA and RNA that may be single-stranded or, more typically, double-stranded or a mixture of single- and double-stranded regions.
  • polynucleotide can be composed of triple-stranded regions comprising RNA or DNA or both RNA and DNA.
  • a polynucleotide may also contain one or more modified bases or DNA or RNA backbones modified for stability or for other reasons.
  • Modified bases include, for example, tritylated bases and unusual bases such as inosine.
  • a variety of modifications can be made to DNA and RNA; thus, "polynucleotide” embraces chemically, enzymatically, or metabolically modified forms.
  • the polypeptide of the present invention can be composed of amino acids joined to each other by peptide bonds or modified peptide bonds, i.e., peptide isosteres, and may contain amino acids other than the 20 gene-encoded amino acids.
  • the polypeptides may be modified by either natural processes, such as posttranslational processing, or by chemical modification techniques which are well known in the art. Such modifications are well described in basic texts and in more detailed monographs, as well as in a voluminous research literature. Modifications can occur anywhere in a polypeptide, including the peptide backbone, the amino acid side-chains and the amino or carboxyl termini.
  • polypeptides may be branched, for example, as a result of ubiquitination, and they may be cyclic, with or without branching. Cyclic, branched, and branched cyclic polypeptides may result from posttranslation natural processes or may be made by synthetic methods.
  • Modifications include acetylation, acylation, ADP-ribosylation, amidation, covalent attachment of flavin, covalent attachment of a heme moiety, covalent attachment of a nucleotide or nucleotide derivative, covalent attachment of a lipid or lipid derivative, covalent attachment of phosphotidylinositol, cross-linking, cyclization, disulfide bond formation, demethylation, formation of covalent cross-links, formation of cysteine, formation of pyroglutamate, formylation, gamma-carboxylation, glycosylation, GPI anchor formation, hydroxylation, iodination, methylation, myristoylation, oxidation, pegylation, proteolytic processing, phosphorylation, prenylation, racemization, selenoylation, sulfation, transfer- RNA mediated addition of amino acids to proteins such as arginylation, and ubiquitination.
  • SEQ ID NO:X refers to a polynucleotide sequence described, for example, in
  • SEQ ID NO:Y refers to a polypeptide sequence described in column 6 of Table 1A.
  • SEQ ID NO:X is identified by an integer specified in column 4 of Table 1A.
  • the polypeptide sequence SEQ ID NO:Y is a translated open reading frame (ORF) encoded by polynucleotide SEQ ID NO:X.
  • Clone ED NO:Z refers to a cDNA clone described in column 2 of Table 1 A.
  • a polypeptide having functional activity refers to a polypeptide capable of displaying one or more known functional activities associated with a full-length (complete) protein. Such functional activities include, but are not limited to, biological activity, antigenicity [ability to bind (or compete with a polypeptide for binding) to an anti- polypeptide antibody], immunogenicity (ability to generate antibody which binds to a specific polypeptide of the invention), ability to form multimers with polypeptides of the invention, and ability to bind to a receptor or ligand for a polypeptide.
  • polypeptides of the invention can be assayed for functional activity (e.g. biological activity) using or routinely modifying assays known in the art, as well as assays described herein. Specifically, one of skill in the art may routinely assay neoplastic disease associated polypeptides (including fragments and variants) of the invention for activity using assays as described in Examples 21-62 and 65.
  • a polypeptide having biological activity refers to a polypeptide exhibiting activity similar to, but not necessarily identical to, an activity of a polypeptide of the present invention, including mature forms, as measured in a particular biological assay, with or without dose dependency. In the case where dose dependency does exist, it need not be identical to that of the polypeptide, but rather substantially similar to the dose-dependence in a given activity as compared to the polypeptide of the present invention (i.e., the candidate polypeptide will exhibit greater activity or not more than about 25-fold less and, preferably, not more than about tenfold less activity, and most preferably, not more than about three-fold less activity relative to the polypeptide of the present invention).
  • the first column in Table 1A provides the gene number in the application responding to the clone identifier.
  • the second column in Table 1A provides a unique "Clone ED NO:Z" for a cDNA clone related to each contig sequence disclosed in Table 1A.
  • This clone ID references the cDNA clone which contains at least the 5' most sequence of the assembled contig and at least a portion of SEQ ID NO:X was determined by directly sequencing the referenced clone.
  • the reference clone may have more sequence than described in the sequence listing or the clone may have less. In the vast majority of cases, however, the clone is believed to encode a full-length polypeptide. In the case where a clone is not full-length, a full-length cDNA can be obtained by methods described elsewhere herein.
  • the third column in Table 1A provides a unique "Contig ID” identification for each contig sequence.
  • the fourth column provides the "SEQ ID NO:” identifier for each of the contig polynucleotide sequences disclosed in Table 1A.
  • the fifth column, "ORF (From- To)" provides the location (i.e., nucleotide position numbers) within the polynucleotide sequence "SEQ ID NO:X” that delineate the preferred open reading frame (ORF) shown in the sequence listing and referenced in Table 1A, column 6, as SEQ ID NO:Y.
  • the preferred ORF is the reverse complement of the referenced polynucleotide sequence.
  • the sixth column in Table 1A provides the corresponding SEQ ID NO:Y for the polypeptide sequence encoded by the preferred ORF delineated in column 5.
  • the invention provides an amino acid sequence comprising, or alternatively consisting of, a polypeptide encoded by the portion of SEQ ID NO:X delineated by "ORF (From-To)". Also provided are polynucleotides encoding such amino acid sequences and the complementary strand thereto.
  • polypeptides of the invention comprise, or alternatively consist of, at least one, two, three, four, five or more of the predicted epitopes as described in Table 1A.
  • Column 8 in Table 1A provides an expression profile and library code: count for h of the contig sequences (SEQ ID NO:X) disclosed in Table 1A, which can routinely be combined with the information provided in Table 4 and used to determine the tissues, cells, and/or cell line libraries which predominantly express the polynucleotides of the invention.
  • the first number in column 8 represents the tissue/cell source identifier code corresponding to the code and description provided in Table 4.
  • the second number in column 8 represents the number of times a sequence corresponding to the reference polynucleotide sequence was identified in the tissue/cell source.
  • tissue/cell source identifier codes in which the first two letters are "AR” designate information generated using DNA array technology. Utilizing this technology, cDNAs were amplified by PCR and then transferred, in duplicate, onto the array. Gene expression was assayed through hybridization of first strand cDNA probes to the DNA array. cDNA probes were generated from total RNA extracted from a variety of different tissues and cell lines. Probe synthesis was performed in the presence of 33 P dCTP, using oligo(dT) to prime reverse transcription.
  • Table IB summarizes additional polynucleotides encompassed by the invention eluding cDNA clones related to the sequences (Clone ID NO:Z), contig sequences (contig identifier (Contig ID:) contig nucleotide sequence identifiers (SEQ ID NO:X)), and genomic sequences (SEQ ID NO:B).
  • the first column provides a unique clone identifier, "Clone ID NO:Z”, for a cDNA clone related to each contig sequence.
  • the second column provides the sequence identifier, "SEQ ID NO:X”, for each contig sequence.
  • the third column provides a unique contig identifier, "Contig ID:” for each contig sequence.
  • the fourth column provides a BAC identifier "BAC ED NO:A” for the BAC clone referenced in the corresponding row of the table.
  • the fifth column provides the nucleotide sequence identifier, "SEQ ID NO:B" for a fragment of the BAC clone identified in column four of the corresponding row of the table.
  • the sixth column provides the location (i.e., nucleotide position numbers) within the polynucleotide sequence of SEQ ID NO:B which delineate certain polynucleotides of the invention that are also exemplary members of polynucleotide sequences that encode polypeptides of the invention (e.g., polypeptides containing amino acid sequences encoded by the polynucleotide sequences delineated in column six, and fragments and variants thereof).
  • Table 2 further characterizes certain encoded polypeptides of the invention, by providing the results of comparisons to protein and protein family databases.
  • the first column provides a unique clone identifier, "Clone ID NO:”, corresponding to a cDNA clone disclosed in Table 1A.
  • the second column provides the unique contig identifier, "Contig ID:” which allows correlation with the information in Table 1A.
  • the third column provides the sequence identifier, "SEQ ID NO:”, for the contig polynucleotide sequences.
  • the fourth column provides the analysis method by which the homology/identity disclosed in the Table was determined.
  • the fifth column provides a description of the PFAM/NR hit identified by each analysis.
  • the NR database which comprises the NBRF PIR database, the NCBI GenPept database, and the SIB SwissProt and TrEMBL databases, was made non-redundant using the computer program nrdb2 (Warren Gish, Washington University in Saint Louis).
  • nrdb2 Warren Gish, Washington University in Saint Louis.
  • Each of the polynucleotides shown in Table 1A, column 3 e.g., SEQ ID NO:X or the 'Query' sequence
  • the computer program BLASTX was used to compare a 6-frame translation of the Query sequence to the NR database (for information about the BLASTX algorithm please see Altshul et al., J. Mol. Biol. 215:403-410 (1990); and Gish and States, Nat. Genet.
  • the percent identity is determined by dividing the number of exact matches between the two aligned sequences in the HSP, dividing by the number of Query amino acids in the HSP and multiplying by 100.
  • the polynucleotides of SEQ ID NO:X which encode the polypeptide sequence that generates an HSP are delineated by columns 8 and 9 of Table 2.
  • HMM Hidden Markov Model
  • a HMM derived from PFAM version 2.1 was said to be a significant match to a polypeptide of the invention if the score returned by HMMER 1.8 was greater than 0.8 times the HMMER 1.8 score obtained with the most distantly related known member of that protein family.
  • the description of the PFAM family which shares . a significant match with a polypeptide of the invention is listed in column 5 of Table 2, and the database accession number of the PFAM hit is provided in column 6.
  • Column 7 provides the score returned by HMMER version 1.8 for the alignment.
  • Columns 8 and 9 delineate the polynucleotides of SEQ ID NO:X which encode the polypeptide sequence which show a significant match to a PFAM protein family.
  • the invention provides a protein comprising, or alternatively consisting of, a polypeptide encoded by the polynucleotides of SEQ ID NO:X delineated in columns 8 and 9 of Table 2. Also provided are polynucleotides encoding such proteins, and the complementary strand thereto.
  • nucleotide sequence SEQ ID NO:X and the translated SEQ ID NO:Y are sufficiently accurate and otherwise suitable for a variety of uses well known in the art and described further below.
  • the nucleotide sequences of SEQ ID NO:X are useful for designing nucleic acid hybridization probes that will detect nucleic acid sequences contained in SEQ ID NO:X or the cDNA contained in Clone ID NO:Z. These probes will also hybridize to nucleic acid molecules in biological samples, thereby enabling immediate applications in chromosome mapping, linkage analysis, tissue identification and/or typing, and a variety of forensic and diagnostic methods of the invention.
  • polypeptides identified from SEQ ID NO:Y may be used to generate antibodies which bind specifically to these polypeptides, or fragments thereof, and/or to the polypeptides encoded by the cDNA clones identified in, for example, Table 1A.
  • DNA sequences generated by sequencing reactions can contain sequencing errors.
  • the errors exist as misidentified nucleotides, or as insertions or deletions of nucleotides in the generated DNA sequence.
  • the erroneously inserted or deleted nucleotides cause frame shifts in the reading frames of the predicted amino acid sequence.
  • the predicted amino acid sequence diverges from the actual amino acid sequence, even though the generated DNA sequence may be greater than 99.9% identical to the actual DNA sequence (for example, one base insertion or deletion in an open reading frame of over 1000 bases).
  • the present invention provides not only the generated nucleotide sequence identified as SEQ ID NO:X, and a predicted translated amino acid sequence identified as SEQ ID NO:Y, but also a sample of plasmid DNA containing cDNA Clone ID NO:Z (deposited with the ATCC on October 5, 2000, and receiving ATCC designation numbers PTA 2574 and PTA 2575; deposited with the ATCC on January 5, 2001, and having depositor reference numbers TS-1, TS-2, AC-1, and AC-2; and/or as set forth, for example, in Table 1A, 6 and 7).
  • nucleotide sequence of each deposited clone can readily be determined by sequencing the deposited clone in accordance with known methods. Further, techniques known in the art can be used to verify the nucleotide sequences of SEQ ID NO:X. [54] The predicted amino acid sequence can then be verified from such deposits.
  • amino acid sequence of the protein encoded by a particular clone can also be directly determined by peptide sequencing or by expressing the protein in a suitable host cell containing the deposited human cDNA, collecting the protein, and determining its sequence.
  • Partial cDNA clones can be made full-length by utilizing the rapid amplification of cDNA ends (RACE) procedure described in Frohman, M.A., et al., Proc. Nat'l. Acad. Sci. USA, 85:8998-9002 (1988).
  • RACE rapid amplification of cDNA ends
  • RNA Poly A+ or total RNA is reverse transcribed with Superscript II (Gibco/BRL) and an antisense or complementary primer specific to the cDNA sequence.
  • the primer is removed from the reaction with a Microcon Concentrator (Amicon).
  • the first- strand cDNA is then tailed with dATP and terminal deoxynucleotide transferase (Gibco/BRL).
  • the second strand is synthesized from the dA-tail in PCR buffer, Taq DNA polymerase (Perkin-Elmer Cetus), an oligo-dT primer containing three adjacent restriction sites (Xhol, Sail and Clal) at the 5' end and a primer containing just these restriction sites.
  • This double-stranded cDNA is PCR amplified for 40 cycles with the same primers as well as a nested cDNA-specific antisense primer.
  • the PCR products are size-separated on an ethidium bromide-agarose gel and the region of gel containing cDNA products the predicted size of missing protein-coding DNA is removed.
  • cDNA is purified from the agarose with the Magic PCR Prep kit (Promega), restriction digested with Xhol or Sail, and ligated to a plasmid such as pBluescript SKU (Stratagene) at Xhol and EcoRV sites.
  • This DNA is transformed into bacteria and the plasmid clones sequenced to identify the correct protein-coding inserts. Correct 5' ends are confirmed by comparing this sequence with the putatively identified homologue and overlap with the partial cDNA clone. Similar methods known in the art and/or commercial kits are used to amplify and recover 3' ends.
  • kits are commercially available for purchase. Similar reagents and methods to those above are supplied in kit form from Gibco/BRL for both 5' and 3' RACE for recovery of full length genes. A second kit is available from Clontech which is a modification of a related technique, SLIC (single-stranded ligation to single-stranded cDNA), developed by Dumas et al., Nucleic Acids Res., 19:5227-32 (1991). The major differences in procedure are that the RNA is alkaline hydrolyzed after reverse transcription and RNA ligase is used to join a restriction site-containing anchor primer to the first-strand cDNA. This obviates the necessity for the dA-tailing reaction which results in a polyT stretch that is difficult to sequence past.
  • SLIC single-stranded ligation to single-stranded cDNA
  • An alternative to generating 5' or 3' cDNA from RNA is to use cDNA library double-stranded DNA.
  • An asymmetric PCR-amplified antisense cDNA strand is synthesized with an antisense cDNA-specific primer and a plasmid-anchored primer. These primers are removed and a symmetric PCR reaction is performed with a nested cDNA-specific antisense primer and the plasmid-anchored primer.
  • RNA oligonucleotide is ligated to the 5' ends of a population of RNA presumably containing full-length gene RNA transcript and a primer set containing a primer specific to the ligated RNA oligonucleotide and a primer specific to a known sequence of the gene of interest, is used to PCR amplify the 5' portion of the desired full length gene which may then be sequenced and used to generate the full length gene.
  • This method starts with total RNA isolated from the desired source, poly A RNA may be used but is not a prerequisite for this procedure.
  • RNA preparation may then be treated with phosphatase if necessary to eliminate 5' phosphate groups on degraded or damaged RNA which may interfere with the later RNA ligase step.
  • the phosphatase if used is then inactivated and the RNA is treated with tobacco acid pyrophosp hatase in order to remove the cap structure present at the 5' ends of messenger RNAs.
  • This reaction leaves a 5' phosphate group at the 5' end of the cap cleaved RNA which can then be ligated to an RNA oligonucleotide using T4 RNA ligase.
  • This modified RNA preparation can then be used as a template for first strand cDNA synthesis using a gene specific oligonucleotide.
  • the first strand synthesis reaction can then be used as a template for PCR amplification of the desired 5' end using a primer specific to the ligated RNA oligonucleotide and a primer specific to the known sequence of the gene of interest.
  • the resultant product is then sequenced and analyzed to confirm that the 5' end sequence belongs to the relevant gene.
  • the present invention also relates to vectors or plasmids which include such DNA sequences, as well as the use of the DNA sequences.
  • the material deposited with the ATCC (deposited with the ATCC on October 5, 2000, and receiving ATCC designation numbers PTA 2574 and PTA 2575; deposited with the ATCC on January 5, 2001, and receiving ATCC designation numbers TS-1, TS-2, AC-1, and AC-2; and/or as set forth, for example, in Table 1A, Table 6, or Table 7) is a mixture of cDNA clones derived from a variety of human tissue and cloned in either a plasmid vector or a phage vector, as described, for example, in Table 7. These deposits are referred to as "the deposits" herein.
  • the tissues from which some of the clones were derived are listed in Table 7, and the vector in which the corresponding cDNA is contained is also indicated in Table 7.
  • the deposited material includes cDNA clones corresponding to SEQ ID NO:X described, for example, in Table 1A (Clone ID NO:Z).
  • a clone which is isolatable from the ATCC Deposits by use of a sequence listed as SEQ ID NO:X may include the entire coding region of a human gene or in other cases such clone may include a substantial portion of the coding region of a human gene.
  • sequence listing may in some instances list only a portion of the DNA sequence in a clone included in the ATCC Deposits, it is well within the ability of one skilled in the art to sequence the DNA included in a clone contained in the ATCC Deposits by use of a sequence (or portion thereof) described in, for example Tables lAor 2 by procedures hereinafter further described, and others apparent to those skilled in the art.
  • Table 7 Also provided in Table 7 is the name of the vector which contains the cDNA clone. Each vector is routinely used in the art. The following additional information is provided for convenience.
  • Phagemid pBS contains an ampicillin resistance gene and pBK contains a neomycin resistance gene.
  • Phagemid pBS may be excised from the Lambda Zap and Uni-Zap XR vectors, and phagemid pBK may be excised from the Zap Express vector. Both phagemids may be transformed into E. coli strain XL-1 Blue, also available from Stratagene.
  • Vectors pSportl, pCMVSport 1.0, pCMVSport 2.0 and pCMVSport 3.0 were obtained from Life Technologies, Inc., P. O. Box 6009, Gaithersburg, MD 20897. All Sport vectors contain an ampicillin resistance gene and may be transformed into E. coli strain DH10B, also available from Life Technologies. See, for instance, Gruber, C. E., et al., Focus 75:59- (1993). Vector lafmid BA (Bento Soares, Columbia University, New York, NY) contains an ampicillin resistance gene and can be transformed into E. coli strain XL-1 Blue.
  • Vector pCR 2.1 which is available from Invitrogen, 1600 Faraday Avenue, Carlsbad, CA 92008, contains an ampicillin resistance gene and may be transformed into E. coli strain DH10B, available from Life Technologies. See, for instance, Clark, J. M., Nuc. Acids Res. 16:9611-9686 (1988) and Mead, D. et al., Bio/Technology 9: (1991).
  • the present invention also relates to the genes corresponding to S ⁇ Q ID NO:X,
  • S ⁇ Q ID NO:Y and/or the deposited clone (Clone ID NO:Z).
  • the corresponding gene can be isolated in accordance with known methods using the sequence information disclosed herein. Such methods include preparing probes or primers from the disclosed sequence and identifying or amplifying the corresponding gene from appropriate sources of genomic material.
  • allelic variants, orthologs, and/or species homologs are also provided in the present invention. Procedures known in the art can be used to obtain full-length genes, allelic variants, splice variants, full-length coding portions, orthologs, and/or species homologs of genes corresponding to S ⁇ Q ID NO:X or the complement thereof, polypeptides encoded by genes corresponding to S ⁇ Q ID NO:X or the complement thereof, and/or the cDNA contained in Clone ID NO:Z, using information from the sequences disclosed herein or the clones deposited with the ATCC.
  • allelic variants and/or species homologs may be isolated and identified by making suitable probes or primers from the sequences provided herein and screening a suitable nucleic acid source for allelic variants and/or the desired homologue.
  • polypeptides of the invention can be prepared in any suitable manner.
  • Such polypeptides include isolated naturally occurring polypeptides, recombinantly produced polypeptides, synthetically produced polypeptides, or polypeptides produced by a combination of these methods. Means for preparing such polypeptides are well understood in the art.
  • the polypeptides may be in the form of the secreted protein, including the mature form, or may be a part of a larger protein, such as a fusion protein (see below). It is often advantageous to include an additional amino acid sequence which contains secretory or leader sequences, pro-sequences, sequences which aid in purification, such as multiple histidine residues, or an additional sequence for stability during recombinant production.
  • the polypeptides of the present invention are preferably provided in an isolated form, and preferably are substantially purified.
  • a recombinantly produced version of a polypeptide, including the secreted polypeptide can be substantially purified using techniques described herein or otherwise known in the art, such as, for example, 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, synthetic or recombinant sources using techniques described herein or otherwise known in the art, such as, for example, antibodies of the invention raised against the polypeptides of the present invention in methods which are well known in the art.
  • the present invention provides a polynucleotide comprising, or alternatively consisting of, the nucleic acid sequence of SEQ ID NO:X, and/or the cDNA sequence contained in Clone ID NO:Z.
  • the present invention also provides a polypeptide comprising, or alternatively, consisting of, the polypeptide sequence of SEQ ID NO:Y, a polypeptide encoded by SEQ ID NO:X or a complement thereof, a polypeptide encoded by the cDNA contained in Clone ID NO:Z, and/or the polypeptide sequence encoded by a nucleotide sequence in SEQ ID NO:B as defined in column 6 of Table IB.
  • Polynucleotides encoding a polypeptide comprising, or alternatively consisting of the polypeptide sequence of SEQ ID NO:Y, a polypeptide encoded by SEQ ID NO:X, a polypeptide encoded by the cDNA contained in Clone ID NO:Z, and/or a polypeptide sequence encoded by a nucleotide sequence in SEQ ID NO:B as defined in column 6 of Table IB are also encompassed by the invention.
  • the present invention further encompasses a polynucleotide comprising, or alternatively consisting of, the complement of the nucleic acid sequence of SEQ ID NO:X, a nucleic acid sequence encoding a polypeptide encoded by the complement of the nucleic acid sequence of SEQ ID NO:X, and/or the cDNA contained in Clone ID NO:Z.
  • a polynucleotide comprising, or alternatively consisting of, the complement of the nucleic acid sequence of SEQ ID NO:X, a nucleic acid sequence encoding a polypeptide encoded by the complement of the nucleic acid sequence of SEQ ID NO:X, and/or the cDNA contained in Clone ID NO:Z.
  • representative examples of polynucleotides of the invention comprise, or alternatively consist of, one, two, three, four, five, six, seven, eight, nine, ten, or more of the sequences delineated in Table IB column 6, or any combination thereof.
  • polynucleotides of the invention comprise, or alternatively consist of, one, two, three, four, five, six, seven, eight, nine, ten, or more of the complementary strand(s) of the sequences delineated in Table IB column 6, or any combination thereof.
  • the above-described polynucleotides of the invention comprise, or alternatively consist of, sequences delineated in Table IB, column 6, and have a nucleic acid sequence which is different from that of the BAC fragment having the sequence disclosed in SEQ ID NO:B (see Table IB, column 5).
  • the above-described polynucleotides of the invention comprise, or alternatively consist of, sequences delineated in Table IB, column 6, and have a nucleic acid sequence which is different from that published for the BAC clone identified as BAC ID NO:A (see Table IB, column 4). In additional embodiments, the above-described polynucleotides of the invention comprise, or alternatively consist of, sequences delineated in Table IB, column 6, and have a nucleic acid sequence which is different from that contained in the BAC clone identified as BAC ED NO:A (see Table IB, column 4).
  • polypeptides encoded by these polynucleotides are also encompassed by the invention. Additionally, fragments and variants of the above- described polynucleotides and polypeptides are also encompassed by the invention.
  • representative examples of polynucleotides of the invention comprise, or alternatively consist of, one, two, three, four, five, six, seven, eight, nine, ten, or more of the sequences delineated in column 6 of Table IB which correspond to the same Clone ID NO:Z (see Table IB, column 1), or any combination thereof.
  • polynucleotides of the invention comprise, or alternatively consist of, one, two, three, four, five, six, seven, eight, nine, ten, or more of the complementary strand(s) of the sequences delineated in column 6 of Table IB which correspond to the same Clone ID NO:Z (see Table IB, column 1), or any combination thereof.
  • the above-described polynucleotides of the invention comprise, or alternatively consist of, sequences delineated in column 6 of Table IB which correspond to the same Clone ED NO:Z (see Table IB, column 1) and have a nucleic acid sequence which is different from that of the BAC fragment having the sequence disclosed in SEQ ID NO:B (see Table IB, column 5).
  • polynucleotides of the invention comprise, or alternatively consist of, sequences delineated in column 6 of Table IB which correspond to the same Clone ID NO:Z (see Table IB, column 1) and have a nucleic acid sequence which is different from that published for the BAC clone identified as BAC ID NO:A (see Table IB, column 4).
  • the above-described polynucleotides of the invention comprise, or alternatively consist of, sequences delineated in column 6 of Table IB which correspond to the same Clone ID NO:Z (see Table IB, column 1) and have a nucleic acid sequence which is different from that contained in the BAC clone identified as BAC ID NO: A (see Table IB, column 4).
  • Polypeptides encoded by these polynucleotides, other polynucleotides that encode these polypeptides, and antibodies that bind these polypeptides are also encompassed by the invention. Additionally, fragments and variants of the above- 'described polynucleotides and polypeptides are also encompassed by the invention.
  • polynucleotides of the invention comprise, or alternatively consist of, one, two, three, four, five, six, seven, eight, nine, ten, or more of the sequences delineated in column 6 of Table IB which correspond to the same contig sequence identifer SEQ ID NO:X (see Table IB, column 2), or any combination thereof.
  • polynucleotides of the invention comprise, or alternatively consist of, one, two, three, four, five, six, seven, eight, nine, ten, or more of the complementary strand(s) of the sequences delineated in column 6 of Table IB which correspond to the same contig sequence identifer SEQ ED NO:X (see Table IB, column 2), or any combination thereof.
  • the above-described polynucleotides of the invention comprise, or altematively consist of, sequences delineated in column 6 of Table IB which correspond to the same contig sequence identifer SEQ ID NO:X (see Table IB, column 2) and have a nucleic acid sequence which is different from that of the BAC fragment having the sequence disclosed in SEQ ED NO:B (see Table IB, column 5).
  • polynucleotides of the invention comprise, or alternatively consist of, sequences delineated in column 6 of Table IB which correspond to the same contig sequence identifer SEQ ED NO:X (see Table IB, column 2) and have a nucleic acid sequence which is different from that published for the BAC clone identified as BAC ED NO: A (see Table IB, column 4).
  • the above-described polynucleotides of the invention comprise, or alternatively consist of, sequences delineated in column 6 of Table IB which correspond to the same contig sequence identifer SEQ ED NO:X (see Table IB, column 2) and have a nucleic acid sequence which is different from that contained in the BAC clone identified as BAC ED NO: A (See Table IB, column 4).
  • Polypeptides encoded by these polynucleotides, other polynucleotides that encode these polypeptides, and antibodies that bind these polypeptides are also encompassed by the invention. Additionally, fragments and variants of the above-described polynucleotides and polypeptides are also encompassed by the invention.
  • representative examples of polynucleotides of the invention comprise, or alternatively consist of, one, two, three, four, five, six, seven, eight, nine, ten, or more of the sequences delineated in the same row of Table IB column 6, or any combination thereof.
  • Additional, representative examples of polynucleotides of the invention comprise, or alternatively consist of, one, two, three, four, five, six, seven, eight, nine, ten, or more of the complementary strand(s) of the sequences delineated in the same row of Table IB column 6, Or any combination thereof.
  • the polynucleotides of the invention comprise, or alternatively consist of, one, two, three, four, five, six, seven, eight, nine, ten, or more of the complementary strand(s) of the sequences delineated in the same row of Table IB column 6, wherein sequentially delineated sequences in the table (i.e.
  • above-described polynucleotides of the invention comprise, or alternatively consist of, sequences delineated in the same row of Table IB, column 6, and have a nucleic acid sequence which is different from that of the BAC fragment having the sequence disclosed in SEQ ED NO:B (see Table IB, column 5).
  • the above-described polynucleotides of the invention comprise, or alternatively consist of, sequences delineated in the same row of Table IB, column 6, and have a nucleic acid sequence which is different from that published for the BAC clone identified as BAC ID NO: A (see Table IB, column 4).
  • the above-described polynucleotides of the invention comprise, or alternatively consist of, sequences delineated in the same row of Table IB, column 6, and have a nucleic acid sequence which is different from that contained in the BAC clone identified as BAC ED NO:A (see Table IB, column 4).
  • Polypeptides encoded by these polynucleotides, other polynucleotides that encode these polypeptides, and antibodies that bind these polypeptides are also encompassed by the invention.
  • polynucleotides of the invention comprise, or alternatively consist of, one, two, three, four, five, six, seven, eight, nine, ten, or more of the sequences delineated in column 6 of Table IB, and the polynucleotide sequence of SEQ ID NO:X (e.g., as defined in Table IB, column 2) or fragments or variants thereof.
  • Polypeptides encoded by these polynucleotides, other polynucleotides that encode these polypeptides, and antibodies that bind these polypeptides are also encompassed by the invention.
  • polynucleotides of the invention comprise, or alternatively consist of, one, two, three, four, five, six, seven, eight, nine, ten, or more of the sequences delineated in column 6 of Table IB which correspond to the same Clone ID NO:Z (see Table IB, column 1), and the polynucleotide sequence of SEQ ID NO:X (e.g., as defined in Table 1A or IB) or fragments or variants thereof.
  • the delineated sequence(s) and polynucleotide sequence of SEQ ID NO:X correspond to the same Clone ID NO:Z.
  • Polypeptides encoded by these polynucleotides, other polynucleotides that encode these polypeptides, and antibodies that bind these polypeptides are also encompassed by the invention.
  • polynucleotides of the invention comprise, or altematively consist of, one, two, three, four, five, six, seven, eight, nine, ten, or more of the sequences delineated in the same row of column 6 of Table IB, and the polynucleotide sequence of SEQ ED NO:X (e.g., as defined in Table 1A or IB) or fragments or variants thereof.
  • the delineated sequence(s) and polynucleotide sequence of SEQ UD NO:X correspond to the same row of column 6 of Table IB.
  • polynucleotides of the invention comprise, or alternatively consist of a polynucleotide sequence in which the 3' 10 polynucleotides of one of the sequences delineated in column 6 of Table IB and the 5' 10 polynucleotides of the sequence of SEQ ED NO:X are directly contiguous.
  • Nucleic acids which hybridize to the complement of these 20 contiguous polynucleotides under stringent hybridization conditions or alternatively, under lower stringency conditions are also encompassed by the invention.
  • Polypeptides encoded by these polynucleotides and/or nucleic acids, other polynucleotides and/or nucleic acids that encode these polypeptides, and antibodies that bind these polypeptides are also encompassed by the invention. Additionally, fragments and variants of the above-described polynucleotides, nucleic acids, and polypeptides are also encompassed by the invention.
  • polynucleotides of the invention comprise, or alternatively consist of, a polynucleotide sequence in which the 3' 10 polynucleotides of one of the sequences delineated in column 6 of Table IB and the 5' 10 polynucleotides of a fragment or variant of the sequence of SEQ ID NO:X are directly contiguous Nucleic acids which hybridize to the complement of these 20 contiguous polynucleotides under stringent hybridization conditions or alternatively, under lower stringency conditions, are also encompassed by the invention.
  • Polypeptides encoded by these polynucleotides and/or nucleic acids, other polynucleotides and/or nucleic acids encoding these polypeptides, and antibodies that bind these polypeptides are also encompassed by the invention. Additionally, fragments and variants of the above-described polynucleotides, nucleic acids, and polypeptides are also encompassed by the invention.
  • polynucleotides of the invention comprise, or alternatively consist of, a polynucleotide sequence in which the 3' 10 polynucleotides of the sequence of SEQ ED NO:X and the 5' 10 polynucleotides of the sequence of one of the sequences delineated in column 6 of Table IB are directly contiguous. Nucleic acids which hybridize to the complement of these 20 contiguous polynucleotides under stringent hybridization conditions or alternatively, under lower stringency conditions, are also encompassed by the invention.
  • Polypeptides encoded by these polynucleotides and/or nucleic acids, other polynucleotides and/or nucleic acids encoding these polypeptides, and antibodies that bind these polypeptides are also encompassed by the invention. Additionally, fragments and variants of the above-described polynucleotides, nucleic acids, and polypeptides are also encompassed by the invention.
  • polynucleotides of the invention comprise, or alternatively consist of, a polynucleotide sequence in which the 3' 10 polynucleotides of a fragment or variant of the sequence of SEQ ID NO:X and the 5' 10 polynucleotides of the sequence of one of the sequences delineated in column 6 of Table IB are directly contiguous.
  • Nucleic acids which hybridize to the complement of these 20 contiguous polynucleotides under stringent hybridization conditions or alternatively, under lower stringency conditions, are also encompassed by the invention.
  • Polypeptides encoded by these polynucleotides and/or nucleic acids, other polynucleotides and/or nucleic acids encoding these polypeptides, and antibodies that bind these polypeptides are also encompassed by the invention. Additionally, fragments and variants of the above-described polynucleotides, nucleic acids, and polypeptides, are also encompassed by the invention.
  • polynucleotides of the invention comprise, or alternatively consist of, a polynucleotide sequence in which the 3' 10 polynucleotides of one of the sequences delineated in column 6 of Table IB and the 5' 10 polynucleotides of another sequence in column 6 are directly contiguous.
  • Nucleic acids which hybridize to the complement of these 20 contiguous polynucleotides under stringent hybridization conditions or alternatively, under lower stringency conditions, are also encompassed by the invention.
  • Polypeptides encoded by these polynucleotides and/or nucleic acids, other polynucleotides and/or nucleic acids encoding these polypeptides, and antibodies that bind these polypeptides are also encompassed by the invention. Additionally, fragments and variants of the above- described polynucleotides, nucleic acids, and polypeptides are also encompassed by the invention.
  • polynucleotides of the invention comprise, or alternatively consist of, a polynucleotide sequence in which the 3' 10 polynucleotides of one of the sequences delineated in column 6 of Table IB and the 5' 10 polynucleotides of another sequence in column 6 corresponding to the same Clone YD NO:Z (see Table IB, column 1) are directly contiguous. Nucleic acids which hybridize to the complement of these 20 lower stringency conditions, are also encompassed by the invention.
  • Polypeptides encoded by these polynucleotides and/or nucleic acids, other polynucleotides and/or nucleic acids encoding these polypeptides, and antibodies that bind these polypeptides are also encompassed by the invention. Additionally, fragments and variants of the above-described polynucleotides, nucleic acids, and polypeptides are also encompassed by the invention.
  • polynucleotides of the invention comprise, or alternatively consist of, a polynucleotide sequence in which the 3' 10 polynucleotides of one sequence in column 6 corresponding to the same contig sequence identifer SEQ ID NO:X (see Table IB, column 2) are directly contiguous. Nucleic acids which hybridize to the complement of these 20 contiguous polynucleotides under stringent hybridization conditions or alternatively, under lower stringency conditions, are also encompassed by the invention.
  • Polypeptides encoded by these polynucleotides and/or nucleic acids, other polynucleotides and/or nucleic acids encoding these polypeptides, and antibodies that bind these polypeptides are also encompassed by the invention. Additionally, fragments and variants of the above- described polynucleotides, nucleic acids, and polypeptides are also encompassed by the invention.
  • polynucleotides of the invention comprise, or alternatively consist of a polynucleotide sequence in which the 3' 10 polynucleotides of one of the sequences delineated in column 6 of Table IB and the 5' 10 polynucleotides of another sequence in column 6 corresponding to the same row are directly contiguous.
  • the 3' 10 polynucleotides of one of the sequences delineated in column 6 of Table IB is directly contiguous with the 5' 10 polynucleotides of the next sequential exon delineated in Table IB, column 6.
  • Nucleic acids which hybridize to the complement of these 20 contiguous polynucleotides under stringent hybridization conditions or alternatively, under lower stringency conditions are also encompassed by the invention.
  • Polypeptides encoded by these polynucleotides and/or nucleic acids, other polynucleotides and/or nucleic acids encoding these polypeptides, and antibodies that bind these polypeptides are also encompassed by the invention. Additionally, fragments and variants of the above-described polynucleotides, nucleic acids, and polypeptides are also encompassed by the invention.
  • polynucleotide sequences such as EST sequences, are publicly available and accessible through sequence databases and may have been publicly available prior to conception of the present invention. Preferably, such related polynucleotides are specifically excluded from the scope of the present invention.
  • each contig sequence (SEQ ED NO:X) listed in the fourth column of Table 1 A preferably excluded are one or more polynucleotides comprising a nucleotide sequence described by the general formula of a-b, where a is any integer between 1 and the final nucleotide minus 15 of SEQ ED NO:X, b is an integer of 15 to the final nucleotide of SEQ ID NO:X, where both a and b correspond to the positions of nucleotide residues shown in SEQ ID NO:X, and where b is greater than or equal to a + 14.
  • polynucleotides comprising a nucleotide sequence described by the general formula of a-b, where a and b are integers as defined in columns 4 and 5, respectively, of Table 3.
  • the polynucleotides of the invention do not consist of at least one, two, three, four, five, ten, or more of the specific polynucleotide sequences referenced by the Genbank Accession No. as disclosed in column 6 of Table 3 (including for example, published sequence in connection with a particular BAC clone).
  • preferably excluded from the invention are the specific polynucleotide sequence(s) contained in the clones corresponding to at least one, two, three, four, five, ten, or more of the available material having the accession numbers identified in the sixth column of this Table (including for example, the actual sequence contained in an identified BAC clone). In no way is this listing meant to encompass all of the sequences which may be excluded by the general formula, it is just a representative example. All references available through these accessions are hereby inco ⁇ orated by reference in their entirety.
  • HUJDC24 31 953517 1 - 1408 15 - 1422 AI467849, AI742229, AW148851,AI916736, AI281433,AA804534, AI564217,AI523942, AW364769, AW299493, AI620008, AI123945, AI377579, Z78359, AI191913,AI610109, AW085483, AA923067, AI769651,AI928062, AI078678, AA854786, AI025849,AI581271, AA609195,AA812157, All 50639, AI261346, AI433379, AA599239, AI743119,AW194073, AI760076, AA507048, AI041747,AW002493, AW391749,AI492025, AA890051,AA113313, AA004830, C06203, AW169615,AA814762, C75084, AI814568,
  • HWLLB11 102 954849 1-731 15-745 AI745636, andAA102414.
  • HNTEF53 103 954852 1 - 2342 15-2356 AA557324, AI655577, AI696732, AI923200, AA863360, AW262723, AI697332, AW275990, AI436648,AW276183, R56515, AI362521, R53456, R53457, D62878, AA337301.AA652746, AW264444, R56123, AA319338,D79346, D79250, N56346, AA886832, andAL138223.
  • HHAWC08 106 957942 1 - 1870 15-1884 AI860000,AI984180, AI052115,AW027098, AA507892, AI269682, AI860186, AA910656, AA911665,AI122607, AI982553, AI492064, AI148135,W56156,
  • HPLBP54 127 1212679 1 - 1901 15-1915 C16805,C17480,D58975, C18261,C16945,C18700, C18638,C18027,C16784, C 18260, C 18222, C17488, D58770,C16946,C18483, C17653,C17965,D58978, C 17490, C 18701,C 17740, C 18747, C 18449, C 17748, D58838,C18252,C17885, C18988,D58702,C18964, C19012,D58739,C18542, D58759, C16754, C17095, C17755,C17886,C16794, C18933,D58837,D59219, C17939, C17663, C17522, C17714, C17766,D58840, D58983,D59184,C16856, C18711.C17278, C17232, C17696, C17749,D58550, C
  • HRABU93 129 1206777 1 - 2267 15-2281 AW161406,W22101, AW163611,AW140025, AA326433,R90781, R53169,AA446363, U83543, AI380305, H19578, H23911,H51386,H19075, H46576, AI609704, D86957, A87006, AC004775, and AC005742.
  • HWLQH41 130 1176226 1 -1249 15 - 1263 AI417842,AI127930, AI202751,AW237652, AW241522,AI633241, AA828649, AI056043, AI097032, AA595596, AI332697,AW009101, AI391721,AI288942, AI742779, AI741429, AW338282, AI633779, AI218345, AI971047, AI290280, AI754557, AA812206, AA586780, AWl 17606, AI924489, AI284061,AI689103, AW207270,AW294513, AA568817,AI478517, AI657130, AI074577, AI990149, AA806346, N51855,AI149292,D61975, AI033281,AA384212, AI917469, N53363, N73526, H23985, R28358, AA677342, AA
  • HCEPH84 140 1148128 1 - 1499 15-1513 AA442393,AA019932, W76203, AI372800, R56176.AI372801, AW248425, AA350199, T77421, AA054057, AI298004, T75382, T47787, AA018176,F08410, AA058754, R55903, R36279, AAl 91072, AAl 80848, AAl 94490, F13127,AA193053, AA326216,R16315, AA663485, AAl 66864, AA204736, AA249839, F05591,AI651722, AAl 67722, Z42609, AW138074, AA339074, AA054202, AA056327, AI479964,AA207119, AI221332, F26963, F36674, F28731, AI937722, F36659, AI609568, F25641, F
  • HSHBW40 150 1144361 1-1120 15-1134 AA393046, AA449274, AI417305,H54361, AA450152,H89797, AA045551,AA315053, W28083,AI925931, AI810820, AA310873, R83856, R33893, T78727, R18673,AA306399, AA306395, AA126773, AA324948, AA450094, AW379133, T77732, T27901,W25779,T51655, W25559, AWl 89057, R14561,T78180,H48688, AW365287, AI342499, W25778, R56439, AI858990, AA374368, AW362215,AI609827, AA344165, AI655305, AA121371.H77659, R18418,R34534,H87657, AA378311,AI269259, R08
  • HGOCA12 173 1172054 1 - 1234 15 - 1248 AW444890, AA461185, AA435513, AA602372, AA723271, AA774585, AI971328, AI004443, W72923, F36785, F17742, F19634, W94217, AA346094, AA346095, F24441, AA393548, AA461361, and AB027004.
  • HCE5E94 174 1070802 1 - 1465 15 - 1479 AI142098, AI061467, AA555087, AI816028, AI064696, All 32975, AI955216, AI954923, AH 85092, AI961492, AA557261, AA594123, AI683136, AI821060, AA167739, AI492571, AA167833, AI128382, AI039982, AI801532, AI884567, AI708278, AA877359, AA771931, AI225219, AW169567, AW149057, W02976, AA570678, AA847293, AA738472, AI916354, AI568477, AI687057, AA570139, AA485692, AI075416, AW025307, AA009525, AI870831, N79600, AA524522, AA128450, AA447933,
  • the present invention is directed to variants of the polynucleotide sequence disclosed in SEQ ID NO:X or the complementary strand thereto, nucleotide sequences encoding the polypeptide of SEQ ID NO:Y, the nucleotide sequence of SEQ ID NO.X encoding the polypeptide sequence as defined in column 7 of Table 1A, nucleotide sequences encoding the polypeptide as defined in column 7 of Table 1A, the nucleotide sequence as defined in columns 8 and 9 of Table 2, nucleotide sequences encoding the polypeptide encoded by the nucleotide sequence as defined in columns 8 and 9 of Table 2, the nucleotide sequence as defined in column 6 of Table IB, nucleotide sequences encoding the polypeptide encoded by the nucleotide sequence as defined in column 6 of Table IB, the cDNA sequence contained in Clone ED NO:Z, and/or nucleotide sequences encoding the polypeptide encoded by the c
  • the present invention also encompasses variants of the polypeptide sequence disclosed in SEQ ED NO:Y, the polypeptide sequence as defined in column 7 of Table 1A, a polypeptide sequence encoded by the polynucleotide sequence in SEQ ED NO.X, a polypeptide sequence encoded by the nucleotide sequence as defined in columns 8 and 9 of Table 2, a polypeptide sequence encoded by the nucleotide sequence as defined in column 6 of Table IB, a polypeptide sequence encoded by the complement of the polynucleotide sequence in SEQ ED NO:X, and/or a polypeptide sequence encoded by the cDNA sequence contained in Clone ED NO:Z.
  • Variant refers to a polynucleotide or polypeptide differing from the polynucleotide or polypeptide of the present invention, but retaining essential properties thereof. Generally, variants are overall closely similar, and, in many regions, identical to the polynucleotide or polypeptide of the present invention.
  • one aspect of the invention provides an isolated nucleic acid molecule comprising, or alternatively consisting of, a polynucleotide having a nucleotide sequence selected from the group consisting of: (a) a nucleotide sequence described in SEQ ID NO:X or contained in the cDNA sequence of Clone ED NO:Z; (b) a nucleotide sequence in SEQ ID NO:X or the cDNA in Clone ED NO:Z which encodes the complete amino acid sequence of SEQ ED NO:Y or the complete amino acid sequence encoded by the cDNA in Clone ID NO:Z; (c) a nucleotide sequence in SEQ ID NO.X or the cDNA in Clone ID NO:Z which encodes a mature polypeptide; (d) a nucleotide sequence in SEQ ED NO:X or the cDNA sequence of Clone ID NO:Z, which encodes a biologically active fragment of a polypeptide; (e) a nucleotide sequence
  • the present invention is also directed to nucleic acid molecules which comprise, or alternatively consist of, a nucleotide sequence which is at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99% or 100%, identical to, for example, any of the nucleotide sequences in (a), (b), (c), (d), (e), (f), (g), (h), (i), or (j) above, the nucleotide coding sequence in SEQ ID NO.X or the complementary strand thereto, the nucleotide coding sequence of the cDNA contained in Clone ED NO:Z or the complementary strand thereto, a nucleotide sequence encoding the polypeptide of SEQ ED NO:Y, a nucleotide sequence encoding a polypeptide sequence encoded by the nucleotide sequence in SEQ ED NO:X, a polypeptide sequence encoded by the complement of the polynucleotide sequence in SEQ ED NO:X
  • polynucleotides which hybridize to the complement of these nucleic acid molecules under stringent hybridization conditions or alternatively, under lower stringency conditions are also encompassed by the invention, as are polypeptides encoded by these polynucleotides and nucleic acids.
  • the invention encompasses nucleic acid molecules which comprise, or alternatively, consist of a polynucleotide which hybridizes under stringent hybridization conditions, or alternatively, under lower stringency conditions, to a polynucleotide in (a), (b), (c), (d), (e), (f), (g), (h), or (i), above, as are polypeptides encoded by these polynucleotides.
  • polynucleotides which hybridize to the complement of these nucleic acid molecules under stringent hybridization conditions, or alternatively, under lower stringency conditions are also encompassed by the invention, as are polypeptides encoded by these polynucleotides.
  • the invention provides a purified protein comprising, or alternatively consisting of, a polypeptide having an amino acid sequence selected from the group consisting of: (a) the complete amino acid sequence of SEQ YD NO:Y or the complete amino acid sequence encoded by the cDNA in Clone ED NO:Z; (b) the amino acid sequence of a mature form of a polypeptide having the amino acid sequence of SEQ ID NO:Y or the amino acid sequence encoded by the cDNA in Clone ED NO:Z; (c) the amino acid sequence of a biologically active fragment of a polypeptide having the complete amino acid sequence of SEQ YD NO:Y or the complete amino acid sequence encoded by the cDNA in Clone ID NO:Z; and (d) the amino acid sequence of an antigenic fragment of a polypeptide having the complete amino acid sequence of SEQ ED NO:Y or the complete amino acid sequence encoded by the cDNA in Clone ED NO:Z.
  • the present invention is also directed to proteins which comprise, or alternatively consist of, an amino acid sequence which is at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99% or 100%, identical to, for example, any of the amino acid sequences in (a), (b), (c), or (d), above, the amino acid sequence shown in SEQ YD NO:Y, the amino acid sequence encoded by the cDNA contained in Clone ED NO:Z, the amino acid sequence of the polypeptide encoded by the nucleotide sequence in SEQ ED NO:X as defined in columns 8 and 9 of Table 2, the amino acid sequence of the polypeptide encoded by the nucleotide sequence in SEQ YD NO:B as defined in column 6 of Table IB, the amino acid sequence as defined in column 7 of Table 1A, an amino acid sequence encoded by the nucleotide sequence in SEQ ED NO:X, and an amino acid sequence encoded by the complement of the polynucleotide sequence in SEQ Y
  • polypeptides are also provided (e.g., those fragments described herein).
  • Further proteins encoded by polynucleotides which hybridize to the complement of the nucleic acid molecules encoding these amino acid sequences under stringent hybridization conditions or alternatively, under lower stringency conditions, are also encompassed by the invention, as are the polynucleotides encoding these proteins.
  • nucleotide sequence of the nucleic acid is identical to the reference sequence except that the nucleotide sequence may include up to five point mutations per each 100 nucleotides of the reference nucleotide sequence encoding the polypeptide.
  • nucleotide sequence may include up to five point mutations per each 100 nucleotides of the reference nucleotide sequence encoding the polypeptide.
  • nucleic acid 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.
  • the query sequence may be an entire sequence referred to in Table 1A or 2 as the ORF (open reading frame), or any fragment specified as described herein.
  • nucleic acid molecule or polypeptide is at least 80%, 85%, 90%, 95%, 96%, 97%, 98% or 99% identical to a nucleotide sequence of the present invention can be determined conventionally using known computer programs.
  • a preferred method for determining the best overall match between a query sequence (a sequence of the present invention) and a subject sequence, also referred to as a global sequence alignment, can be determined using the FASTDB computer program based on the algorithm of Brutlag et al. (Comp. App. Biosci. 6:237-245 (1990)). In a sequence alignment the query and subject sequences are both DNA sequences.
  • RNA sequence can be compared by converting U's to T's.
  • the result of said global sequence alignment is expressed as percent identity.
  • the percent identity is corrected by calculating the number of bases of the query sequence that are 5' and 3' of the subject sequence, which are not matched/aligned, as a percent of the total bases of the query sequence. Whether a nucleotide is matched/aligned is determined by results of the FASTDB sequence alignment.
  • This percentage is then subtracted from the percent identity, calculated by the above FASTDB program using the specified parameters, to arrive at a final percent identity score.
  • This corrected score is what is used for the purposes of the present invention. Only bases outside the 5' and 3' bases of the subject sequence, as displayed by the FASTDB alignment, which are not matched/aligned with the query sequence, are calculated for the purposes of tnanually adjusting the percent identity score.
  • a 90 base subject sequence is aligned to a 100 base query sequence to determine percent identity.
  • the deletions occur at the 5' end of the subject sequence and therefore, the FASTDB alignment does not show a matched/alignment of the first 10 bases at 5' end.
  • the 10 unpaired bases represent 10% of the sequence (number of bases at the 5' and 3' ends not matched/total number of bases in the query sequence) so 10% is subtracted from the percent identity score calculated by the FASTDB program. If the remaining 90 bases were perfectly matched the final percent identity would be 90%.
  • a 90 base subject sequence is compared with a 100 base query sequence.
  • deletions are internal deletions so that there are no bases on the 5' or 3' of the subject sequence which are not matched/aligned with the query.
  • percent identity calculated by FASTDB is not manually corrected.
  • bases 5' and 3' of the subject sequence which are not matched/aligned with the query sequence are manually corrected for. No other manual corrections are to be made for the purposes of the present invention.
  • amino acid sequence of the subject polypeptide is identical to the query sequence except that the subject polypeptide sequence may include up to five amino acid alterations per each 100 amino acids of the query amino acid sequence.
  • the subject polypeptide sequence may include up to five amino acid alterations per each 100 amino acids of the query amino acid sequence.
  • up to 5% of the amino acid residues in the subject sequence may be inserted, deleted, (indels) or substituted with another amino acid.
  • These alterations of the reference sequence may occur at the amino or carboxy terminal positions of the reference amino acid sequence or anywhere between those terminal positions, interspersed either individually among residues in the reference sequence or in one or more contiguous groups within the reference sequence.
  • a preferred method for determining the best overall match between a query sequence (a sequence of the present invention) and a subject sequence can be determined using the FASTDB computer program based on the algorithm of Brutlag et al. (Comp. App. Biosci.6:237-245 (1990)).
  • the query and subject sequences are either both nucleotide sequences or both amino acid sequences.
  • the result of said global sequence alignment is expressed as percent identity.
  • the percent identity is corrected by calculating the number of residues of the query sequence that are N- and C- terminal of the subject sequence, which are not matched/aligned with a corresponding subject residue, as a percent of the total bases of the query sequence. Whether a residue is matched/aligned is determined by results of the FASTDB sequence alignment. This percentage is then subtracted from the percent identity, calculated by the above FASTDB program using the specified parameters, to arrive at a final percent identity score. This final percent identity score is what is used for the purposes of the present invention.
  • a 90 amino acid residue subject sequence is aligned with a 100 residue query sequence to determine percent identity.
  • the deletion occurs at the N-terminus of the subject sequence and therefore, the FASTDB alignment does not show a matching/alignment of the first 10 residues at the N-terminus.
  • the 10 unpaired residues represent 10% of the sequence (number of residues at the N- and C- termini not matched/total tiumber of residues in the query sequence) so 10% is subtracted from the percent identity score calculated by the FASTDB program. If the remaining 90 residues were perfectly matched the final percent identity would be 90%.
  • a 90 residue subject sequence is compared with a 100 residue query sequence.
  • deletions are internal deletions so there are no residues at the N- or C-termini of the subject sequence which are not matched/aligned with the query.
  • percent identity calculated by FASTDB is not manually corrected.
  • residue positions outside the N- and C-terminal ends of the subject sequence, as displayed in the FASTDB alignment, which are not matched/aligned with the query sequnce are manually corrected for. No other manual corrections are to made for the purposes of the present invention.
  • the polynucleotide variants of the invention may contain alterations in the coding regions, non-coding regions, or both. Especially preferred are polynucleotide variants containing alterations which produce silent substitutions, additions, or deletions, but do not alter the properties or activities of the encoded polypeptide. Nucleotide variants produced by silent substitutions due to the degeneracy of the genetic code are preferred. Moreover, polypeptide variants in which less than 50, less than 40, less than 30, less than 20, less than
  • Polynucleotide variants can be produced for a variety of reasons, e.g., to optimize codon expression for a particular host (change codons in the human mRNA to those preferred by a bacterial host such as E. coli).
  • Naturally occurring variants are called "allelic variants," and refer to one of several alternate forms of a gene occupying a given locus on a chromosome of an organism. (Genes
  • allelic variants can vary at either the polynucleotide and/or polypeptide level and are included in the present invention.
  • non-naturally occurring variants may be produced by mutagenesis techniques or by direct synthesis.
  • variants may be generated to improve or alter the characteristics of the polypeptides of the present invention.
  • one or more amino acids can be deleted from the N-terminus or C-terminus of the polypeptide of the present invention without substantial loss of biological function.
  • Ron et al. J. Biol. Chem. 268: 2984-2988 (1993)
  • variant KGF proteins having heparin binding activity even after deleting 3, 8, or 27 amino-terminal amino acid residues.
  • Interferon gamma exhibited up to ten times higher activity after deleting 8-10 amino acid residues from the carboxy terminus of this protein. (Dobeli et al., J. Biotechnology 7:199-216 (1988).)
  • the invention further includes polypeptide variants which show a functional activity (e.g., biological activity) of the polypeptides of the invention.
  • a functional activity e.g., biological activity
  • variants include deletions, insertions, inversions, repeats, and substitutions selected according to general rules known in the art so as have little effect on activity.
  • the present application is directed to nucleic acid molecules at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99% or 100% identical to the nucleic acid sequences disclosed herein, (e.g., encoding a polypeptide having the amino acid sequence of an N and/or C terminal deletion), irrespective of whether they encode a polypeptide having functional activity. This is because even where a particular nucleic acid molecule does not encode a polypeptide having functional activity, one of skill in the art would still 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 functional activity include, inter alia, (1) isolating a gene or allelic or ⁇ plice variants thereof in a cDNA library; (2) in situ hybridization (e.g., "FISH") to metaphase chromosomal spreads to provide precise chromosomal location of the gene, as described in Verma et al., Human Chromosomes: A Manual of Basic Techniques, Pergamon Press, New York (1988); (3) Northern Blot analysis for detecting mRNA expression in specific tissues (e.g., normal or diseased tissues); and (4) in situ hybridization (e.g., histochemistry) for detecting mRNA expression in specific tissues (e.g., normal or diseased tissues).
  • in situ hybridization e.g., histochemistry
  • nucleic acid molecules having sequences at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99% or 100% identical to the nucleic acid sequences disclosed herein, which do, in fact, encode a polypeptide having functional activity.
  • a polypeptide having "functional activity” is meant, a polypeptide capable of displaying one or more known functional activities associated with a full-length (complete) protein of the invention.
  • Such functional activities include, but are not limited to, biological activity, antigenicity [ability to bind (or compete with a polypeptide of the invention for binding) to an anti-polypeptide of the invention antibody], immunogenicity (ability to generate antibody which binds to a specific polypeptide of the invention), ability to form multimers with polypeptides of the invention, and ability to bind to a receptor or ligand for a polypeptide of the invention.
  • polypeptides, and fragments, variants and derivatives of the invention can be assayed by various methods.
  • various immunoassays known in the art can be used, including but not limited to, competitive and non-competitive assay systems using techniques such as radioimmunoassays, ELISA (enzyme linked immunosorbent assay), "sandwich” immunoassays, immunoradiometric assays, gel diffusion precipitation reactions, immunodiffusion assays, in situ immunoassays (using colloidal gold, enzyme or radioisotope labels, for example), western blots, precipitation reactions, agglutination assays (e.g., gel agglutination assays, hemagglutination assays), complement fixation assays, immunofluorescence assays, protein A assays, and immunoelectrophoresis assays, etc.
  • competitive and non-competitive assay systems using techniques such as radioimmunoassays, ELISA (enzyme linked immunosorbent assay), "sandwich” immunoassays, immunoradiometric
  • antibody binding is detected by detecting a label on the primary antibody.
  • the primary antibody is detected by detecting binding of a secondary antibody or reagent to the primary antibody.
  • the secondary antibody is labeled. Many means are known in the art for detecting binding in an immunoassay and are within the scope of the present invention.
  • binding can be assayed, e.g., by means well-known in the art, such as, for example, reducing and non- reducing gel chromatography, protein affinity chromatography, and affinity blotting. See generally, Phizicky et al., Microbiol. Rev. 59:94-123 (1995).
  • the ability of physiological correlates of a polypeptide of the present invention to bind to a substrate(s) of the polypeptide of the invention can be routinely assayed using techniques known in the art.
  • nucleic acid molecules having a sequence at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99%, or 100% identical to, for example, the nucleic acid sequence of the cDNA contained in Clone YD NO:Z, the nucleic acid sequence referred to in Table 1A (SEQ ID NO:X), the nucleic acid sequence disclosed in Table 2 (e.g,.
  • nucleic acid sequence delineated in columns 8 and 9) or fragments thereof will encode polypeptides "having functional activity.”
  • degenerate variants of any of these nucleotide sequences all encode the same polypeptide, in many instances, this will be clear to the skilled artisan even without performing the above described comparison assay.
  • a reasonable number will also encode a polypeptide having functional activity. This is because the skilled artisan is fully aware of amino acid substitutions that are either less likely or not likely to significantly effect protein function (e.g., replacing one aliphatic amino acid with a second aliphatic amino acid), as further described below.
  • the first strategy exploits the tolerance of amino acid substitutions by natural selection during the process of evolution. By comparing amino acid sequences in different species, conserved amino acids can be identified. These conserved amino acids are likely important for protein function. In contrast, the amino acid positions where substitutions have been tolerated by natural selection indicates that these positions are not critical for protein function. Thus, positions tolerating amino acid substitution could be modified while still maintaining biological activity of the protein.
  • the second strategy uses genetic engineering to introduce amino acid changes at specific positions of a cloned gene to identify regions critical for protein function. For example, site directed mutagenesis or alanine-scanning mutagenesis (introduction of single alanine mutations at every residue in the molecule) can be used. See Cunningham and Wells, Science 244:1081-1085 (1989). The resulting mutant molecules can then be tested for biological activity.
  • tolerated conservative amino acid substitutions involve replacement of the aliphatic or hydrophobic amino acids Ala, Val, Leu and He; replacement of the hydroxyl residues Ser and Thr; replacement of the acidic residues Asp and Glu; replacement of the amide residues Asn and Gin, replacement of the basic residues Lys, Arg, and His; replacement of the aromatic residues Phe, Tyr, and T ⁇ , and replacement of the small-sized amino acids Ala, Ser, Thr, Met, and Gly.
  • variants of the present invention include (i) substitutions with one or more of the non- conserved amino acid residues, where the substituted amino acid residues may or may not be one encoded by the genetic code, or (ii) substitutions with one or more of the amino acid residues having a substituent group, or (iii) fusion of the mature polypeptide with another compound, such as a compound to increase the stability and/or solubility of the polypeptide (for example, polyethylene glycol), (iv) fusion of the polypeptide with additional amino acids, such as, for example, an IgG Fc fusion region peptide, serum albumin (preferably human serum albumin) or a fragment thereof, or leader or secretory sequence, or a sequence facilitating purification, or (v) fusion of the polypeptide with another compound, such as albumin (including but not limited to recombinant albumin (see, e.g., U.S.
  • polypeptide variants containing amino acid substitutions of charged amino acids with other charged or neutral amino acids may produce proteins with improved characteristics, such as less aggregation. Aggregation of pharmaceutical formulations both reduces activity and increases clearance due to the aggregate's immunogenic activity. See Pinckard et al., Clin. Exp. Immunol. 2:331-340 (1967); Robbins et al., Diabetes 36: 838-845 (1987); Cleland et al., Crit. Rev. Therapeutic Drug Carrier Systems 10:307-377 (1993).
  • a further embodiment of the invention relates to polypeptides which comprise the amino acid sequence of a polypeptide having an amino acid sequence which contains at least one amino acid substitution, but not more than 50 amino acid substitutions, even more preferably, not more than 40 amino acid substitutions, still more preferably, not more than 30 amino acid substitutions, and still even more preferably, not more than 20 amino acid substitutions from a polypeptide sequence disclosed herein.
  • a polypeptide prefferably has an amino acid sequence which comprises the amino acid sequence of a polypeptide of SEQ ID NO:Y, an amino acid sequence encoded by SEQ ID NO:X, an amino acid sequence encoded by the portion of SEQ ID NO:X as defined in columnns 8 and 9 of Table 2, an amino acid sequence encoded by the complement of SEQ ED NO:X, and/or an amino acid sequence encoded by cDNA contained in Clone ED NO:Z which contains, in order of ever-increasing preference, at least one, but not more than 10, 9, 8, 7, 6, 5, 4, 3, 2 or 1 amino acid substitutions.
  • the polypeptides of the invention comprise, or altematively, consist of, fragments or variants of a reference amino acid sequence selected from: (a) the amino acid sequence of SEQ YD NO:Y or fragments thereof (e.g., the mature form and/or other fragments described herein); (b) the amino acid sequence encoded by SEQ ID NO:X or fragments thereof; (c) the amino acid sequence encoded by the complement of SEQ ID NO:X or fragments thereof; (d) the amino acid sequence encoded by the portion of SEQ ED NO:X as defined in columns 8 and 9 of Table 2 or fragments thereof; and (e) the amino acid sequence encoded by cDNA contained in Clone ED NO:Z or fragments thereof; wherein the fragments or variants have 1-5, 5-10, 5-25, 5-50, 10-50 or 50-150, amino acid residue additions, substitutions, and/or deletions when compared to the reference amino acid sequence.
  • the amino acid substitutions are conservative. Polyn
  • polynucleotide fragment refers to a polynucleotide having a nucleic acid sequence which, for example: is a portion of the cDNA contained in Clone ID NO:Z or the complementary strand thereto; is a portion of the polynucleotide sequence encoding the polypeptide encoded by the cDNA contained in Clone ID NO:Z or the complementary strand thereto; is a portion of a polynucleotide sequence encoding the amino acid sequence encoded by the region of SEQ ID NO:X as defined in columns 8 and 9 of Table 2 or the complementary strand thereto; is a portion of the polynucleotide sequence of SEQ ED NO.X as defined in columns 8 and 9 of Table 2 or the complementary strand thereto; is a portion of the polynucleotide sequence of SEQ ED NO.X as defined in columns 8 and 9 of Table 2 or the complementary strand thereto; is a portion of the polynucleotide sequence of SEQ ED NO
  • the polynucleotide fragments of the invention are preferably at least about 15 nt, and more preferably at least about 20 nt, still more preferably at least about 30 nt, and even more preferably, at least about 40 nt, at least about 50 nt, at least about 75 nt, or at least about 150 nt in length.
  • a fragment "at least 20 nt in length,” for example, is intended to include 20 or more contiguous bases from the cDNA sequence contained in Clone ED NO:Z, or the nucleotide sequence shown in SEQ ED NO:X or the complementary stand thereto.
  • nucleotide fragments include, but are not limited to, as diagnostic probes and primers as discussed herein.
  • larger fragments e.g., at least 160, 170, 180, 190, 200, 250, 500, 600, 1000, or 2000 nucleotides in length ) are also encompassed by the invention.
  • polynucleotide fragments of the invention comprise, or alternatively consist of, a sequence from about nucleotide number 1-50, 51-100, 101-150, 151-200, 201-250, 251-300, 301-350, 351-400, 401-450, 451-500, 501-550, 551- 600, 601-650, 651-700, 701-750, 751-800, 801-850, 851-900, 901-950, 951-1000, 1001- 1050, 1051-1100, 1 101-1 150, 1 151-1200, 1201-1250, 1251-1300, 1301-1350, 1351-1400, 1401-1450, 1451-1500, 1501-1550, 1551-1600, 1601-1650, 1651-1700, 1701-1750, 1751- 1800, 1801-1850, 1851-1900, 1901-1950, 1951-2000, 2001-2050, 2051-2100, 2101-2150, 2151-2200, 2201-2250,
  • polynucleotide fragments of the invention Comprise, or alternatively consist of, a sequence from about nucleotide number 1-50, 51-100, 101-150, 151-200, 201 -250, 251-300, 301-350, 351-400, 401-450, 451-500, 501-550, 551- 600, 601-650, 651-700, 701-750, 751-800, 801-850, 851-900, 901-950, 951-1000, 1001- 1050, 1051-1100, 1 101-1 150, 1 151-1200, 1201-1250, 1251-1300, 1301-1350, 1351-1400, 1401-1450, 1451-1500, 1501-1550, 1551-1600, 1601-1650, 1651-1700, 1701-1750, 1751- 1800, 1801-1850, 1851-1900, 1901-1950, 1951-2000, 2001-2050, 2051-2100, 2101-2150, 2151-2200, 2201-2250,
  • polynucleotide fragments of the invention comprise, or alternatively consist of, a nucleic acid sequence comprising one, two, three, four, five, six, seven, eight, nine, ten, or more of the above described polynucleotide fragments of the invention in combination with a polynucleotide sequence delineated in Table IB column 6.
  • polynucleotide fragments of the invention comprise, or altematively consist of, a nucleic acid sequence comprising one, two, three, four, five, six, seven, eight, nine, ten, or more of the above described polynucleotide fragments of the invention in combination with a polynucleotide sequence that is the complementary strand of a sequence delineated in column 6 of Table IB.
  • the above-described polynucleotide fragments of the invention comprise, or alternatively consist of, sequences delineated in Table IB, column 6, and have a nucleic acid sequence which is different from that of the BAC fragment having the sequence disclosed in SEQ ID NO:B (see Table I B, column 5).
  • the above-described polynucleotide fragments of the invention comprise, or alternatively consist of, sequences delineated in Table IB, column 6, and have a nucleic acid sequence which is different from that published for the BAC clone identified as BAC ED NO:A (see Table IB, column 4).
  • the above-described polynucleotides of the invention comprise, or alternatively consist of, sequences delineated Table IB, column 6, and have a nucleic acid sequence which is different from that contained in the BAC clone identified as BAC ED NO:A (see Table IB, column 4).
  • polynucleotides of the invention comprise, or alternatively consist of, one, two, three, four, five, six, seven, eight, nine, ten, or more fragments of the sequences delineated in column 6 of Table IB, and the polynucleotide sequence of SEQ ID NO:X (e.g., as defined in Table IB, column 2) or fragments or variants thereof.
  • polypeptides encoded by these polynucleotides, other polynucleotides that encode these polypeptides, and antibodies that bind these polypeptides are also encompassed by the invention.
  • polynucleotides of the invention comprise, or altematively consist of, one, two, three, four, five, six, seven, eight, nine, ten, or more fragments of the sequences delineated in column 6 of Table IB which correspond to the same Clone ID NO:Z (see Table IB, column 1), and the polynucleotide sequence of SEQ ID NO:X (e.g., as defined in Table 1A or IB) or fragments or variants thereof.
  • Polypeptides encoded by these polynucleotides, other polynucleotides that encode these polypeptides, and antibodies that bind these polypeptides are also encompassed by the invention.
  • polynucleotides of the invention comprise, or alternatively consist of, one, two, three, four, five, six, seven, eight, nine, ten, or more fragments of the sequences delineated in the same row of column 6 of Table IB, and the polynucleotide sequence of SEQ ED NO:X (e.g., as defined in Table 1A or IB) or fragments or variants thereof.
  • Polypeptides encoded by these polynucleotides, other polynucleotides that encode these polypeptides, and antibodies that bind these polypeptides are also encompassed by the invention.
  • polynucleotides of the invention comprise, or alternatively consist of a polynucleotide sequence in which the 3' 10 polynucleotides of one of the sequences delineated in column 6 of Table IB and the 5' 10 polynucleotides of the sequence of SEQ ID NO:X are directly contiguous. Nucleic acids which hybridize to the complement of these 20 contiguous polynucleotides under stringent hybridization conditions or alternatively, under lower stringency conditions, are also encompassed by the invention.
  • Polypeptides encoded by these polynucleotides and/or nucleic acids, other polynucleotides and/or nucleic acids that encode these polypeptides, and antibodies that bind these polypeptides are also encompassed by the invention. Additionally, fragments and variants of the above-described polynucleotides, nucleic acids, and polypeptides are also encompassed by the invention.
  • polynucleotides of the invention comprise, or alternatively consist of a polynucleotide sequence in which the 3' 10 polynucleotides of one of the sequences delineated in column 6 of Table IB and the 5' 10 polynucleotides of a fragment or variant of the sequence of SEQ ID NO:X (e.g., as described herein) are directly contiguous Nucleic acids which hybridize to the complement of these 20 contiguous polynucleotides under stringent hybridization conditions or alternatively, under lower stringency conditions, are also encompassed by the invention.
  • Polypeptides encoded by these polynucleotides and/or nucleic acids, other polynucleotides and/or nucleic acids encoding these polypeptides, and antibodies that bind these polypeptides are also encompassed by the invention. Additionally, fragments and variants of the above-described polynucleotides, nucleic acids, and polypeptides are also encompassed by the invention.
  • polynucleotides of the invention comprise, or alternatively consist of a polynucleotide sequence in which the 3' 10 polynucleotides of a fragment or variant of the sequence of SEQ ID NO:X and the 5' 10 polynucleotides of the sequence of one of the sequences delineated in column 6 of Table IB are directly contiguous. Nucleic acids which hybridize to the complement of these 20 contiguous polynucleotides under stringent hybridization conditions or alternatively, under lower stringency conditions, are also encompassed by the invention.
  • Polypeptides encoded by these polynucleotides and/or nucleic acids, other polynucleotides and/or nucleic acids encoding these polypeptides, and antibodies that bind these polypeptides are also encompassed by the invention. Additionally, fragments and variants of the above-described polynucleotides, nucleic acids, and polypeptides are also encompassed by the invention.
  • polynucleotides of the invention comprise, or alternatively consist of a polynucleotide sequence in which the 3' 10 polynucleotides of one of the sequences delineated in column 6 of Table IB and the 5' 10 polynucleotides of another sequence in column 6 are directly contiguous.
  • the 3' 10 polynucleotides of one of the sequences delineated in column 6 of Table IB is directly contiguous with the 5' 10 polynucleotides of the next sequential exon delineated in Table IB, column 6.
  • Nucleic acids which hybridize to the complement of these 20 contiguous polynucleotides under stringent hybridization conditions or alternatively, under lower stringency conditions are also encompassed by the invention.
  • Polypeptides encoded by these polynucleotides and/or nucleic acids, other polynucleotides and/or nucleic acids encoding these polypeptides, and antibodies that bind these polypeptides are also encompassed by the invention. Additionally, fragments and variants of the above-described polynucleotides, nucleic acids, and polypeptides are also encompassed by the invention.
  • a "polypeptide fragment” refers to an amino acid sequence which is a portion of that contained in SEQ ID NO:Y, a portion of an amino acid sequence encoded by the portion of SEQ ID NO:X as defined in columnns 8 and 9 of Table 2, a portion of an amino acid sequence encoded by the polynucleotide sequence of SEQ ED NO:X, a portion of an amino acid sequence encoded by the complement of the polynucleotide sequence in SEQ YD NO:X, and/or a portion of an amino acid sequence encoded by the cDNA contained in Clone ED NO:Z.
  • Protein (polypeptide) fragments may be "free-standing," or comprised within a larger polypeptide of which the fragment forms a part or region, most preferably as a single continuous region.
  • Representative examples of polypeptide fragments of the invention include, for example, fragments comprising, or alternatively consisting of, from about amino acid number 1-20, 21-40, 41-60, 61-80, 81-100, 101-120, 121-140, 141-160, 161-180, 181-200, 201-220, 221-240, 241-260, 261-280, 281- 300, 301-320, 321-340, 341-360, 361-380, 381-400, 401-420, 421-440, 441-460, 461-480, 481-500, 501-520, 521 -540, 541-560, 561-580, 581-600, 601-620, 621-640, 641-660, 661- 680, 681-700, 701-720, 721-740, 741-760, 761
  • polypeptide fragments of the invention include, for example, fragments comprising, or alternatively consisting of, from about amino acid number 1-20, 21-40, 41-60, 61-80, 81-100, 101-120, 121-140, 141-160, 161-180, 181-200, 201-220, 221-240, 241-260, 261-280, 281-300, 301-320, 321-340, 341- 360, 361-380, 381 -400, 401-420, 421-440, 441-460, 461-480, 481-500, 501-520, 521-540, 541-560, 561-580, 581-600, 601-620, 621-640, 641-660, 661-680, 681-700, 701-720, 721- 740, 741-760, 761-780, 781-800, 801-820, 821-840, 841-860, 861-880, 881-900, 901-920, 921-940, 941-
  • polypeptide fragments of the invention may be at least about 10, 15, 20, 25, 30, 35, 40, 45, 50, 55, 60, 65, 70, 75, 80, 85, 90, 100, 110, 120, 130, 140, or 150 amino acids in length.
  • “about” includes the particularly recited ranges or values, or ranges or values larger or smaller by several (5, 4, 3, 2, or 1) amino acids, at either extreme or at both extremes.
  • Polynucleotides encoding these polypeptide fragments are also encompassed by the invention.
  • polypeptide fragments include the secreted protein as well as the mature form. Further preferred polypeptide fragments include the secreted protein or the mature form having a continuous series of deleted residues from the amino or the carboxy terminus, or both. For example, any number of amino acids, ranging from 1-60, can be deleted from the amino terminus of either the secreted polypeptide or the mature form. Similarly, any number of amino acids, ranging from 1-30, can be deleted from the carboxy terminus of the secreted protein or mature form. Furthermore, any combination of the above amino and carboxy tenninus deletions are preferred. Similarly, polynucleotides encoding these polypeptide fragments arc also preferred.
  • the present invention further provides polypeptides having one or more residues deleted from the amino terminus of the amino acid sequence of a polypeptide disclosed herein (e.g., a polypeptide of SEQ YD NO:Y, a polypeptide encoded by the polynucleotide sequence contained in SEQ ED NO:X or the complement thereof, a polypeptide encoded by the portion of SEQ ID NO:X as defined in columns 8 and 9 of Table 2, a polypeptide encoded by the portion of SEQ YD NO:B as defined in column 6 of Table IB, and/or a polypeptide encoded by the cDNA contained in Clone ED NO:Z).
  • a polypeptide disclosed herein e.g., a polypeptide of SEQ YD NO:Y, a polypeptide encoded by the polynucleotide sequence contained in SEQ ED NO:X or the complement thereof, a polypeptide encoded by the portion of SEQ ID NO:X as defined in columns 8 and 9
  • N-terminal deletions may be described by the general formula m-q, where q is a whole integer representing the total number of amino acid residues in a polypeptide of the invention (e.g., the polypeptide disclosed in SEQ ID NO:Y, or the polypeptide encoded by the portion of SEQ YD NO:X as defined in columns 8 and 9 of Table 2), and m is defined as any integer ranging from 2 to q-6. Polynucleotides encoding these polypeptides are also encompassed by the invention.
  • the present invention further provides polypeptides having one or more residues from the carboxy terminus of the amino acid sequence of a polypeptide disclosed herein (e.g., a polypeptide of SEQ ID NO:Y, a polypeptide encoded by the polynucleotide sequence contained in SEQ ID NO:X, a polypeptide encoded by the portion of SEQ YD NO:X as defined in columns 8 and 9 of Table 2, and/or a polypeptide encoded by the cDNA contained in Clone ED NO:Z).
  • a polypeptide of SEQ ID NO:Y e.g., a polypeptide of SEQ ID NO:Y, a polypeptide encoded by the polynucleotide sequence contained in SEQ ID NO:X, a polypeptide encoded by the portion of SEQ YD NO:X as defined in columns 8 and 9 of Table 2, and/or a polypeptide encoded by the cDNA contained in Clone ED NO:Z.
  • C-terminal deletions may be described by the general formula 1-n, where n is any whole integer ranging from 6 to q-1, and where n corresponds to the position of amino acid residue in a polypeptide of the invention. Polynucleotides encoding these polypeptides are also encompassed by the invention. [138] In addition, any of the above described N- or C-terminal deletions can be Combined to produce a N- and C-terminal deleted polypeptide.
  • the invention also provides polypeptides having one or more amino acids deleted from both the amino and the carboxyl termini, which may be described generally as having residues m-n of a polypeptide encoded by SEQ ID NO:X (e.g., including, but not limited to, the preferred polypeptide disclosed as SEQ ID NO:Y and the polypeptide encoded by the portion of SEQ ED NO:X as defined in columns 8 and 9 of Table 2), the cDNA contained in Clone YD NO:Z, and/or the complement thereof, where n and m are integers as described above. Polynucleotides encoding these polypeptides are also encompassed by the invention.
  • the present application is also directed to proteins containing polypeptides at least 80%, 85%, 90%, 95%, 96%, 97%, 98% or 99% identical to a polypeptide sequence set forth herein. In preferred embodiments, the application is directed to proteins containing polypeptides at least 80%, 85%, 90%, 95%, 96%, 97%, 98% or 99% identical to polypeptides having the amino acid sequence of the specific N- and C-terminal deletions. Polynucleotides encoding these polypeptides are also encompassed by the invention.
  • Any polypeptide sequence encoded by, for example, the polynucleotide sequences set forth as SEQ YD NO:X or the complement thereof, (presented, for example, in Tables 1 A and 2), the cDNA contained in Clone ID NO:Z, or the polynucleotide sequence as defined in column 6 of Table IB, may be analyzed to determine certain prefe ⁇ ed regions of the polypeptide.
  • amino acid sequence of a polypeptide encoded by a polynucleotide sequence of SEQ YD NO:X e.g., the polypeptide of SEQ ED NO:Y and the polypeptide encoded by the portion of SEQ YD NO:X as defined in columnns 8 and 9 of Table 2
  • the cDNA contained in Clone YD NO:Z may be analyzed using the default parameters of the DNASTAR computer algorithm (DNASTAR, Inc., 1228 S. Park St., Madison, WI 53715 USA; http://www.dnastar.com/).
  • Polypeptide regions that may be routinely obtained using the DNASTAR computer algorithm include, but are not limited to, Gamier-Robson alpha-regions, beta-regions, tum-regions, and coil-regions; Chou-Fasman alpha-regions, beta-regions, and tum-regions; Kyte-Doolittle hydrophilic regions and hydrophobic regions; Eisenberg alpha- and beta-amphipathic regions; Ka ⁇ lus-Schulz flexible regions; Emini surface-forming regions; and Jameson- Wolf regions of high antigenic index.
  • highly prefe ⁇ ed polynucleotides of the invention in this regard are those that encode polypeptides comprising regions that combine several structural features, such as several (e.g., 1, 2, 3 or 4) of the features set out above.
  • Kyte-Doolittle hydrophilic regions and hydrophobic regions, Emini surface-forming regions, and Jameson- Wolf regions of high antigenic index can routinely be used to determine polypeptide regions that exhibit a high degree of potential for antigenicity. Regions of high antigenicity are determined from data by DNASTAR analysis by choosing values which represent regions of the polypeptide which are likely to be exposed on the surface of the polypeptide in an environment in which antigen recognition may occur in the process of initiation of an immune response.
  • polypeptide fragments of the invention are fragments comprising, or alternatively, consisting of, an amino acid sequence that displays a functional activity (e.g. biological activity) of the polypeptide sequence of which the amino acid sequence is a fragment.
  • a polypeptide displaying a "functional activity” is meant a polypeptide capable of one or more known functional activities associated with a full-length protein, such as, for example, biological activity, antigenicity, immunogenicity, and/or multimerization, as described herein.
  • polypeptides of the invention comprise, or alternatively consist of, one, two, three, four, five or more of the antigenic fragments of the polypeptide of SEQ ED NO:Y, or portions thereof. Polynucleotides encoding these polypeptides are also encompassed by the invention.
  • the present invention encompasses polypeptides comprising, or alternatively consisting of, an epitope of: the polypeptide sequence shown in SEQ ED NO:Y; a polypeptide sequence encoded by SEQ ID NO:X or the complementary strand thereto; the polypeptide sequence encoded by the portion of SEQ YD NO:X as defined in columns 8 and 9 of Table 2; the polypeptide sequence encoded by the portion of SEQ ED NO:B as defined in column 6 of Table IB or the complement thereto; the polypeptide sequence encoded by the cDNA contained in Clone ID NO:Z; or the polypeptide sequence encoded by a polynucleotide that hybridizes to the sequence of SEQ ID NO:X, the complement of the sequence of SEQ ED NO:X, the complement of a portion of SEQ ED NO:X as defined in columns 8 and 9 of Table 2, or the cDNA sequence contained in Clone ED NO:Z under stringent hybridization conditions or alternatively, under lower stringency hybridization as defined
  • the present invention further encompasses polynucleotide sequences encoding an epitope of a polypeptide sequence of the invention (such as, for example, the sequence disclosed in SEQ YD NO:X, or a fragment thereof), polynucleotide sequences of the complementary strand of a polynucleotide sequence encoding an epitope of the invention, and polynucleotide sequences which hybridize to the complementary strand under stringent hybridization conditions or alternatively, under lower stringency hybridization conditions defined supra.
  • epitope of a polypeptide sequence of the invention such as, for example, the sequence disclosed in SEQ YD NO:X, or a fragment thereof
  • polynucleotide sequences of the complementary strand of a polynucleotide sequence encoding an epitope of the invention and polynucleotide sequences which hybridize to the complementary strand under stringent hybridization conditions or alternatively, under lower stringency hybridization conditions defined supra.
  • the present invention encompasses a polypeptide comprising an epitope, as well as the polynucleotide encoding this polypeptide.
  • An "immunogenic epitope,” as used herein, is defined as a portion of a protein that elicits an antibody response in an animal, as determined by any method known in the art, for example, by the methods for generating antibodies described infra. (See, for example, Geysen et al., Proc. Natl. Acad. Sci. USA 81 :3998- 4002 (1983)).
  • antigenic epitope is defined as a portion of a protein to which an antibody can immunospecifically bind its antigen as determined by any method well known in the art, for example, by the immunoassays described herein. Immunospecific binding excludes non-specific binding but does not necessarily exclude cross- reactivity with other antigens. Antigenic epitopes need hot necessarily be immunogenic.
  • Fragments which function as epitopes may be produced by any conventional means. (See, e.g., Houghten, R. A., Proc. Natl. Acad. Sci. USA 82:5131-5135 (1985) further described in U.S. Patent No. 4,631 ,211.)
  • antigenic epitopes preferably contain a sequence of at least 4, at least 5, at least 6, at least 7, more preferably at least 8, at least 9, at least 10, at least 11, at least 12, at least 13, at least 14, at least 15, at least 20, at least 25, at least 30, at least 40, at least 50, and, most preferably, between about 15 to about 30 amino acids.
  • Prefe ⁇ ed polypeptides comprising immunogenic or antigenic epitopes are at least 10, 15, 20, 25, 30, 35, 40, 45, 50, 55, 60, 65, 70, 75, 80, 85, 90, 95, or 100 amino acid residues in length.
  • Additional non-exclusive prefe ⁇ ed antigenic epitopes include the antigenic epitopes disclosed herein, as well as portions thereof.
  • Antigenic epitopes are useful, for example, to raise antibodies, including monoclonal antibodies, that specifically bind the epitope.
  • Prefe ⁇ ed antigenic epitopes include the antigenic epitopes disclosed herein, as well as any combination of two, three, four, five or more of these antigenic epitopes.
  • Antigenic epitopes can be used as the target molecules in immunoassays. (See, for instance, Wilson et al., Cell 37:767-778 (1984); Sutcliffe et al., Science 219:660-666 (1983)).
  • Non-limiting examples of epitopes of polypeptides that can be used to generate antibodies of the invention include a polypeptide comprising, or alternatively consisting of, at least one, two, three, four, five, six or more of the portion(s) of SEQ ED NO:Y specified in column 7 of Table 1A. These polypeptide fragments have been determined to bear antigenic epitopes of the proteins of the invention by the analysis of the Jameson- Wolf antigenic index which is included in the DNAStar suite of computer programs.
  • a polypeptide contains at least one, two, three, four, five, six or more of the portion(s) of SEQ ED NO:Y shown in column 7 of Table 1A, but it may contain additional flanking residues on either the amino or carboxyl termini of the recited portion.
  • additional flanking sequences are preferably sequences naturally found adjacent to the portion; i.e., contiguous sequence shown in SEQ YD NO:Y.
  • the flanking sequence may, however, be sequences from a heterolgous polypeptide, such as from another protein described herein or from a heterologous polypeptide not described herein.
  • epitope portions of a polypeptide of the invention comprise one, two, three, or more of the portions of SEQ YD NO:Y shown in column 7 of Table 1A.
  • immunogenic epitopes can be used, for example, to induce antibodies according to methods well known in the art. See, for instance, Sutcliffe et al., supra; Wilson et al., supra; Chow et al., Proc. Natl. Acad. Sci. USA 82:910-914; and Bittle et al., J. Gen. Virol. 66:2347-2354 (1985).
  • immunogenic epitopes include the immunogenic epitopes disclosed herein, as well as any combination of two, three, four, five or more of these immunogenic epitopes.
  • the polypeptides comprising one or more immunogenic epitopes may be presented for eliciting an antibody response together with a carrier protein, such as an albumin, to an animal system (such as rabbit or mouse), or, ifthe polypeptide is of sufficient length (at least about 25 amino acids), the polypeptide may be presented without a carrier.
  • immunogenic epitopes comprising as few as 8 to 10 amino acids have been shown to be sufficient to raise antibodies capable of binding to, at the very least, linear epitopes in a denatured polypeptide (e.g., in Western blotting).
  • Epitope-bearing polypeptides of the present invention may be used to induce antibodies according to methods well known in the art including, but not limited to, in vivo immunization, in vitro immunization, and phage display methods. See, e.g., Sutcliffe et al., supra; Wilson et al., supra, and Bittle et al., J. Gen. Virol., 66:2347-2354 (1985).
  • animals may be immunized with free peptide; however, anti-peptide antibody titer may be boosted by coupling the peptide to a macromolecular carrier, such as keyhole limpet hemacyanin (KLH) or tetanus toxoid.
  • KLH keyhole limpet hemacyanin
  • peptides containing cysteine residues may be coupled to a carrier using a linker such as maleimidobenzoyl- N- hydroxysuccinimide ester (MBS), while other peptides may be coupled to carriers using a more general linking agent such as glutaraldehyde.
  • Animals such as rabbits, rats and mice are immunized with either free or carrier- coupled peptides, for instance, by intraperitoneal and/or intradermal injection of emulsions containing about 100 ⁇ g of peptide or carrier protein and Freund's adjuvant or any other adjuvant known for stimulating an immune response.
  • booster injections may be needed, for instance, at intervals of about two weeks, to provide a useful titer of anti-peptide antibody which can be detected, for example, by ELISA assay using free peptide adsorbed to a solid surface.
  • the titer of anti-peptide antibodies in serum from an immunized animal may be increased by selection of anti-peptide antibodies, for instance, by adso ⁇ tion to the peptide on a solid support and elution of the selected antibodies according to methods well known in the art.
  • polypeptides of the present invention can be fused to heterologous polypeptide sequences.
  • polypeptides of the present invention may be fused with the constant domain of immunoglobulins (IgA, IgE, IgG, IgM), or portions thereof (CHI, CH2, CH3, or any combination thereof and portions thereof, resulting in chimeric polypeptides.
  • polypeptides and/or antibodies of the present invention may be fused with albumin (including but not limited to recombinant human serum albumin or fragments or variants thereof (see, e.g., U.S. Patent No. 5,876,969, issued March 2, 1999, EP Patent 0 413 622, and U.S. Patent No. 5,766,883, issued June 16, 1998, herein inco ⁇ orated by reference in their entirety)).
  • albumin including but not limited to recombinant human serum albumin or fragments or variants thereof (see, e.g., U.S. Patent No. 5,876,969, issued March 2, 1999, EP Patent 0 413 622, and U.S. Patent No. 5,766,883, issued June 16, 1998, herein inco ⁇ orated by reference in their entirety)).
  • polypeptides and/or antibodies of the present invention are fused with the mature form of human serum albumin (i.e., amino acids 1 - 585 of human semm albumin as shown in Figures 1 and 2 of EP Patent 0 322 094) which is herein inco ⁇ orated by reference in its entirety.
  • polypeptides and/or antibodies of the present invention are fused with polypeptide fragments comprising, or alternatively consisting of, amino acid residues 1-z of human semm albumin, where z is an integer from 369 to 419, as described in U.S.
  • Patent 5,766,883 herein inco ⁇ orated by reference in its entirety.
  • Polypeptides and/or antibodies of the present invention may be fused to either the N- or C-terminal end of the heterologous protein (e.g., immunoglobulin Fc polypeptide or human semm albumin polypeptide).
  • Polynucleotides encoding fusion proteins of the invention are also encompassed by the invention. [155] Such fusion proteins as those described above may facilitate purification and may increase half-life in vivo. This has been shown 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.
  • IgG fusion proteins that have a disulfide- linked dimeric structure due to the IgG portion desulfide bonds have also been found to be more efficient in binding and neutralizing other molecules than monomeric polypeptides or fragments thereof alone.
  • Nucleic acids encoding the above epitopes can also be recombined with a gene of interest as an epitope tag (e.g., the hemagglutinin (HA) tag or flag tag) to aid in detection and purification of the expressed polypeptide.
  • an epitope tag e.g., the hemagglutinin (HA) tag or flag tag
  • HA hemagglutinin
  • a system described by Janknecht et al. allows for the ready purification of non-denatured fusion proteins expressed in human cell lines (Janknecht et al., 1991 , Proc. Natl. Acad. Sci. USA 88:8972- 897).
  • the gene of interest is subcloned into a vaccinia recombination plasmid such that the open reading frame of the gene is translationally fused to an amino-terminal tag consisting of six histidine residues.
  • the tag serves as a matrix binding domain for the fusion protein. Extracts from cells infected with the recombinant vaccinia vims are loaded onto Ni2+ nitriloacetic acid-agarose column and histidine-tagged proteins can be selectively eluted with imidazole-containing buffers.
  • any polypeptide of the present invention can be used to generate fusion proteins.
  • the polypeptide of the present invention when fused to a second protein, can be used as an antigenic tag.
  • Antibodies raised against the polypeptide of the present invention can be used to indirectly detect the second protein by binding to the polypeptide.
  • secreted proteins target cellular locations based on trafficking signals
  • polypeptides of the present invention which are shown to be secreted can be used as targeting molecules once fused to other proteins.
  • domains that can be fused to polypeptides of the present invention include not only heterologous signal sequences, but also other heterologous functional regions.
  • the fusion does not necessarily need to be direct, but may occur through linker sequences.
  • proteins of the invention are fusion proteins comprising an amino acid sequence that is an N and/or C- terminal deletion of a polypeptide of the invention.
  • the invention is directed to a fusion protein comprising an amino acid sequence that is at least 90%, 95%, 96%, 97%, 98% or 99% identical to a polypeptide sequence of the invention. Polynucleotides encoding these proteins are also encompassed by the invention.
  • fusion proteins may also be engineered to improve characteristics of the polypeptide of the present invention. For instance, a region of additional amino acids, particularly charged amino acids, may be added to the N-terminus of the polypeptide to improve stability and persistence during purification from the host cell or 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. The addition of peptide moieties to facilitate handling of polypeptides are familiar and routine techniques in the art.
  • polypeptides of the present invention can be combined with heterologous polypeptide sequences.
  • the polypeptides of the present invention may be fused with heterologous polypeptide sequences, for example, the polypeptides of the present invention may be fused with the constant domain of immunoglobulins (IgA, IgE, IgG, IgM) or portions thereof (CHI, CH2, CH3, and any combination thereof, including both entire domains and portions thereof), or albumin (including, but not limited to, native or recombinant human albumin or fragments or variants thereof (see, e.g., U.S. Patent No.
  • EP-A-O 464 533 (Canadian counte ⁇ art 2045869) discloses fusion proteins comprising various portions of constant region of immunoglobulin molecules together with another human protein or part thereof.
  • the Fc part in a fusion protein is beneficial in therapy and diagnosis, and thus can result in, for example, improved pharmacokinetic properties (EP-A 0232 262).
  • deleting the Fc part after the fusion protein has been expressed, detected, and purified, would be desired.
  • the Fc portion may hinder therapy and diagnosis if the fusion protein is used as an antigen for immunizations.
  • human proteins such as hEL-5
  • Fc portions for the pu ⁇ ose of high-throughput screening assays to identify antagonists of hEL-5. See, D. Bennett et al., J. Molecular Recognition 8:52-58 (1995); K. Johanson et al., J. Biol. Chem. 270:9459-9471 (1995).
  • the polypeptides of the present invention can be fused to marker sequences, such as a polypeptide which facilitates purification of the fused polypeptide.
  • the marker amino acid sequence is a hexa-histidine peptide, such as the tag provided in a pQE vector (QIAGEN, Inc., 9259 Eton Avenue, Chatsworth, CA, 91311), among others, many of which are commercially available.
  • hexa-histidine provides for convenient purification of the fusion protein.
  • Another peptide tag useful for purification, the "HA" tag co ⁇ esponds to an epitope derived from the influenza hemagglutinin protein ( ⁇ Wilson et al., Cell 37:767 (1984)).
  • DNA shuffling may be employed to modulate the activities of polypeptides of the invention, such methods can be used to generate polypeptides with altered activity, as well as agonists and antagonists of the polypeptides. See, generally, U.S. Patent Nos. 5,605,793; 5,811,238; 5,830,721; 5,834,252; and 5,837,458, and Patten et al., Cu ⁇ .
  • alteration of polynucleotides co ⁇ esponding to SEQ ID NO:X and the polypeptides encoded by these polynucleotides may be achieved by DNA shuffling.
  • DNA shuffling involves the assembly of two or more DNA segments by homologous or site-specific recombination to generate variation in the polynucleotide sequence.
  • polynucleotides of the invention, or the encoded polypeptides may be altered by being subjected to random mutagenesis by e ⁇ or-prone PCR, random nucleotide insertion or other methods prior to recombination.
  • one or more components, motifs, sections, parts, domains, fragments, etc., of a polynucleotide encoding a polypeptide of the invention may be recombined with one or more components, motifs, sections, parts, domains, fragments, etc. of one or more heterologous molecules.
  • any of these above fusions can be engineered using the polynucleotides or the polypeptides of the present invention.
  • the present invention also relates to vectors containing the polynucleotide of the present invention, host cells, and the production of polypeptides by synthetic and recombinant techniques.
  • the vector may be, for example, a phage, plasmid, viral, or retroviral vector. Retroviral vectors may be replication competent or replication defective. In the latter case, viral propagation generally will occur only in complementing host cells.
  • the polynucleotides of the invention 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. If the vector is a vims, it may be packaged in vitro using an appropriate packaging cell line and then transduced into host cells.
  • the polynucleotide insert should be operatively linked to an appropriate promoter, such as 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 constmcts 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 constmcts will preferably include a translation initiating codon at the beginning and a termination codon (UAA, UGA or UAG) appropriately positioned at the end of the polypeptide to be translated.
  • the expression vectors will preferably include at least one selectable marker.
  • markers include dihydrofolate reductase, G418, glutamine synthase, or neomycin resistance for eukaryotic cell culture, and tetracycline, kanamycin or ampicillin resistance genes for culturing in E. coli and other bacteria.
  • Representative examples of appropriate hosts include, but are not limited to, bacterial cells, such as E.
  • yeast cells e.g., Saccharomyces cerevisiae or Pichia pastoris (ATCC Accession No. 201178)
  • insect cells such as Drosophila S2 and Spodoptera Sf9 cells
  • animal cells such as CHO, COS, 293, and Bowes melanoma cells
  • plant cells Appropriate culture mediums and conditions for the above-described host cells are known in the art.
  • vectors prefe ⁇ ed for use in bacteria include pQE70, pQE60 and pQE-9, available from QIAGEN, Inc.; pBluescript vectors, Phagescript vectors, pNH8A, pNH16a, pNH18A, pNH46A, available from Stratagene Cloning Systems, Inc.; and ptrc99a, pKK223- 3, pKK233-3, pDR540, pRIT5 available from Pharmacia Biotech, Inc.
  • prefe ⁇ ed eukaryotic vectors are pWLNEO, pSV2CAT, pOG44, pXTl and pSG available from Stratagene; and pSVK3, pBPV, pMSG and pSVL available from Pharmacia.
  • Prefe ⁇ ed expression vectors for use in yeast systems include, but are not limited to pYES2, pYDl, pTEFl/Zeo, pYES2/GS, pPICZ, pGAPZ, pGAPZalph, pPIC9, pPIC3.5, pHEL-D2, pHIL-Sl, pPIC3.5K, pPIC9K, and PAO815 (all available from Invitrogen, Carlbad, CA).
  • Other suitable vectors will be readily apparent to the skilled artisan.
  • Vectors which use glutamine synthase (GS) or DHFR as the selectable markers can be amplified in the presence of the drugs methionine sulphoximine or methotrexate, respectively.
  • An advantage of glutamine synthase based vectors are the availabilty of cell lines (e.g., the murine myeloma cell line, NSO) which are glutamine synthase negative.
  • Glutamine synthase expression systems can also function in glutamine synthase expressing cells (e.g., Chinese Hamster Ovary (CHO) cells) by providing additional inhibitor to prevent the functioning of the endogenous gene.
  • glutamine synthase expression system and components thereof are detailed in PCT publications: WO87/04462; WO86/05807; WO89/01036; WO89/10404; and WO91/06657, which are hereby inco ⁇ orated in their entireties by reference herein. Additionally, glutamine synthase expression vectors can be obtained from Lonza Biologies, Inc. (Portsmouth, NH). Expression and production of monoclonal antibodies using a GS expression system in murine myeloma cells is described in Bebbington et al., Bio/technology 10:169(1992) and in Biblia and Robinson BiotechnoL Prog. 11 :1 (1995) which are herein inco ⁇ orated by reference.
  • the present invention also relates to host cells containing the above-described vector constmcts described herein, and additionally encompasses host cells containing nucleotide sequences of the invention that are operably associated with one or more heterologous control regions (e.g., promoter and/or enhancer) using techniques known of in the art.
  • the host cell can be a higher eukaryotic cell, such as a mammalian cell (e.g., a human derived cell), or a lower eukaryotic cell, such as a yeast cell, or the host cell can be a prokaryotic cell, such as a bacterial cell.
  • a host strain may be chosen which modulates the expression of the inserted gene sequences, or modifies and processes the gene product in the specific fashion desired. Expression from certain promoters can be elevated in the presence of certain inducers; thus expression of the genetically engineered polypeptide may be controlled. Furthermore, different host cells have characteristics and specific mechanisms for the translational and post-translational processing and modification (e.g., phosphorylation, cleavage) of proteins. Appropriate cell lines can be chosen to ensure the desired modifications and processing of the foreign protein expressed.
  • nucleic acids and nucleic acid constmcts of the invention into the host cell can be effected by calcium phosphate transfection, DEAE-dextran mediated transfection, cationic lipid-mediated transfection, electroporation, transduction, infection, or other methods. Such methods are described in many standard laboratory manuals, such as Davis et al., Basic Methods In Molecular Biology (1986). It is specifically contemplated that the polypeptides of the present invention may in fact be expressed by a host cell lacking a recombinant vector.
  • the invention also encompasses primary, secondary, and immortalized host cells of vertebrate origin, particularly mammalian origin, that have been engineered to delete or replace endogenous genetic material (e.g., the coding sequence), and/or to include genetic material (e.g., heterologous polynucleotide sequences) that is operably associated with polynucleotides of the invention, and which activates, alters, and/or amplifies endogenous polynucleotides.
  • endogenous genetic material e.g., the coding sequence
  • genetic material e.g., heterologous polynucleotide sequences
  • heterologous control regions e.g., promoter and/or enhancer
  • endogenous polynucleotide sequences via homologous recombination
  • heterologous control regions e.g., promoter and/or enhancer
  • endogenous polynucleotide sequences via homologous recombination
  • Polypeptides 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 and lectin chromatography. Most preferably, high performance liquid chromatography (“HPLC”) is employed for purification.
  • HPLC high performance liquid chromatography
  • Polypeptides of the present invention can also be recovered from: 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.
  • 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.
  • the yeast Pichia pastoris is used to express polypeptides of the invention in a eukaryotic system. Pichia pastoris is a methylotrophic yeast which can metabolize methanol as its sole carbon source.
  • a main step in the methanol metabolization pathway is the oxidation of methanol to formaldehyde using O 2 .
  • This reaction is catalyzed by the enzyme alcohol oxidase.
  • Pichia pastoris In order to metabolize methanol as its sole carbon source, Pichia pastoris must generate high levels of alcohol oxidase due, in part, to the relatively low affinity of alcohol oxidase for O 2 . Consequently, in a growth medium depending on methanol as a main carbon source, the promoter region of one of the two alcohol oxidase genes (AOX1) is highly active. In the presence of methanol, alcohol oxidase produced from the AOX1 gene comprises up to approximately 30% of the total soluble protein in Pichia pastoris.
  • a heterologous coding sequence such as, for example, a polynucleotide of the present invention, under the transcriptional regulation of all or part of the AOX1 regulatory sequence is expressed at exceptionally high levels in Pichia yeast grown in the presence of methanol.
  • the plasmid vector pPIC9K is used to express DNA encoding a polypeptide of the invention, as set forth herein, in a Pichea yeast system essentially as described in "Pichia Protocols: Methods in Molecular Biology," D.R. Higgins and J. Cregg, eds. The Humana Press, Totowa, NJ, 1998.
  • This expression vector allows expression and secretion of a polypeptide of the invention by virtue of the strong AOX1 promoter linked to the Pichia pastoris alkaline phosphatase (PHO) secretory signal peptide (i.e., leader) located upstream of a multiple cloning site.
  • PHO alkaline phosphatase
  • yeast vectors could be used in place of pPIC9K, such as, pYES2, pYDl, pTEFl/Zeo, pYES2/GS, pPICZ, pGAPZ, pGAPZalpha, pPIC9, pPIC3.5, pHIL-D2, pHEL-Sl, pPIC3.5K, and PAO815, as one skilled in the art would readily appreciate, as long as the proposed expression constmct provides appropriately located signals for transcription, translation, secretion (if desired), and the like, including an in- frame AUG as required.
  • high-level expression of a heterologous coding sequence such as, for example, a polynucleotide of the present invention
  • a heterologous coding sequence such as, for example, a polynucleotide of the present invention
  • an expression vector such as, for example, pGAPZ or pGAPZalpha
  • the invention also encompasses primary, secondary, and immortalized host cells of vertebrate origin, particularly mammalian origin, that have been engineered to delete or replace endogenous genetic material (e.g., coding sequence), and/or to include genetic material (e.g., heterologous polynucleotide sequences) that is operably associated with polynucleotides of the invention, and which activates, alters, and/or amplifies endogenous polynucleotides.
  • endogenous genetic material e.g., coding sequence
  • genetic material e.g., heterologous polynucleotide sequences
  • heterologous control regions e.g., promoter and/or enhancer
  • endogenous polynucleotide sequences via homologous recombination
  • heterologous control regions e.g., promoter and/or enhancer
  • endogenous polynucleotide sequences via homologous recombination
  • polypeptides of the invention can be chemically synthesized using techniques known in the art (e.g., see Creighton, 1983, Proteins: Stmctures and Molecular Principles, W.H. Freeman & Co., N.Y., and Hunkapiller et al., Nature, 310:105-111 (1984)).
  • a polypeptide co ⁇ esponding to a fragment of a polypeptide can be synthesized by use of a peptide synthesizer.
  • nonclassical amino acids or chemical amino acid analogs can be introduced as a substitution or addition into the polypeptide sequence.
  • Non-classical amino acids include, but are not limited to, to the D- isomers of the common amino acids, 2,4-diaminobutyric acid, a-amino isobutyric acid, 4- aminobutyric acid, Abu, 2-amino butyric acid, g-Abu, e-Ahx, 6-amino hexanoic acid, Aib, 2-amino isobutyric acid, 3-amino propionic acid, omithine, norleucine, norvaline, hydroxyproline, sarcosine, citmlline, homocitrulline, cysteic acid, t-butylglycine, t- butylalanine, phenylglycine, cyclohexylalanine, b-alanine, fluoro-amino acids, designer amino acids such as b-methyl amino acids, Ca-methyl amino acids, Na-methyl amino acids, and amino acid analogs in general. Furthermore, the amino acid can
  • the invention encompasses polypeptides of the present invention which are differentially modified during or after translation, e.g., by glycosylation, acetylation, phosphorylation, amidation, derivatization by known protecting/blocking groups, proteolytic cleavage, linkage to an antibody molecule or other cellular ligand, etc. Any of numerous chemical modifications may be carried out by known techniques, including but not limited, to Specific chemical cleavage by cyanogen bromide, trypsin, chymotrypsin, papain, V8 protease, NaBH 4 ; acetylation, formylation, oxidation, reduction; metabolic synthesis in the presence of tunicamycin; etc.
  • 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 procaryotic 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.
  • suitable enzymes include horseradish peroxidase, alkaline phosphatase, beta-galactosidase, or acetylcholinesterase; examples of suitable prosthetic group complexes include streptavidin/biotin and avidin/biotin; examples of suitable fluorescent materials include umbelliferone, fluorescein, fluorescein isothiocyanate, rhodamine, dichlorotriazinylamine fluorescein, dansyl chloride or phycoerythrin; an example of a luminescent material includes luminol; examples of bioluminescent materials include luciferase, luciferin, and aequorin; and examples of suitable radioactive material include iodine ( 12, I, l23 I, 125 I, 1 1 I), carbon ( 14 C), sulfur ( 35 S), tritium ( 3 H), indium ( In, 112 In, , 13m In, 115m In), technetium ( 99 Tc,
  • a polypeptide of the present invention or fragment or variant thereof is attached to macrocyclic chelators that associate with radiometal ions, including but not limited to, 177 Lu, 90 Y, 166 Ho, and 153 Sm, to polypeptides.
  • the radiometal ion associated with the macrocyclic chelators is In.
  • the radiometal ion associated with the macrocyclic chelator is 90 Y.
  • the macrocyclic chelator is 1, 4,7,10-tetraazacyclododecane- N,N',N",N'"-tetraacetic acid (DOTA).
  • DOTA is attached to an antibody of the invention or fragment thereof via a linker molecule.
  • linker molecules useful for conjugating DOTA to a polypeptide are commonly known in the art - see, for example, DeNardo et al., Clin Cancer Res. 4(10):2483-90 (1998); Peterson et al., Bioconjug. Chem. 10(4):553-7 (1999); and Zimmerman et al, Nucl. Med. Biol. 26(8):943-50 (1999); which are hereby inco ⁇ orated by reference in their entirety.
  • the proteins of the invention may be modified by either natural processes, such as posttranslational processing, or by chemical modification techniques which are well known in the art. It will be appreciated that the same type of modification may be present in the same or varying degrees at several sites in a given polypeptide.
  • Polypeptides of the invention may be branched, for example, as a result of ubiquitination, and they may be cyclic, with or without branching. Cyclic, branched, and branched cyclic polypeptides may result from posttranslation natural processes or may be made by synthetic methods.
  • Modifications include acetylation, acylation, ADP-ribosylation, amidation, covalent attachment of flavin, covalent attachment of a heme moiety, covalent attachment of a nucleotide or nucleotide derivative, covalent attachment of a lipid or lipid derivative, covalent attachment of phosphotidylinositol, cross-linking, cyclization, disulfide bond formation, demethylation, formation of covalent cross-links, formation of cysteine, formation of pyroglutamate, formylation, gamma-carboxylation, glycosylation, GPI anchor formation, hydroxylation, iodination, methylation, myristoylation, oxidation, pegylation, proteolytic processing, phosphorylation, prenylation, racemization, selenoylation, sulfation, transfer- RNA mediated addition of amino acids to proteins such as arginylation, and ubiquitination.
  • the chemical moieties for derivitization may be selected from water soluble polymers such as polyethylene glycol, ethylene glycol/propylene glycol copolymers, carboxymethylcellulose, dextran, polyvinyl alcohol and the like.
  • the polypeptides may be modified at random positions within the molecule, or at predetermined positions within the molecule and may include one, two, three or more attached chemical moieties.
  • the polymer may be of any molecular weight, and may be branched or unbranched.
  • the prefe ⁇ ed molecular weight is between about 1 kDa and about 100 kDa (the term "about” indicating that in preparations of polyethylene glycol, some molecules will weigh more, some less, than the stated molecular weight) for ease in handling and manufacturing.
  • Other sizes may be used, depending on the desired therapeutic profile (e.g., the duration of sustained release desired, the effects, if any on biological activity, the ease in handling, the degree or lack of antigenicity and other known effects of the polyethylene glycol to a therapeutic protein or analog).
  • the polyethylene glycol may have an average molecular weight of about 200, 500, 1000, 1500, 2000, 2500, 3000, 3500, 4000, 4500, 5000, 5500, 6000, 6500, 7000, 7500, 8000, 8500, 9000, 9500, 10,000, 10,500, 1 1,000, 11,500, 12,000, 12,500, 13,000, 13,500, 14,000, 14,500, 15,000, 15,500, 16,000, 16,500, 17,000, 17,500, 18,000, 18,500, 19,000, 19,500, 20,000, 25,000, 30,000, 35,000, 40,000, 45,000, 50,000, 55,000, 60,000, 65,000, 70,000, 75,000, 80,000, 85,000, 90,000, 95,000, or 100,000 kDa.
  • the polyethylene glycol may have a branched stmcture.
  • Branched polyethylene glycols are described, for example, in U.S. Patent No. 5,643,575; Mo ⁇ urgo et al., Appl. Biochem. BiotechnoL 56:59-72 (1996); Vorobjev et al., Nucleosides Nucleotides 18:2145-2150 (1999); and Caliceti et al., Bioconjug. Chem. 70:638-646 (1999), the disclosures of each of which are inco ⁇ orated herein by reference.
  • polyethylene glycol molecules should be attached to the protein with consideration of effects on functional or antigenic domains of the protein.
  • attachment methods available to those skilled in the art, such as, for example, the method disclosed in EP 0 401 384 (coupling PEG to G-CSF), herein inco ⁇ orated by reference; see also Malik et al., Exp. Hematol. 20:1028-1035 (1992), reporting pegylation of GM-CSF using tresyl chloride.
  • polyethylene glycol may be covalently bound through amino acid residues via a reactive group, such as a free amino or carboxyl group.
  • Reactive groups are those to which an activated polyethylene glycol molecule may be bound.
  • the amino acid residues having a free amino group may include lysine residues and the N-terminal amino acid residues; those having a free carboxyl group may include aspartic acid residues glutamic acid residues and the C-terminal amino acid residue.
  • Sulfhydryl groups may also be used as a reactive group for attaching the polyethylene glycol molecules. Prefe ⁇ ed for therapeutic pu ⁇ oses is attachment at an amino group, such as attachment at the N-terminus or lysine group.
  • polyethylene glycol may be attached to proteins via linkage to any of a number of amino acid residues.
  • polyethylene glycol can be linked to proteins via covalent bonds to lysine, histidine, aspartic acid, glutamic acid, or cysteine residues.
  • One or more reaction chemistries may be employed to attach polyethylene glycol to specific amino acid residues (e.g., lysine, histidine, aspartic acid, glutamic acid, or cysteine) of the protein or to more than one type of amino acid residue (e.g., lysine, histidine, aspartic acid, glutamic acid, cysteine and combinations thereof) of the protein.
  • specific amino acid residues e.g., lysine, histidine, aspartic acid, glutamic acid, or cysteine
  • polyethylene glycol as an illustration of the present composition, one may select from a variety of polyethylene glycol molecules (by molecular weight, branching, etc.), the proportion of polyethylene glycol molecules to protein (polypeptide) molecules in the reaction mix, the type of pegylation reaction to be performed, and the method of obtaining the selected N-terminally pegylated protein.
  • the method of obtaining the N-terminally pegylated preparation i.e., separating this moiety from other monopegylated moieties if necessary
  • Selective proteins chemically modified at the N-terminus modification may be accomplished by reductive alkylation which exploits differential reactivity of different types of primary amino groups (lysine versus the N-terminal) available for derivatization in a particular protein. Under the appropriate reaction conditions, substantially selective derivatization of the protein at the N-terminus with a carbonyl group containing polymer is achieved.
  • pegylation of the proteins of the invention may be accomplished by any number of means.
  • polyethylene glycol may be attached to the protein either directly or by an intervening linker.
  • Linkerless systems for attaching polyethylene glycol to proteins are described in Delgado et al., Crit. Rev. Thera. Dmg Carrier Sys. 9:249-304 (1992); Francis et al., Intern. J. of Hematol. 68:1-18 (1998); U.S. Patent No. 4,002,531; U.S. Patent No. 5,349,052; WO 95/06058; and WO 98/32466, the disclosures of each of which are inco ⁇ orated herein by reference.
  • One system for attaching polyethylene glycol directly to amino acid residues of proteins without an intervening linker employs tresylated MPEG, which is produced by the modification of monmethoxy polyethylene glycol (MPEG) using tresylchloride (ClSO 2 CH CF 3 ).
  • MPEG monmethoxy polyethylene glycol
  • ClSO 2 CH CF 3 tresylchloride
  • polyethylene glycol is directly attached to amine groups of the protein.
  • the invention includes protein- polyethylene glycol conjugates produced by reacting proteins of the invention with a polyethylene glycol molecule having a 2,2,2-trifluoreothane sulphonyl group.
  • Polyethylene glycol can also be attached to proteins using a number of different intervening linkers. For example, U.S.
  • Patent No. 5,612,460 discloses urethane linkers for connecting polyethylene glycol to proteins.
  • Protein-polyethylene glycol conjugates wherein the polyethylene glycol is attached to the protein by a linker can also be produced by reaction of proteins with compounds such as MPEG-succinimidylsuccinate, MPEG activated with 1 , 1 '-carbonyldiimidazole, MPEG-2,4,5-trichloropenylcarbonate, MPEG-p- nitrophenolcarbonate, and various MPEG-succinate derivatives.
  • MPEG-succinimidylsuccinate MPEG activated with 1 , 1 '-carbonyldiimidazole
  • MPEG-2,4,5-trichloropenylcarbonate MPEG-p- nitrophenolcarbonate
  • various MPEG-succinate derivatives A number of additional polyethylene glycol derivatives and reaction chemistries for attaching polyethylene glycol to proteins are described in International Publication No.
  • WO 98/32466 the entire disclosure of which is inco ⁇ orated herein by reference.
  • Pegylated protein products produced using the reaction chemistries set out herein are included within the scope of the invention.
  • the number of polyethylene glycol moieties attached to each protein of the invention may also vary.
  • the pegylated proteins of the invention may be linked, on average, to 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 12, 15, 17, 20, or more polyethylene glycol molecules.
  • the average degree of substitution within ranges such as 1-3, 2-4, 3-5, 4-6, 5-7, 6-8, 7-9, 8-10, 9-11, 10-12, 11-13, 12-14, 13-15, 14-16, 15-17, 16-18, 17-19, or 18-20 polyethylene glycol moieties per protein molecule.
  • Methods for determining the degree of substitution are discussed, for example, in Delgado et al., Crit. Rev. Thera. Dmg Carrier Sys. 9:249-304 (1992).
  • polypeptides of the invention can be recovered and purified from chemical synthesis and recombinant cell cultures by standard methods which include, but are not limited to, ammonium sulfate or ethanol precipitation, acid extraction, anion or cation exchange chromatography, phosphocellulose chromatography, hydrophobic interaction chromatography, affinity chromatography, hydroxylapatite chromatography and lectin chromatography. Most preferably, high performance liquid chromatography (“HPLC”) is employed for purification. Well known techniques for refolding protein may be employed to regenerate active conformation when the polypeptide is denatured during isolation and/or purification.
  • HPLC high performance liquid chromatography
  • the polypeptides of the invention may be in monomers or multimers (i.e., dimers, trimers, tetramers and higher multimers). Accordingly, the present invention relates to monomers and multimers of the polypeptides of the invention, their preparation, and Compositions (preferably, Therapeutics) containing them.
  • the polypeptides of the invention are monomers, dimers, trimers or tetramers.
  • the multimers of the invention are at least dimers, at least trimers, or at least tetramers.
  • Multimers encompassed by the invention may be homomers or heteromers.
  • the term homomer refers to a multimer containing only polypeptides co ⁇ esponding to a protein of the invention (e.g., the amino acid sequence of SEQ YD NO:Y, an amino acid sequence encoded by SEQ ID NO:X or the complement of SEQ ED NO:X, the amino acid sequence encoded by the portion of SEQ YD NO:X as defined in columns 8 and 9 of Table 2, and/or an amino acid sequence encoded by cDNA contained in Clone ED NO:Z (including fragments, variants, splice variants, and fusion proteins, co ⁇ esponding to these as described herein)).
  • homomers may contain polypeptides having identical or different amino acid sequences.
  • a homomer of the invention is a multimer containing only polypeptides having an identical amino acid sequence.
  • a homomer of the invention is a multimer containing polypeptides having different amino acid sequences.
  • the multimer of the invention is a homodimer (e.g., containing two polypeptides having identical or different amino acid sequences) or a homotrimer (e.g., containing three polypeptides having identical and/or different amino acid sequences).
  • the homomeric multimer of the invention is at least a homodimer, at least a homotrimer, or at least a homotetramer.
  • heteromer refers to a multimer containing one or more heterologous polypeptides (i.e., polypeptides of different proteins) in addition to the polypeptides of the invention.
  • the multimer of the invention is a heterodimer, a heterotrimer, or a heterotetramer.
  • the heteromeric multimer of the invention is at least a heterodimer, at least a heterotrimer, or at least a heterotetramer.
  • Multimers of the invention may be the result of hydrophobic, hydrophilic, ionic and/or covalent associations and/or may be indirectly linked by, for example, liposome formation.
  • multimers of the invention such as, for example, homodimers or homotrimers
  • heteromultimers of the invention such as, for example, heterotrimers or heterotetramers, are formed when polypeptides of the invention contact antibodies to the polypeptides of the invention (including antibodies to the heterologous polypeptide sequence in a fusion protein of the invention) in solution.
  • multimers of the invention are formed by covalent associations with and/or between the polypeptides of the invention.
  • covalent associations may involve one or more amino acid residues contained in the polypeptide sequence (e.g., that recited in SEQ ID NO:Y, encoded by the portion of SEQ YD NO:X as defined in columns 8 and 9 of Table 2, and/or encoded by the cDNA contained in Clone ED NO:Z).
  • the covalent associations are cross-linking between cysteine residues located within the polypeptide sequences which interact in the native (i.e., naturally occurring) polypeptide.
  • the covalent associations are the consequence of chemical or recombinant manipulation.
  • covalent associations may involve one or more amino acid residues contained in the heterologous polypeptide sequence in a fusion protein.
  • covalent associations are between the heterologous sequence contained in a fusion protein of the invention (see, e.g., US Patent Number 5,478,925).
  • the covalent associations are between the heterologous sequence contained in a Fc fusion protein of the invention (as described herein).
  • covalent associations of fusion proteins of the invention are between heterologous polypeptide sequence from another protein that is capable of forming covalently associated multimers, such as for example, osteoprotegerin (see, e.g., International Publication NO: WO 98/49305, the contents of which are herein inco ⁇ orated by reference in its entirety).
  • two or more polypeptides of the invention are joined through peptide linkers. Examples include those peptide linkers described in U.S. Pat. No. 5,073,627 (hereby inco ⁇ orated by reference). Proteins comprising multiple polypeptides of the invention separated by peptide linkers may be produced using conventional recombinant DNA technology.
  • Leucine zipper and isoleucine zipper domains are polypeptides that promote multimerization of the proteins in which they are found.
  • Leucine zippers were originally identified in several DNA-binding proteins (Landschulz et al., Science 240:1759, (1988)), and have since been found in a variety of different proteins.
  • Leucine zippers are naturally occu ⁇ ing peptides and derivatives thereof that dimerize or trimerize.
  • leucine zipper domains suitable for producing soluble multimeric proteins of the invention are those described in PCT application WO 94/10308, hereby inco ⁇ orated by reference.
  • Recombinant fusion proteins comprising a polypeptide of the invention fused to a polypeptide sequence that dimerizes or trimerizes in solution are expressed in suitable host cells, and the resulting soluble multimeric fusion protein is recovered from the culture supernatant using techniques known in the art.
  • Trimeric polypeptides of the invention may offer the advantage of enhanced biological activity.
  • Preferred leucine zipper moieties and isoleucine moieties are those that preferentially form trimers.
  • One example is a leucine zipper derived from lung surfactant protein D (SPD), as descnbed in Hoppe et al. (FEBS Letters 344:191, (1994)) and in U.S. patent application Ser. No. 08/446,922, hereby inco ⁇ orated by reference.
  • Other peptides derived from naturally occurring trimeric proteins may be employed in preparing trimeric polypeptides of the invention.
  • proteins of the invention are associated by interactions between Flag® polypeptide sequence contained in fusion proteins of the invention containing Flag® polypeptide sequence.
  • proteins of the invention are associated by interactions between heterologous polypeptide sequence contained in Flag® fusion proteins of the invention and anti -Flag® antibody.
  • the multimers of the invention may be generated using chemical techniques known in the art.
  • polypeptides desired to be contained in the multimers of the invention may be chemically cross-linked using linker molecules and linker molecule length optimization techniques known in the art (see, e.g., US Patent Number 5,478,925, which is herein inco ⁇ orated by reference in its entirety).
  • linker molecules and linker molecule length optimization techniques known in the art
  • multimers of the invention may be generated using techniques known in the art to form one or more inter-molecule cross-links between the cysteine residues located within the sequence of the polypeptides desired to be contained in the multimer (see, e.g., US Patent Number 5,478,925, which is herein inco ⁇ orated by reference in its entirety).
  • polypeptides of the invention may be routinely modified by the addition of cysteine or biotin to the C-terminus or N-terminus of the polypeptide and techniques known in the art may be applied to generate multimers containing one or more of these modified polypeptides (see, e.g., US Patent Number 5,478,925, which is herein inco ⁇ orated by reference in its entirety). Additionally, techniques known in the art may be applied to generate liposomes containing the polypeptide components desired to be contained in the multimer of the invention (see, e.g., US Patent Number 5,478,925, which is herein inco ⁇ orated by reference in its entirety). [205] Alternatively, multimers of the invention may be generated using genetic engineering techniques known in the art.
  • polypeptides contained in multimers of the invention are produced recombinantly using fusion protein technology described herein or otherwise known in the art (see, e.g., US Patent Number 5,478,925, which is herein inco ⁇ orated by reference in its entirety).
  • polynucleotides coding for a homodimer of the invention are generated by ligating a polynucleotide sequence encoding a polypeptide of the invention to a sequence encoding a linker polypeptide and then further to a synthetic polynucleotide encoding the translated product of the polypeptide in the reverse orientation from the original C-terminus to the N- terminus (lacking the leader sequence) (see, e.g., US Patent Number 5,478,925, which is herein inco ⁇ orated by reference in its entirety).
  • recombinant techniques described herein or otherwise known in the art are applied to generate recombinant polypeptides of the invention which contain a transmembrane domain (or hydrophobic or signal peptide) and which can be inco ⁇ orated by membrane reconstitution techniques into liposomes (see, e.g., US Patent Number 5,478,925, which is herein inco ⁇ orated by reference in its entirety).
  • polypeptides of the invention relate to antibodies and T-cell antigen receptors (TCR) which immunospecifically bind a polypeptide, polypeptide fragment, or variant of the invention (e.g., a polypeptide or fragment or variant of the amino acid sequence of SEQ ED NO:Y or a polypeptide encoded by the cDNA contained in Clone ED No:Z, and/or an epitope, of the present invention) as determined by immunoassays well known in the art for assaying specific antibody-antigen binding.
  • TCR T-cell antigen receptors
  • Antibodies of the invention include, but are not limited to, polyclonal, monoclonal, multispecific, human, humanized or chimeric antibodies, single chain antibodies, Fab fragments, F(ab') fragments, fragments produced by a Fab expression library, anti-idiotypic (anti-Id) antibodies (including, e.g., anti-Id antibodies to antibodies of the invention), intracellularly-made antibodies (i.e., intrabodies), and epitope-binding fragments of any of the above.
  • antibody refers to immunoglobulin molecules and immunologically active portions of immunoglobulin molecules, i.e., molecules that contain an antigen binding site that immunospecifically binds an antigen.
  • the immunoglobulin molecules of the invention can be of any type (e.g., IgG, IgE, IgM, IgD, IgA and IgY), class (e.g., IgGl, IgG2, IgG3, IgG4, IgAl and IgA2) or subclass of immunoglobulin molecule.
  • the immunoglobulin molecules of the invention are IgGl .
  • the immunoglobulin molecules of the invention are IgG4.
  • the antibodies are human antigen-binding antibody fragments of the present invention and include, but are not limited to, Fab, Fab' and F(ab')2, Fd, single- chain Fvs (scFv), single-chain antibodies, disulfide-linked Fvs (sdFv) and fragments comprising either a VL or VH domain.
  • Antigen-binding antibody fragments, including single-chain antibodies may comprise the variable region(s) alone or in combination with the entirety or a portion of the following: hinge region, CHI, CH2, and CH3 domains. Also included in the invention are antigen-binding fragments also comprising any combination of variable region(s) with a hinge region, CHI, CH2, and CH3 domains.
  • the antibodies of the invention may be from any animal origin including birds and mammals.
  • the antibodies are human, murine (e.g., mouse and rat), donkey, ship rabbit, goat, guinea pig, camel, horse, or chicken.
  • "human” antibodies include antibodies having the amino acid sequence of a human immunoglobulin and include antibodies isolated from human immunoglobulin libraries or from animals transgenic for one or more human immunoglobulin and that do not express endogenous immunoglobulins, as described infra and, for example in, U.S. Patent No. 5,939,598 by Kucherlapati et al.
  • the antibodies of the present invention may be monospecific, bispecific, trispecific or of greater multispecificity. Multispecific antibodies may be specific for different epitopes of a polypeptide of the present invention or may be specific for both a polypeptide of the present invention as well as for a heterologous epitope, such as a heterologous polypeptide or solid support material. See, e.g., PCT publications WO 93/17715; WO 92/08802; WO 91/00360; WO 92/05793; Tutt, et al., J. Immunol. 147:60-69 (1991); U.S. Patent Nos. 4,474,893; 4,714,681 ; 4,925,648; 5,573,920; 5,601,819; Kostelny et al., J. Immunol. 148:1547-1553 (1992).
  • Antibodies of the present invention may be described or specified in terms of the epitope(s) or portion(s) of a polypeptide of the present invention which they recognize or specifically bind.
  • the epitope(s) or polypeptide portion(s) may be specified as described herein, e.g., by N-terminal and C-terminal positions, or by size in contiguous amino acid residues, or listed in the Tables and Figures.
  • Prefe ⁇ ed epitopes of the invention include the predicted epitopes shown in column 7 of Table 1A, as well as polynucleotides that encode these epitopes.
  • Antibodies which specifically bind any epitope or polypeptide of the present invention may also be excluded. Therefore, the present invention includes antibodies that Specifically bind polypeptides of the present invention, and allows for the exclusion of the same.
  • Antibodies of the present invention may also be described or specified in terms of their cross-reactivity. Antibodies that do not bind any other analog, ortholog, or homolog of a polypeptide of the present invention are included. Antibodies that bind polypeptides with at least 95%, at least 90%, at least 85%, at least 80%, at least 75%, at least 70%, at least 65%, at least 60%, at least 55%, and at least 50% identity (as calculated using methods known in the art and described herein) to a polypeptide of the present invention are also included in the present invention. In specific embodiments, antibodies of the present invention cross-react with murine, rat and/or rabbit homologs of human proteins and the co ⁇ esponding epitopes thereof.
  • Antibodies that do not bind polypeptides with less than 95%, less than 90%, less than 85%, less than 80%, less than 75%, less than 70%, less than 65%, less than 60%, less than 55%, and less than 50% identity (as calculated using methods known in the art and described herein) to a polypeptide of the present invention are also included in the present invention.
  • the above-described cross-reactivity is with respect to any single specific antigenic or immunogenic polypeptide, or combination(s) of 2, 3, 4, 5, or more of the specific antigenic and/or immunogenic polypeptides disclosed herein.
  • antibodies which bind polypeptides encoded by polynucleotides which hybridize to a polynucleotide of the present invention under stringent hybridization conditions are also included in the present invention.
  • Preferred binding affinities include those with a dissociation constant or Kd less than 5 X 10 " M, 10 " M, 5 X IO “3 M, IO “3 M, 5 X 10 "4 M, 10 “4 M, 5 X IO “5 M, 10 “5 M, 5 X IO “6 M, 10 “6 M, 5 X 10 "7 M, IO 7 M, 5 X 10 "8 M, IO “8 M, 5 X IO “9 M, IO "9 M, 5 X 10 "10 M, 10 “10 M, 5 X 10 "11 M, 10 "11 M, 5 X IO “12 M, IO “12 M, 5 X 10 "13 M, 10 “13 M, 5 X IO “14 M, IO “14 M, 5 X 10 “15 M, or 10 '15 M.
  • the invention also provides antibodies that competitively inhibit binding of an antibody to an epitope of the invention as determined by any method known in the art for determining competitive binding, for example, the immunoassays described herein. In prefe ⁇ ed embodiments, the antibody competitively inhibits binding to the epitope by at least 95%, at least 90%, at least 85 %, at least 80%, at least 75%, at least 70%, at least 60%, or at least 50%.
  • Antibodies of the present invention may act as agonists or antagonists of the polypeptides of the present invention.
  • the present invention includes antibodies which dismpt the receptor/Iigand interactions with the polypeptides of the invention either partially or fully.
  • antibodies of the present invention bind an antigenic epitope disclosed herein, or a portion thereof.
  • the invention features both receptor-specific antibodies and ligand-specific antibodies.
  • the invention also features receptor-specific antibodies which do not prevent ligand binding but prevent receptor activation.
  • Receptor activation i.e., signaling
  • receptor activation can be determined by techniques described herein or otherwise known in the art. For example, receptor activation can be determined by detecting the phosphorylation (e.g., tyrosine or serine/threonine) of the receptor or its substrate by immunoprecipitation followed by western blot analysis (for example, as described supra).
  • antibodies are provided that inhibit ligand activity or receptor activity by at least 95%, at least 90%, at least 85%, at least 80%, at least 75%, at least 70%, at least 60%, or at least 50% of the activity in absence of the antibody.
  • the invention also features receptor-specific antibodies which both prevent ligand binding and receptor activation as well as antibodies that recognize the receptor-ligand complex, and, preferably, do not specifically recognize the unbound receptor or the unbound ligand.
  • receptor-specific antibodies which both prevent ligand binding and receptor activation as well as antibodies that recognize the receptor-ligand complex, and, preferably, do not specifically recognize the unbound receptor or the unbound ligand.
  • neutralizing antibodies which bind the ligand and prevent binding of the ligand to the receptor, as well as antibodies which bind the ligand, thereby preventing receptor activation, but do not prevent the ligand from binding the receptor.
  • antibodies which activate the receptor are also act as receptor agonists, i.e., potentiate or activate either all or a subset of the biological activities of the ligand-mediated receptor activation, for example, by inducing dimerization of the receptor.
  • the antibodies may be specified as agonists, antagonists or inverse agonists for biological activities comprising the specific biological activities of the peptides of the invention disclosed herein.
  • the above antibody agonists can be made using methods known in the art. See, e.g., PCT publication WO 96/40281; U.S. Patent No. 5,811,097; Deng et al., Blood 92(6):1981-1988 (1998); Chen et al., Cancer Res. 58(16):3668-3678 (1998); Ha ⁇ op et al., J. Immunol. 161(4):1786-1794 (1998); Zhu et al., Cancer Res.
  • Antibodies of the present invention may be used, for example, to purify, detect, and target the polypeptides of the present invention, including both in vitro and in vivo diagnostic and therapeutic methods.
  • the antibodies have utility in immunoassays for qualitatively and quantitatively measuring levels of the polypeptides of the present invention in biological samples. See, e.g., Harlow et al., Antibodies: A Laboratory Manual, (Cold Spring Harbor Laboratory Press, 2nd ed. 1988); inco ⁇ orated by reference herein in its entirety. .
  • the antibodies of the present invention may be used either alone or in combination with other compositions.
  • the antibodies may further be recombinantly fused to a heterologous polypeptide at the N- or C-terminus or chemically conjugated (including covalent and non-covalent conjugations) to polypeptides or other compositions.
  • antibodies of the present invention may be recombinantly fused or conjugated to molecules useful as labels in detection assays and effector molecules such as heterologous polypeptides, dmgs, radionuclides, or toxins. See, e.g., PCT publications WO 92/08495; WO 91/14438; WO 89/12624; U.S. Patent No.
  • the antibodies of the invention include derivatives that are modified, i.e, by the covalent attachment of any type of molecule to the antibody such that covalent attachment does not prevent the antibody from generating an anti-idiotypic response.
  • the antibody derivatives include antibodies that have been modified, e.g., by glycosylation, acetylation, pegylation, phosphylation, amidation, derivatization by known protecting/blocking groups, proteolytic cleavage, linkage to a cellular ligand or other protein, etc. Any of numerous chemical modifications may be carried out by known techniques, including, but not limited to specific chemical cleavage, acetylation, formylation, metabolic synthesis of tunicamycin, etc. Additionally, the derivative may contain one or more non-classical amino acids.
  • the antibodies of the present invention may be generated by any suitable method known in the art.
  • Polyclonal antibodies to an antigen-of- interest can be produced by various procedures well known in the art.
  • a polypeptide of the invention can be administered to various host animals including, but not limited to, rabbits, mice, rats, etc. to induce the production of sera containing polyclonal antibodies specific for the antigen.
  • adjuvants may be used to increase the immunological response, depending on the host species, and include but are not limited to, Freund's (complete and incomplete), mineral gels such as aluminum hydroxide, surface active substances such as lysolecithin, pluronic polyols, polyanions, peptides, oil emulsions, keyhole limpet hemocyanins, dinitrophenol, and potentially useful human adjuvants such as BCG (bacille Calmette-Guerin) and corynebacterium parvum. Such adjuvants are also well known in the art.
  • Monoclonal antibodies can be prepared using a wide variety of techniques known in the art including the use of hybridoma, recombinant, and phage display technologies, or a combination thereof.
  • monoclonal antibodies can be produced using hybridoma techniques including those known in the art and taught, for example, in Harlow et al., Antibodies: A Laboratory Manual, (Cold Spring Harbor Laboratory Press, 2nd ed. 1988); Hammerling, et al., in: Monoclonal Antibodies and T-Cell Hybridomas 563-681 (Elsevier, N.Y., 1981) (said references inco ⁇ orated by reference in their entireties).
  • the term "monoclonal antibody' * as used herein is not limited to antibodies produced through hybridoma technology.
  • the term “monoclonal antibody” refers to an antibody that is derived from a single clone, including any eukaryotic, prokaryotic, or phage clone, and not the method by which it is produced.
  • mice can be immunized with a polypeptide of the invention or a cell expressing such peptide.
  • an immune response e.g., antibodies specific for the antigen are detected in the mouse semm
  • the mouse spleen is harvested and splenocytes isolated.
  • the splenocytes are then fused by well known techniques to any suitable myeloma cells, for example cells from cell line SP20 available from the ATCC.
  • Hybridomas are selected and cloned by limited dilution.
  • hybridoma clones are then assayed by methods known in the art for cells that secrete antibodies capable of binding a polypeptide of the invention.
  • Ascites fluid which generally contains high levels of antibodies, can be generated by immunizing mice with positive hybridoma clones.
  • the present invention provides methods of generating monoclonal antibodies as well as antibodies produced by the method comprising culturing a hybridoma cell secreting an antibody of the invention wherein, preferably, the hybridoma is generated by fusing splenocytes isolated from a mouse immunized with an antigen of the invention with myeloma cells and then screening the hybridomas resulting from the fusion for hybridoma Clones that secrete an antibody able to bind a polypeptide of the invention.
  • Another well known method for producing both polyclonal and monoclonal human B cell lines is transformation using Epstein Ban Vims (EBV).
  • Protocols for generating EBV-transformed B cell lines are commonly known in the art, such as, for example, the protocol outlined in Chapter 7.22 of Cu ⁇ ent Protocols in Immunology, Coligan et al., Eds., 1994, John Wiley & Sons, NY, which is hereby inco ⁇ orated in its entirety by reference.
  • the source of B cells for transformation is commonly human peripheral blood, but B cells for transformation may also be derived from other sources including, but not limited to, lymph nodes, tonsil, spleen, tumor tissue, and infected tissues. Tissues are generally made into single cell suspensions prior to EBV transformation. Additionally, steps may be taken to either physically remove or inactivate T cells (e.g., by treatment with cyclosporin A) in B cell-containing samples, because T cells from individuals seropositive for anti-EBV antibodies can suppress B cell immortalization by EBV.
  • EBV lines are generally polyclonal. However, over prolonged periods of cell cultures, EBV lines may become monoclonal or polyclonal as a result of the selective outgrowth of particular B cell clones.
  • polyclonal EBV transformed lines may be subcloned (e.g., by limiting dilution culture) or fused with a suitable fusion partner and plated at limiting dilution to obtain monoclonal B cell lines.
  • suitable fusion partners for EBV transformed cell lines include mouse myeloma cell lines (e.g., SP2/0, X63-Ag8.653), heteromyeloma cell lines (human x mouse; e.g, SPAM-8, SBC-H20, and CB-F7), and human cell lines (e.g., GM 1500, SKO-007, RPMI 8226, and KR-4).
  • the present invention also provides a method of generating polyclonal or monoclonal human antibodies against polypeptides of the invention or fragments thereof, comprising EBV-transformation of human B cells.
  • Antibody fragments which recognize specific epitopes may be generated by known techniques.
  • Fab and F(ab')2 fragments of the invention may be produced by proteolytic cleavage of immunoglobulin molecules, using enzymes such as papain (to produce Fab fragments) or pepsin (to produce F(ab')2 fragments).
  • F(ab')2 fragments contain the variable region, the light chain constant region and the CHI domain of the heavy chain.
  • the antibodies of the present invention can also be generated using various phage display methods known in the art. In phage display methods, functional antibody domains are displayed on the surface of phage particles which carry the polynucleotide sequences encoding them.
  • such phage can be utilized to display antigen binding domains expressed from a repertoire or combinatorial antibody library (e.g., human or murine).
  • Phage expressing an antigen binding domain that binds the antigen of interest can be selected or identified with antigen, e.g., using labeled antigen or antigen bound or captured to a solid surface or bead.
  • Phage used in these methods are typically filamentous phage including fd and Ml 3 binding domains expressed from phage with Fab, Fv or disulfide stabilized Fv antibody domains recombinantly fused to either the phage gene III or gene VIII protein.
  • the antibody coding regions from the phage can be isolated and used to generate whole antibodies, including human antibodies, or any other desired antigen binding fragment, and expressed in any desired host, including mammalian cells, insect cells, plant cells, yeast, and bacteria, e.g., as described in detail below.
  • a chimeric antibody is a molecule in which different portions of the antibody are derived from different animal species, such as antibodies having a variable region derived from a murine monoclonal antibody and a human immunoglobulin constant region.
  • Methods for producing chimeric antibodies are known in the art. See e.g., Morrison, Science 229: 1202 (1985); Oi et al., BioTechniques 4:214 (1986); Gillies et al., (1989) J. Immunol. Methods 125:191-202; U.S. Patent Nos. 5,807,715; 4,816,567; and 4,816397, which are inco ⁇ orated herein by reference in their entirety.
  • Humanized antibodies are antibody molecules from non-human species antibody that binds the desired antigen having one or more complementarity determining regions (CDRs) from the non- human species and a framework regions from a human immunoglobulin molecule.
  • CDRs complementarity determining regions
  • framework residues in the human framework regions will be substituted with the co ⁇ esponding residue from the CDR donor antibody to alter, preferably improve, antigen binding.
  • These framework substitutions are identified by methods well known in the art, e.g., by modeling of the interactions of the CDR and framework residues to identify framework residues important for antigen binding and sequence comparison to identify unusual framework residues at particular positions. (See, e.g., Queen et al., U.S. Patent No.
  • Antibodies can be humanized using a variety of techniques known in the art including, for example, CDR-grafting (EP 239,400; PCT publication WO 91/09967; U.S. Patent Nos. 5,225,539; 5,530,101; and 5,585,089), veneering or resurfacing (EP 592,106; EP 519,596; Padlan, Molecular Immunology 28(4/5):489-498 (1991); Studnicka et al., Protein Engineering 7(6):805-814 (1994); Roguska. et al., PNAS 91 :969-973 (1994)), and chain shuffling (U.S. Patent No. 5,565,332).
  • Human antibodies are particularly desirable for therapeutic treatment of human patients.
  • Human antibodies can be made by a variety of methods known in the art including phage display methods described above using antibody libraries derived from human immunoglobulin sequences. See also, U.S. Patent Nos. 4,444,887 and 4,716,111 ; and PCT publications WO 98/46645, WO 98/50433, WO 98/24893, WO 98/16654, WO 96/34096, WO 96/33735, and WO 91/10741; each of which is inco ⁇ orated herein by reference in its entirety.
  • Human antibodies can also be produced using transgenic mice which are incapable Of expressing functional endogenous immunoglobulins, but which can express human immunoglobulin genes.
  • the human heavy and light chain immunoglobulin gene complexes may be introduced randomly or by homologous recombination into mouse embryonic stem cells.
  • the human variable region, constant region, and diversity region may be introduced into mouse embryonic stem cells in addition to the human heavy and light chain genes.
  • the mouse heavy and light chain immunoglobulin genes may be rendered non-functional separately or simultaneously with the introduction of human immunoglobulin loci by homologous recombination. In particular, homozygous deletion of the JH region prevents endogenous antibody production.
  • the modified embryonic stem cells are expanded and microinjected into blastocysts to produce chimeric mice.
  • the chimeric mice are then bred to produce homozygous offspring which express human antibodies.
  • the transgenic mice are immunized in the normal fashion with a selected antigen, e.g., all or a portion of a polypeptide of the invention.
  • Monoclonal antibodies directed against the antigen can be obtained from the immunized, transgenic mice using conventional hybridoma technology.
  • the human immunoglobulin transgenes harbored by the transgenic mice rea ⁇ ange during B cell differentiation, and subsequently undergo class switching and somatic mutation.
  • antibodies to the polypeptides of the invention can, in turn, be utilized to generate anti-idiotype antibodies that "mimic" polypeptides of the invention using techniques well known to those skilled in the art. (See, e.g., Greenspan & Bona, FASEB J. 7(5):437-444; (1989) and Nissinoff, J. Immunol. 147(8):2429-2438 (1991)).
  • antibodies which bind to and competitively inhibit polypeptide multimerization and or binding of a polypeptide of the invention to a ligand can be used to generate anti-idiotypes that "mimic" the polypeptide multimerization and/or binding domain and, as a consequence, bind to and neutralize polypeptide and/or its ligand.
  • Such neutralizing anti-idiotypes or Fab fragments of such anti-idiotypes can be used in therapeutic regimens to neutralize polypeptide ligand(s)/receptor(s).
  • anti-idiotypic antibodies can be used to bind a polypeptide of the invention and or to bind its ligand(s)/receptor(s), and thereby block its biological activity.
  • antibodies which bind to and enhance polypeptide multimerization and/or binding, and or receptor/Iigand multimerization, binding and/or signaling can be used to generate anti-idiotypes that function as agonists of a polypeptide of the invention and/or its ligand receptor.
  • Such agonistic anti-idiotypes or Fab fragments of such anti-idiotypes can be used in therapeutic regimens as agonists of the polypeptides of the invention or its ligand(s)/receptor(s).
  • anti-idiotypic antibodies can be used to bind a polypeptide of the invention and/or to bind its ligand(s)/receptor(s), and thereby promote or enhance its biological activity.
  • Intrabodies of the invention can be produced using methods known in the art, such as those disclosed and reviewed in Chen et al., Hum. Gene Ther. 5:595-601 (1994); Marasco, W.A., Gene Ther. 4:11-15 (1997); Rondon and Marasco, Annu. Rev. Microbiol. 51:257-283 (1997); Proba et al., J. Mol. Biol. 275:245-253 (1998); Cohen et al., Oncogene 17:2445-2456 (1998); Ohage and Steipe, J. Mol. Biol. 291:1119-1128 (1999); Ohage et al., J. Mol. Biol. 291:1129-1134 (1999); Wirtz and Steipe, Protein Sci. 8:2245-2250 (1999); Zhu et al., J. Immunol. Methods 231 :207-222 (1999); and references cited therein.
  • the invention further provides polynucleotides comprising a nucleotide sequence encoding an antibody of the invention and fragments thereof.
  • the invention also encompasses polynucleotides that hybridize under stringent or altematively, under lower stringency hybridization conditions, e.g., as defined supra, to polynucleotides that encode an antibody, preferably, that specifically binds to a polypeptide of the invention, preferably, an antibody that binds to a polypeptide having the amino acid sequence of SEQ ED NO:Y, to a polypeptide encoded by a portion of SEQ ID NO:X as defined in columns 8 and 9 of Table 2, and/or to a polypeptide encoded by the cDNA contained in Clone ID NO:Z.
  • the polynucleotides may be obtained, and the nucleotide sequence of the polynucleotides determined, by any method known in the art.
  • a polynucleotide encoding the antibody may be assembled from chemically synthesized oligonucleotides (e.g., as described in Kutmeier et al., BioTechniques 17:242 (1994)), which, briefly, involves the synthesis of overlapping oligonucleotides containing portions of the sequence encoding the antibody, annealing and ligating of those oligonucleotides, and then amplification of the ligated oligonucleotides by PCR.
  • a polynucleotide encoding an antibody may be generated from nucleic acid from a suitable source. If a clone containing a nucleic acid encoding a particular antibody is not available, but the sequence of the antibody molecule is known, a nucleic acid encoding the immunoglobulin may be chemically synthesized or obtained from a suitable source (e.g., an antibody cDNA library, or a cDNA library generated from, or nucleic acid, preferably poly A+ RNA, isolated from, any tissue or cells expressing the antibody, such as hybridoma cells selected to express an antibody of the invention) by PCR amplification using synthetic primers hybridizable to the 3' and 5' ends of the sequence or by cloning using an oligonucleotide probe specific for the particular gene sequence to identify, e.g., a cDNA clone from a cDNA library that encodes the antibody. Amplified nucleic acids generated by a suitable source (e.
  • nucleotide sequence and co ⁇ esponding amino acid sequence of the antibody may be manipulated using methods well known in the art for the manipulation of nucleotide sequences, e.g., recombinant DNA techniques, site directed mutagenesis, PCR, etc.
  • the amino acid sequence of the heavy and/or light chain variable domains may be inspected to identify the sequences of the complementarity determining regions (CDRs) by methods that are well know in the art, e.g., by comparison to known amino acid sequences of other heavy and light chain variable regions to determine the regions of sequence hypervariability.
  • CDRs complementarity determining regions
  • one or more of the CDRs may be inserted within framework regions, e.g., into human framework regions to humanize a non-human antibody, as described supra.
  • the framework regions may be naturally occurring or consensus framework regions, and preferably human framework regions (see, e.g., Chothia et al., J. Mol. Biol.
  • the polynucleotide generated by the combination of the framework regions and CDRs encodes an antibody that specifically binds a polypeptide of the invention.
  • one or more amino acid substitutions may be made within the framework regions, and, preferably, the amino acid substitutions improve binding of the antibody to its antigen. Additionally, such methods may be used to make amino acid substitutions or deletions of one or more variable region cysteine residues participating in an intrachain disulfide bond to generate antibody molecules lacking one or more intrachain disulfide bonds.
  • Other alterations to the polynucleotide are encompassed by the present invention and within the skill of the art.
  • a chimeric antibody is a molecule in which different portions are derived from different animal species, such as those having a variable region derived from a murine mAb and a human immunoglobulin constant region, e.g., humanized antibodies.
  • the antibodies of the invention can be produced by any method known in the art for the synthesis of antibodies, in particular, by chemical synthesis or preferably, by recombinant expression techniques. Methods of producing antibodies include, but are not limited to, hybridoma technology, EBV transformation, and other methods discussed herein as well as through the use recombinant DNA technology, as discussed below. [240] Recombinant expression of an antibody of the invention, or fragment, derivative or analog thereof, (e.g., a heavy or light chain of an antibody of the invention or a single chain antibody of the invention), requires constmction of an expression vector containing a polynucleotide that encodes the antibody.
  • the vector for the production of the antibody molecule may be produced by recombinant DNA technology using techniques well known in the art.
  • methods for preparing a protein by expressing a polynucleotide containing an antibody encoding nucleotide sequence are described herein. Methods which are well known to those skilled in the art can be used to construct expression vectors containing antibody coding sequences and appropriate transcriptional and translational control signals. These methods include, for example, in vitro recombinant DNA techniques, synthetic techniques, and in vivo genetic recombination.
  • the invention thus, provides replicable vectors comprising a nucleotide sequence encoding an antibody molecule of the invention, or a heavy or light chain thereof, or a heavy or light chain variable domain, operably linked to a promoter.
  • Such vectors may include the nucleotide sequence encoding the constant region of the antibody molecule (see, e.g., PCT Publication WO 86/05807; PCT Publication WO 89/01036; and U.S. Patent No. 5,122,464) and the variable domain of the antibody may be cloned into such a vector for expression of the entire heavy or light chain.
  • the expression vector is transfe ⁇ ed to a host cell by conventional techniques and the transfected cells are then cultured by conventional techniques to produce an antibody of the invention.
  • the invention includes host cells containing a polynucleotide encoding an antibody of the invention, or a heavy or light chain thereof, or a single chain antibody of the invention, operably linked to a heterologous promoter.
  • vectors encoding both the heavy and light chains may be co-expressed in the host cell for expression of the entire immunoglobulin molecule, as detailed below.
  • host-expression vector systems may be utilized to express the antibody molecules of the invention.
  • Such host-expression systems represent vehicles by which the coding sequences of interest may be produced and subsequently purified, but also represent cells which may, when transformed or transfected with the appropriate nucleotide coding sequences, express an antibody molecule of the invention in situ.
  • These include but are not limited to microorganisms such as bacteria (e.g., E. coli, B.
  • subtilis transformed with recombinant bacteriophage DNA, plasmid DNA or cosmid DNA expression vectors containing antibody coding sequences; yeast (e.g., Saccharomyces, Pichia) transformed with recombinant yeast expression vectors containing antibody coding sequences; insect cell systems infected with recombinant vims expression vectors (e.g., baculovirus) containing antibody coding sequences; plant cell systems infected with recombinant vims expression vectors (e.g., cauliflower mosaic vims, CaMV; tobacco mosaic vims, TMV) or transformed with recombinant plasmid expression vectors (e.g., Ti plasmid) containing antibody coding sequences; or mammalian cell systems (e.g., COS, CHO, BHK, 293, 3T3 cells) harboring recombinant expression constmcts containing promoters derived from the genome of mammalian cells (e.g.,
  • bacterial cells such as Escherichia coli, and more preferably, eukaryotic cells, especially for the expression of whole recombinant antibody molecule, are used for the expression of a recombinant antibody molecule.
  • mammalian cells such as Chinese hamster ovary cells (CHO)
  • CHO Chinese hamster ovary cells
  • a vector such as the major intermediate early gene promoter element from human cytomegalovirus is an effective expression system for antibodies (Foecking et al., Gene 45:101 (1986); Cockett et al., Bio/Technology 8:2 (1990)).
  • a number of expression vectors may be advantageously selected depending upon the use intended for the antibody molecule being expressed.
  • vectors which direct the expression of high levels of fusion protein products that are readily purified may be desirable.
  • Such vectors include, but are not limited, to the E. coli expression vector pUR278 (Ruther et al., EMBO J. 2:1791 (1983)), in which the antibody coding sequence may be ligated individually into the vector in frame with the lac Z coding region so that a fusion protein is produced; pIN vectors (Inouye & Inouye, Nucleic Acids Res. 13:3101-3109 (1985); Van Heeke & Schuster, J. Biol. Chem. 24:5503-5509 .
  • pGEX vectors may also be used to express foreign polypeptides as fusion proteins with glutathione S-transferase (GST).
  • GST glutathione S-transferase
  • fusion proteins are soluble and can easily be purified from lysed cells by adso ⁇ tion and binding to matrix glutathione-agarose beads followed by elution in the presence of free glutathione.
  • the pGEX vectors are designed to include thrombin or factor Xa protease cleavage sites so that the cloned target gene product can be released from the GST moiety.
  • AcNPV Autographa californica nuclear polyhedrosis vims
  • the vims grows in Spodoptera frugiperda cells.
  • the antibody coding sequence may be cloned individually into non-essential regions (for example the polyhedrin gene) of the vims and placed under control of an AcNPV promoter (for example the polyhedrin promoter).
  • a number of viral-based expression systems may be utilized.
  • the antibody coding sequence of interest may be ligated to an adenovims transcription translation control complex, e.g., the late promoter and tripartite leader sequence.
  • This chimeric gene may then be inserted in the adenovims genome by in vitro or in vivo recombination. Insertion in a non- essential region of the viral genome (e.g., region El or E3) will result in a recombinant vims that is viable and capable of expressing the antibody molecule in infected hosts, (e.g., see Logan & Shenk, Proc.
  • Specific initiation signals may also be required for efficient translation of inserted antibody coding sequences. These signals include the ATG initiation codon and adjacent sequences. Furthermore, the initiation codon must be in phase with the reading frame of the desired coding sequence to ensure translation of the entire insert. These exogenous translational control signals and initiation codons can be of a variety of origins, both natural and synthetic. The efficiency of expression may be enhanced by the inclusion of appropriate transcription enhancer elements, transcription terminators, etc. (see Bittner et al., Methods in Enzymol. 153:51-544 (1987)).
  • a host cell strain may be chosen which modulates the expression of the inserted sequences, or modifies and processes the gene product in the specific fashion desired. Such modifications (e.g., glycosylation) and processing (e.g., cleavage) of protein products may be important for the function of the protein.
  • Different host cells have characteristic and specific mechanisms for the post-translational processing and modification of proteins and gene products. Appropriate cell lines or host systems can be chosen to ensure the co ⁇ ect modification and processing of the foreign protein expressed.
  • eukaryotic host cells which possess the cellular machinery for proper processing of the primary transcript, glycosylation, and phosphorylation of the gene product may be used.
  • Such mammalian host cells include but are not limited to CHO, VERY, BHK, Hela, COS, MDCK, 293, 3T3, WI38, and in particular, breast cancer cell lines such as, for example, BT483, Hs578T, HTB2, BT20 and T47D, and normal mammary gland cell line such as, for example, CRL7030 and Hs578Bst.
  • stable expression is prefe ⁇ ed.
  • cell lines which stably express the antibody molecule may be engineered.
  • host cells can be transformed with DNA controlled by appropriate expression control elements (e.g., promoter, enhancer, sequences, transcription terminators, polyadenylation sites, etc.), and a selectable marker.
  • appropriate expression control elements e.g., promoter, enhancer, sequences, transcription terminators, polyadenylation sites, etc.
  • engineered cells may be allowed to grow for 1-2 days in an enriched media, and then are switched to a selective media.
  • the selectable marker in the recombinant plasmid confers resistance to the selection and allows cells to stably integrate the plasmid into their chromosomes and grow to form foci which in turn can be cloned and expanded into cell lines.
  • This method may advantageously be used to engineer cell lines which express the antibody molecule.
  • Such engineered cell lines may be particularly useful in screening and evaluation of compounds that interact directly or indirectly with the antibody molecule.
  • a number of selection systems may be used, including but not limited to the he ⁇ es simplex vims thymidine kinase (Wigler et al., Cell 11 :223 (1977)), hypoxanthine-guanine phosphoribosyltransferase (Szybalska & Szybalski, Proc. Natl. Acad. Sci. USA 48:202 (1992)), and adenine phosphoribosyltransferase (Lowy et al., Cell 22:817 (1980)) genes can be employed in tk-, hgprt- or aprt- cells, respectively.
  • antimetabolite resistance can be used as the basis of selection for the following genes: dhfr, which confers resistance to methotrexate (Wigler et al., Natl. Acad. Sci. USA 77:357 (1980); O'Hare et al., Proc. Natl. Acad. Sci. USA 78:1527 (1981)); gpt, which confers resistance to mycophenolic acid (Mulligan & Berg, Proc. Natl. Acad. Sci.
  • the expression levels of an antibody molecule can be increased by vector amplification (for a review, see Bebbington and Hentschel, The use of vectors based on gene amplification for the expression of cloned genes in mammalian cells in DNA cloning, Vol.3. (Academic Press, New York, 1987)).
  • vector amplification for a review, see Bebbington and Hentschel, The use of vectors based on gene amplification for the expression of cloned genes in mammalian cells in DNA cloning, Vol.3. (Academic Press, New York, 1987)).
  • a marker in the vector system expressing antibody is amplifiable
  • increase in the level of inhibitor present in culture of host cell will increase the number of copies of the marker gene. Since the amplified region is associated with the antibody gene, production of the antibody will also increase (Crouse et al., Mol. Cell. Biol. 3:257 (1983)).
  • Vectors which use glutamine synthase (GS) or DHFR as the selectable markers can be amplified in the presence of the drugs methionine sulphoximine or methotrexate, respectively.
  • An advantage of glutamine synthase based vectors are the availabilty of cell lines (e.g., the murine myeloma cell line, NS0) which are glutamine synthase negative.
  • Glutamine synthase expression systems can also function in glutamine synthase expressing cells (e.g. Chinese Hamster Ovary (CHO) cells) by providing additional inhibitor to prevent the functioning of the endogenous gene.
  • glutamine synthase expression system and components thereof are detailed in PCT publications: WO87/04462; WO86/05807; WO89/01036; WO89/10404; and WO91/06657 which are inco ⁇ orated in their entireties by reference herein. Additionally, glutamine synthase expression vectors that may be used according to the present invention are commercially available from suplliers, including, for example Lonza Biologies, Inc. (Portsmouth, NH). Expression and production of monoclonal antibodies using a GS expression system in murine myeloma cells is described in Bebbington et al., Bio/technology 10:169(1992) and in Biblia and Robinson BiotechnoL Prog.
  • the host cell may be co-transfected with two expression vectors of the invention, the first vector encoding a heavy chain derived polypeptide and the second vector encoding a light chain derived polypeptide.
  • the two vectors may contain identical selectable markers which enable equal expression of heavy and light chain polypeptides.
  • a single vector may be used which encodes, and is capable of expressing, both heavy and light chain polypeptides. In such situations, the light chain should be placed before the heavy chain to avoid an excess of toxic free heavy chain (Proudfoot, Nature 322:52 (1986); Kohler, Proc. Natl. Acad. Sci. USA 77:2197 (1980)).
  • the coding sequences for the heavy and light chains may comprise cDNA or genomic DNA.
  • an antibody molecule of the invention may be purified by any method known in the art for purification of an immunoglobulin molecule, for example, by chromatography (e.g., ion exchange, affinity, particularly by affinity for the specific antigen after Protein A, and sizing column chromatography), centrifugation, differential solubility, or by any other standard technique for the purification of proteins.
  • chromatography e.g., ion exchange, affinity, particularly by affinity for the specific antigen after Protein A, and sizing column chromatography
  • centrifugation e.g., ion exchange, affinity, particularly by affinity for the specific antigen after Protein A, and sizing column chromatography
  • differential solubility e.g., differential solubility
  • the antibodies of the present invention or fragments thereof can be fused to heterologous polypeptide sequences described herein or otherwise known in the art, to facilitate purification.
  • the present invention encompasses antibodies recombinantly fused or chemically conjugated (including both covalently and non-covalently conjugations) to a polypeptide (or portion thereof, preferably at least 10, 20, 30, 40, 50, 60, 70, 80, 90 or 100 amino acids of the polypeptide) of the present invention to generate fusion proteins.
  • the fusion does not necessarily need to be direct, but may occur through linker sequences.
  • the antibodies may be specific for antigens other than polypeptides (or portion thereof, preferably at least 10, 20, 30, 40, 50, 60, 70, 80, 90 or 100 amino acids of the polypeptide) of the present invention.
  • antibodies may be used to target the polypeptides of the present invention to particular cell types, either in vitro or in vivo, by fusing or conjugating the polypeptides of the present invention to antibodies specific for particular cell surface receptors.
  • Antibodies fused or conjugated to the polypeptides of the present invention may also be used in in vitro immunoassays and purification methods using methods known in the art. See e.g., Harbor et al., supra, and PCT publication WO 93/21232; EP 439,095; Naramura et al., Immunol. Lett. 39:91-99 (1994); U.S.
  • the present invention further includes compositions comprising the polypeptides of the present invention fused or conjugated to antibody domains other than the variable regions.
  • the polypeptides of the present invention may be fused or conjugated to an antibody Fc region, or portion thereof.
  • the antibody portion fused to a polypeptide of the present invention may comprise the constant region, hinge region, CHI domain, CH2 domain, and CH3 domain or any combination of whole domains or portions thereof.
  • polypeptides may also be fused or conjugated to the above antibody portions to form multimers.
  • Fc portions fused to the polypeptides of the present invention can form dimers through disulfide bonding between the Fc portions.
  • Higher multimeric forms can be made by fusing the polypeptides to portions of IgA and IgM. Methods for fusing or conjugating the polypeptides of the present invention to antibody portions are known in the art. See, e.g., U.S. Patent Nos.
  • polypeptides co ⁇ esponding to a polypeptide, polypeptide fragment, or a variant of SEQ ID NO:Y may be fused or conjugated to the above antibody portions to increase the in vivo half life of the polypeptides or for use in immunoassays using methods known in the art. Further, the polypeptides co ⁇ esponding to SEQ ID NO:Y may be fused or conjugated to the above antibody portions to facilitate purification.
  • 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.
  • polypeptides of the present invention fused or conjugated to an antibody having disulfide- linked dimeric stmctures may also be more efficient in binding and neutralizing other molecules, than the monomeric secreted protein or protein fragment alone.
  • the Fc part in a fusion protein is beneficial in therapy and diagnosis, and thus can result in, for example, improved pharmacokinetic properties. See, for example, EP A 232,262.
  • the Fc portion may hinder therapy and diagnosis if the fusion protein is used as an antigen for immunizations.
  • human proteins such as hIL-5
  • Fc portions for the pu ⁇ ose of high- throughput screening assays to identify antagonists of hEL-5.
  • the antibodies or fragments thereof of the present invention can be fused to marker sequences, such as a peptide to facilitate purification.
  • the marker amino acid sequence is a hexa-histidine peptide, such as the tag provided in a pQE vector (QIAGEN, Inc., 9259 Eton Avenue, Chatsworth, CA, 91311), among others, many of which are commercially available.
  • hexa-histidine provides for convenient purification of the fusion protein.
  • peptide tags useful for purification include, but are not limited to, the "HA” tag, which co ⁇ esponds to an epitope derived from the influenza hemagglutinin protein (Wilson et al., Cell 37:767 (1984)) and the "flag" tag.
  • the present invention further encompasses antibodies or fragments thereof conjugated to a diagnostic or therapeutic agent.
  • the antibodies can be used diagnostically to, for example, monitor the development or progression of a tumor as part of a clinical testing procedure to, e.g., determine the efficacy of a given treatment regimen. Detection can be facilitated by coupling the antibody to a detectable substance.
  • detectable substances include various enzymes, prosthetic groups, fluorescent materials, luminescent materials, bioluminescent materials, radioactive materials, positron emitting metals using various positron emission tomographies, and nonradioactive paramagnetic metal ions.
  • the detectable substance may be coupled or conjugated either directly to the antibody (or fragment thereof) or indirectly, through an intermediate (such as, for example, a linker known in the art) using techniques known in the art. See, for example, U.S. Patent No. 4,741,900 for metal ions which can be conjugated to antibodies for use as diagnostics according to the present invention.
  • suitable enzymes include horseradish peroxidase, alkaline phosphatase, beta-galactosidase, or acetylcholinesterase;
  • suitable prosthetic group complexes include streptavidin/biotin and avidin biotin;
  • suitable fluorescent materials include umbelliferone, fluorescein, fluorescein isothiocyanate, rhodamine, dichlorotriazinylamme fluorescein, dansyl chloride or phycoerythrin;
  • an example of a luminescent material includes luminol;
  • examples of bioluminescent materials include luciferase, luciferin, and aequorin; and
  • suitable radioactive material include 1251, 1311, 11 lln or 99Tc.
  • an antibody or fragment thereof may be conjugated to a therapeutic moiety such as a cytotoxin, e.g., a cytostatic or cytocidal agent, a therapeutic agent or a radioactive metal ion, e.g., alpha-emitters such . as, for example, 213BL
  • a cytotoxin or cytotoxic agent includes any agent that is detrimental to cells.
  • Examples include paclitaxol, cytochalasin B, gramicidin D, ethidium bromide, emetine, mitomycin, etoposide, tenoposide, vincristine, vinblastine, colchicin, doxombicin, daunombicin, dihydroxy anthracin dione, mitoxantrone, mithramycin, actinomycin D, 1-dehydro testosterone, glucocorticoids, procaine, tetracaine, lidocaine, propranolol, and puromycin and analogs or homologs thereof.
  • Therapeutic agents include, but are not limited to, antimetabolites (e.g., methotrexate, 6- mercaptopurine, 6-thioguanine, cytarabine, 5-fluorouracil decarbazine), alkylating agents (e.g., mechlorethamine, thioepa chlorambucil, melphalan, carmustine (BSNU) and lomustine (CCNU), cyclothosphamide, busulfan, dibromomannitol, streptozotocin, mitomycin C, and cis- dichlorodiamine platinum (U) (DDP) cisplatin), anthracyclines (e.g., daunombicin (formerly daunomycin) and doxombicin), antibiotics (e.g., dactinomycin (formerly actinomycin), bleomycin, mithramycin, and anthramycin (AMC)), and anti-mitotic agents (e.g., vincris
  • the conjugates of the invention can be used for modifying a given biological response, the therapeutic agent or drug moiety is not to be constmed as limited to classical chemical therapeutic agents.
  • the drag moiety may be a protein or polypeptide possessing a desired biological activity.
  • Such proteins may include, for example, a toxin such as abrin, ricin A, pseudomonas exotoxin, or diphtheria toxin; a protein such as tumor necrosis factor, a-interferon, ⁇ -interferon, nerve growth factor, platelet derived growth factor, tissue plasminogen activator, an apoptotic agent, e.g., TNF-alpha, TNF-beta, AIM I (See, International Publication No. WO 97/33899), A M II (See, International Publication No. WO 97/34911), Fas Ligand (Takahashi et al, Int.
  • a toxin such as abrin, ricin A, pseudomonas exotoxin, or diphtheria toxin
  • a protein such as tumor necrosis factor, a-interferon, ⁇ -interferon, nerve growth factor, platelet derived growth factor, tissue plasminogen activator, an a
  • VEGI See, International Publication No. WO 99/23105
  • a thrombotic agent or an anti- angiogenic agent e.g., angiostatin or endostatin
  • biological response modifiers such as, for example, lymphokines, interleukin-1 ("EL-1"), interleukin-2 (“EL-2”), interleukin-6 (“EL-6”), granulocyte macrophage colony stimulating factor (“GM-CSF”), granulocyte colony stimulating factor (“G-CSF”), or other growth factors.
  • EL-1 interleukin-1
  • EL-2 interleukin-2
  • EL-6 interleukin-6
  • GM-CSF granulocyte macrophage colony stimulating factor
  • G-CSF granulocyte colony stimulating factor
  • Antibodies may also be attached to solid supports, which are particularly useful for immunoassays or purification of the target antigen.
  • solid supports include, but are not limited to, glass, cellulose, polyacrylamide, nylon, polystyrene, polyvinyl chloride or polypropylene.
  • an antibody can be conjugated to a second antibody to form an antibody heteroconjugate as described by Segal in U.S. Patent No. 4,676,980, which is inco ⁇ orated herein by reference in its entirety.
  • An antibody, with or without a therapeutic moiety conjugated to it, administered alone or in combination with cytotoxic factor(s) and/or cytokine(s) can be used as a therapeutic.
  • the antibodies of the invention may be utilized for immunophenotyping of cell lines and biological samples. Translation products of the gene of the present invention may be useful as cell-specific markers, or more specifically as cellular markers that are differentially expressed at various stages of differentiation and/or maturation of particular cell types. Monoclonal antibodies directed against a specific epitope, or combination of epitopes, will allow for the screening of cellular populations expressing the marker. Various techniques can be utilized using monoclonal antibodies to screen for cellular populations expressing the marker(s), and include magnetic separation using antibody-coated magnetic beads, "panning" with antibody attached to a solid matrix (i.e., plate), and flow cytometry (See, e.g., U.S.
  • MRD minimal residual disease
  • GVHD Graft-versus-Host Disease
  • these techniques allow for the screening of hematopoietic stem and progenitor cells capable of undergoing proliferation and/or differentiation, as might be found in human umbilical cord blood.
  • the antibodies of the invention may be assayed for immunospecific binding by any method known in the art.
  • the immunoassays which can be used include but are not limited to competitive and non-competitive assay systems using techniques such as western blots, radioimmunoassays, ELISA (enzyme linked immunosorbent assay), "sandwich” immunoassays, immunoprecipitation assays, precipitin reactions, gel diffusion precipitin reactions, immunodiffusion assays, agglutination assays, complement-fixation assays, immunoradiometric assays, fluorescent immunoassays, and protein A immunoassays, to name but a few.
  • Immunoprecipitation protocols generally comprise lysing a population of cells in a lysis buffer such as RIP A buffer (1% NP-40 or Triton X- 100, 1% sodium deoxycholate, 0.1% SDS, 0.15 M NaCl, 0.01 M sodium phosphate at pH 7.2, 1% Trasylol) supplemented with protein phosphatase and/or protease inhibitors (e.g., EDTA, PMSF, aprotinin, sodium vanadate), adding the antibody of interest to the cell lysate, incubating for a period of time (e.g., 1-4 hours) at 4° C, adding protein A and or protein G sepharose beads to the cell lysate, incubating for about an hour or more at 4° C, washing the beads in lysis buffer and resuspending the beads in SDS/sample buffer.
  • a lysis buffer such as RIP A buffer (1% NP-40 or Triton X- 100, 1% sodium de
  • the ability of the antibody of interest to immunoprecipitate a particular antigen can be assessed by, e.g., western blot analysis.
  • One of skill in the art would be knowledgeable as to the parameters that can be modified to increase the binding of the antibody to an antigen and decrease the background (e.g., pre- clearing the cell lysate with sepharose beads).
  • immunoprecipitation protocols see, e.g., Ausubel et al., eds., (1994), Cu ⁇ ent Protocols in Molecular Biology, Vol. 1, John Wiley & Sons, Inc., New York, section 10.16.1.
  • Western blot analysis generally comprises preparing protein samples, electrophoresis of the protein samples in a polyacrylamide gel (e.g., 8%- 20% SDS-PAGE depending on the molecular weight of the antigen), transferring the protein sample from the polyacrylamide gel to a membrane such as nitrocellulose, PVDF or nylon, blocking the membrane in blocking solution (e.g., PBS with 3% BSA or non-fat milk), washing the membrane in washing buffer (e.g., PBS-Tween 20), blocking the membrane with primary antibody (the antibody of interest) diluted in blocking buffer, washing the membrane in washing buffer, blocking the membrane with a secondary antibody (which recognizes the primary antibody, e.g., an anti-human antibody) conjugated to an enzymatic substrate (e.g., horseradish peroxidase or alkaline phosphatase) or radioactive molecule (e.g., 32P or 1251) diluted in blocking buffer, washing the membrane in wash buffer, and detecting the presence of the antigen
  • ELISAs comprise preparing antigen, coating the well of a 96 well microtiter plate with the antigen, adding the antibody of interest conjugated to a detectable compound such as an enzymatic substrate (e.g., horseradish peroxidase or alkaline phosphatase) to the well and incubating for a period of time, and detecting the presence of the antigen.
  • a detectable compound such as an enzymatic substrate (e.g., horseradish peroxidase or alkaline phosphatase)
  • a detectable compound such as an enzymatic substrate (e.g., horseradish peroxidase or alkaline phosphatase)
  • a second antibody conjugated to a detectable compound may be added following the addition of the antigen of interest to the coated well.
  • affinity of the antibody of interest for a particular antigen and the binding off-rates can be determined from the data by scatchard plot analysis.
  • Competition with a second antibody can also be determined using radioimmunoassays.
  • the antigen is incubated with antibody of interest conjugated to a labeled compound (e.g., 3H or 1251) in the presence of increasing amounts of an unlabeled second antibody.
  • Antibodies of the invention may be characterized using immunocytochemisty methods on cells (e.g., mammalian cells, such as CHO cells) transfected with a vector enabling the expression of an antigen or with vector alone using techniques commonly known in the art.
  • cells e.g., mammalian cells, such as CHO cells
  • Antibodies that bind antigen transfected cells, but not vector-only transfected cells, are antigen specific.
  • the present invention is further directed to antibody-based therapies which involve administering antibodies of the invention to an animal, preferably a mammal, and most preferably a human, patient for treating one or more of the disclosed diseases, disorders, or conditions.
  • Therapeutic compounds of the invention include, but are not limited to, antibodies of the invention (including fragments, analogs and derivatives thereof as described herein) and nucleic acids encoding antibodies of the invention (including fragments, analogs and derivatives thereof and anti-idiotypic antibodies as described herein).
  • the antibodies of the invention can be used to treat, inhibit or prevent diseases, disorders or conditions associated with abe ⁇ ant expression and/or activity of a polypeptide of the invention, including, but not limited to, any one or more of the diseases, disorders, or conditions described herein.
  • the treatment and/or prevention of diseases, disorders, or conditions associated with abe ⁇ ant expression and/or activity of a polypeptide of the invention includes, but is not limited to, alleviating symptoms associated with those diseases, disorders or conditions.
  • Antibodies of the invention may be provided in pharmaceutically acceptable compositions as known in the art or as described herein.
  • the present invention is directed to antibody-based therapies which involve administering antibodies of the invention to an animal, preferably a mammal, and most preferably a human, patient for treating one or more diseases, disorders, or conditions, including but not limited to: neural disorders, immune system disorders, muscular disorders, reproductive disorders, gastrointestinal disorders, pulmonary disorders, cardiovascular disorders, renal disorders, proliferative disorders, and/or cancerous diseases and conditions., and/or as described elsewhere herein.
  • Therapeutic compounds of the invention include, but are not limited to, antibodies of the invention (e.g., antibodies directed to the full length protein expressed on the cell surface of a mammalian cell; antibodies directed to an epitope of a polypeptide of the invention (such as, for example, a predicted linear epitope shown in column 7 of Table 1A; or a conforrnational epitope, including fragments, analogs and derivatives thereof as described herein) and nucleic acids encoding antibodies of the invention (including fragments, analogs and derivatives thereof and anti-idiotypic antibodies as described herein).
  • antibodies of the invention e.g., antibodies directed to the full length protein expressed on the cell surface of a mammalian cell
  • antibodies directed to an epitope of a polypeptide of the invention such as, for example, a predicted linear epitope shown in column 7 of Table 1A; or a conforrnational epitope, including fragments, analogs and derivatives thereof as described herein
  • nucleic acids encoding antibodies of the invention
  • the antibodies of the invention can be used to treat, inhibit or prevent diseases, disorders or conditions associated with abe ⁇ ant expression and/or activity of a polypeptide of the invention, including, but not limited to, any one or more of the diseases, disorders, or conditions described herein.
  • the treatment and/or prevention of diseases, disorders, or conditions associated with abe ⁇ ant expression and/or activity of a polypeptide of the invention includes, but is not limited to, alleviating symptoms associated with those diseases, disorders or conditions.
  • Antibodies of the invention may be provided in pharmaceutically acceptable compositions as known in the art or as described herein.
  • a summary of the ways in which the antibodies of the present invention may be used therapeutically includes binding polynucleotides or polypeptides of the present invention locally or systemically in the body or by direct cytotoxicity of the antibody, e.g. as mediated by complement (CDC) or by effector cells (ADCC). Some of these approaches are described in more detail below.
  • the antibodies of this invention may be advantageously utilized in combination with other monoclonal or chimeric antibodies, or with lymphokines or hematopoietic growth factors (such as, e.g., IL-2, IL-3 and EL-7), for example, which serve to increase the number or activity of effector cells which interact with the antibodies.
  • lymphokines or hematopoietic growth factors such as, e.g., IL-2, IL-3 and EL-7
  • the antibodies of the invention may be administered alone or in combination with other types of treatments (e.g., radiation therapy, chemotherapy, hormonal therapy, immunotherapy and anti-tumor agents).
  • treatments e.g., radiation therapy, chemotherapy, hormonal therapy, immunotherapy and anti-tumor agents.
  • administration of products of a species origin or species reactivity in the case of antibodies
  • human antibodies, fragments derivatives, analogs, or nucleic acids are administered to a human patient for therapy or prophylaxis.
  • Prefe ⁇ ed binding affinities include those with a dissociation constant or Kd less than 5 X IO “2 M, 10 "2 M, 5 X 10 "3 M, 10 “3 M, 5 X 10 "4 M, 10 “4 M, 5 X 10 "5 M, 10 "5 M, 5 X IO “6 M, IO “6 M, 5 X 10 " 7 M, IO “7 M, 5 X 10 '8 M, 10 “8 M, 5 X IO “9 M, IO "9 M, 5 X 10 "10 M, 10 “10 M, 5 X 10 "11 M, IO “11 M, 5 X IO "12 M, IO “12 M, 5 X IO “13 M, 10 " 13 M, 5 X IO “14 M, IO “14 M, 5 X IO “15 M, and IO “15 M.
  • nucleic acids comprising sequences encoding antibodies or functional derivatives thereof, are administered to treat, inhibit or prevent a disease or disorder associated with abe ⁇ ant expression and or activity of a polypeptide of the invention, by way of gene therapy.
  • Gene therapy refers to therapy performed by the administration to a subject of an expressed or expressible nucleic acid.
  • the nucleic acids produce their encoded protein that mediates a therapeutic effect.
  • Any of the methods for gene therapy available in the art can be used according to the present invention. Exemplary methods are described below.
  • the compound comprises nucleic acid sequences encoding an antibody, said nucleic acid sequences being part of expression vectors that express the antibody or fragments or chimeric proteins or heavy or light chains thereof in a suitable host.
  • nucleic acid sequences have promoters operably linked to the antibody coding region, said promoter being inducible or constitutive, and, optionally, tissue-specific.
  • nucleic acid molecules are used in which the antibody coding sequences and any other desired sequences are flanked by regions that promote homologous recombination at a desired site in the genome, thus providing for intrachromosomal expression of the antibody encoding nucleic acids (Koller and Smithies, Proc. Natl. Acad. Sci. USA 86:8932-8935 (1989); Zijlstra et al., Nature 342:435-438 (1989).
  • the expressed antibody molecule is a single chain antibody; alternatively, the nucleic acid sequences include sequences encoding both the heavy and light chains, or fragments thereof, of the antibody.
  • nucleic acids into a patient may be either direct, in which case the patient is directly exposed to the nucleic acid or nucleic acid- carrying vectors, or indirect, in which case, cells are first transformed with the nucleic acids in vitro, then transplanted into the patient. These two approaches are known, respectively, as in vivo or ex vivo gene therapy.
  • the nucleic acid sequences are directly administered in vivo, where it is expressed to produce the encoded product. This can be accomplished by any of numerous methods known in the art, e.g., by constmcting them as part of an appropriate nucleic acid expression vector and administering it so that they become intracellular, e.g., by infection using defective or attenuated retro virals or other viral vectors (see U.S. Patent No.
  • microparticle bombardment e.g., a gene gun; Biolistic, Dupont
  • coating lipids or cell-surface receptors or transfecting agents, encapsulation in liposomes, microparticles, or microcapsules, or by administering them in linkage to a peptide which is known to enter the nucleus, by administering it in linkage to a ligand subject to receptor-mediated endocytosis (see, e.g., Wu and Wu, J. Biol. Chem. 262:4429-4432 (1987)) (which can be used to target cell types specifically expressing the receptors), etc.
  • nucleic acid- ligand complexes can be formed in which the ligand comprises a fusogenic viral peptide to dismpt endosomes, allowing the nucleic acid to avoid lysosomal degradation.
  • the nucleic acid can be targeted in vivo for cell specific uptake and expression, by targeting a specific receptor (see, e.g., PCT Publications WO 92/06180; WO 92/22635; WO92/20316; WO93/14188, WO 93/20221).
  • the nucleic acid can be introduced intracellularly and inco ⁇ orated within host cell DNA for expression, by homologous recombination (Koller and Smithies, Proc. Natl. Acad. Sci. USA 86:8932-8935 (1989); Zijlstra et al., Nature 342:435-438 (1989)).
  • viral vectors that contains nucleic acid sequences encoding an antibody of the invention are used.
  • a retroviral vector can be used (see Miller et al., Meth. Enzymol. 217:581-599 (1993)). These retroviral vectors contain the components necessary for the co ⁇ ect packaging of the viral genome and integration into the host cell DNA.
  • the nucleic acid sequences encoding the antibody to be used in gene therapy are cloned into one or more vectors, which facilitates delivery of the gene into a patient.
  • retroviral vectors More detail about retroviral vectors can be found in Boesen et al., Biotherapy 6:291-302 (1994), which describes the use of a retroviral vector to deliver the mdrl gene to hematopoietic stem cells in order to make the stem cells more resistant to chemotherapy.
  • Other references illustrating the use of retroviral vectors in gene therapy are: Clowes et al., J. Clin. Invest. 93:644-651 (1994); Kiem et al., Blood 83:1467-1473 (1994); Salmons and Gunzberg, Human Gene Therapy 4:129-141 (1993); and Grossman and Wilson, Cu ⁇ . Opin. in Genetics and DeveL 3:110-114 (1993).
  • Adenovimses are other viral vectors that can be used in gene therapy. Adenovimses are especially attractive vehicles for delivering genes to respiratory epithelia. Adenovimses naturally infect respiratory epithelia where they cause a mild disease. Other targets for adenovirus-based delivery systems are liver, the central nervous system, endothelial cells, and muscle. Adenoviruses have the advantage of being capable of infecting non-dividing cells. Kozarsky and Wilson, Cu ⁇ ent Opinion in Genetics and Development 3:499-503 (1993) present a review of adenovims-based gene therapy.
  • Adeno-associated vims has also been proposed for use in gene therapy (Walsh et al., Proc. Soc. Exp. Biol. Med. 204:289-300 (1993); U.S. Patent No. 5,436,146).
  • Another approach to gene therapy involves transferring a gene to cells in tissue culture by such methods as electroporation, lipofection, calcium phosphate mediated transfection, or viral infection. Usually, the method of transfer includes the transfer of a selectable marker to the cells. The cells are then placed under selection to isolate those cells that have taken up and are expressing the transfe ⁇ ed gene. Those cells are then delivered to a patient.
  • the nucleic acid is introduced into a cell prior to administration in vivo of the resulting recombinant cell.
  • introduction can be carried out by any method known in the art, including but not limited to transfection, electroporation, microinjection, infection with a viral or bacteriophage vector containing the nucleic acid sequences, cell fusion, chromosome-mediated gene transfer, microcell-mediated gene transfer, spheroplast fusion, etc.
  • Numerous techniques are known in the art for the introduction of foreign genes into cells (see, e.g., Loeffler and Behr, Meth. Enzymol. 217:599-618 (1993); Cohen et al., Meth. Enzymol.
  • the technique should provide for the stable transfer of the nucleic acid to the cell, so that the nucleic acid is expressible by the cell and preferably heritable and expressible by its cell progeny.
  • the resulting recombinant cells can be delivered to a patient by various methods known in the art.
  • Recombinant blood cells e.g., hematopoietic stem or progenitor cells
  • the amount of cells envisioned for use depends on the desired effect, patient state, etc., and can be determined by one skilled in the art.
  • Cells into which a nucleic acid can be introduced for pu ⁇ oses of gene therapy encompass any desired, available cell type, and include but are not limited to epithelial cells, endothelial cells, keratinocytes, fibroblasts, muscle cells, hepatocytes; blood cells such as T lymphocytes, B lymphocytes, monocytes, macrophages, neutrophils, eosinophils, megakaryocytes, granulocytes; various stem or progenitor cells, in particular hematopoietic stem or progenitor cells, e.g., as obtained from bone ma ⁇ ow, umbilical cord blood, peripheral blood, fetal liver, etc.
  • the cell used for gene therapy is autologous to the patient.
  • nucleic acid sequences encoding an antibody are introduced into the cells such that they are expressible by the cells or their progeny, and the recombinant cells are then administered in vivo for therapeutic effect.
  • stem or progenitor cells are used. Any stem and/or progenitor cells which can be isolated and maintained in vitro can potentially be used in accordance with this embodiment of the present invention (see e.g. PCT Publication WO 94/08598; Stemple and Anderson, Cell 71:973-985 (1992); Rheinwald, Meth. Cell Bio. 21A:229 (1980); and Pittelkow and Scott, Mayo Clinic Proc.
  • the nucleic acid to be introduced for pu ⁇ oses of gene therapy comprises an inducible promoter operably linked to the coding region, such that expression of the nucleic acid is controllable by the presence or absence of an appropriate inducer of transcription.
  • the compounds or pharmaceutical compositions of the invention are preferably tested in vitro, and then in vivo for the desired therapeutic or prophylactic activity, prior to use in humans.
  • in vitro assays to demonstrate the therapeutic or prophylactic utility of a compound or pharmaceutical composition include, the effect of a compound on a cell line or a patient tissue sample.
  • the effect of the compound or composition on the cell line and/or tissue sample can be determined utilizing techniques known to those of skill in the art including, but not limited to, rosette formation assays and cell lysis assays.
  • in vitro assays which can be used to determine whether administration of a specific compound is indicated, include in vitro cell culture assays in which a patient tissue sample is grown in culture, and exposed to or otherwise administered a compound, and the effect of such compound upon the tissue sample is observed.
  • the invention provides methods of treatment, inhibition and prophylaxis by administration to a subject of an effective amount of a compound or pharmaceutical composition of the invention, preferably a polypeptide or antibody of the invention.
  • the compound is substantially purified (e.g., substantially free from substances that limit its effect or produce undesired side-effects).
  • the subject is preferably an animal, including but not limited to animals such as cows, pigs, horses, chickens, cats, dogs, etc., and is preferably a mammal, and most preferably human.
  • Formulations and methods of administration that can be employed when the compound comprises a nucleic acid or an immunoglobulin are described above; additional appropriate formulations and routes of administration can be selected from among those described herein below.
  • Various delivery systems are known and can be used to administer a compound of the invention, e.g., encapsulation in liposomes, microparticles, microcapsules, recombinant cells capable of expressing the compound, receptor-mediated endocytosis (see, e.g., Wu and Wu, J. Biol. Chem. 262:4429-4432 (1987)), constmction of a nucleic acid as part of a retroviral or other vector, etc.
  • Methods of introduction include but are not limited to intradermal, intramuscular, intraperitoneal, intravenous, subcutaneous, intranasal, epidural, and oral routes.
  • the compounds or compositions may be administered by any convenient route, for example by infusion or bolus injection, by abso ⁇ tion through epithelial or mucocutaneous linings (e.g., oral mucosa, rectal and intestinal mucosa, etc.) and may be administered together with other biologically active agents. Administration can be systemic or local.
  • Pulmonary administration can also be employed, e.g., by use of an inhaler or nebulizer, and formulation with an aerosolizing agent.
  • the pharmaceutical compounds or compositions of the invention may be desirable to administer the pharmaceutical compounds or compositions of the invention locally to the area in need of treatment; this may be achieved by, for example, and not by way of limitation, local infusion during surgery, topical application, e.g., in conjunction with a wound dressing after surgery, by injection, by means of a catheter, by means of a suppository, or by means of an implant, said implant being of a porous, non-porous, or gelatinous material, including membranes, such as sialastic membranes, or fibers.
  • a protein including an antibody
  • care must be taken to use materials to which the protein does not absorb.
  • the compound or composition can be delivered in a vesicle, in particular a liposome (see Langer, Science 249:1527-1533 (1990); Treat et al., in Liposomes in the Therapy of Infectious Disease and Cancer, Lopez-Berestein and Fidler (eds.), Liss, New York, pp. 353- 365 (1989); Lopez-Berestein, ibid., pp. 317-327; see generally ibid.)
  • the compound or composition can be delivered in a controlled release system.
  • a pump may be used (see Langer, supra; Sefton, CRC Crit. Ref. Biomed. Eng. 14:201 (1987); Buchwald et al., Surgery 88:507 (1980); Saudek et al., N. Engl. J. Med. 321:574 (1989)).
  • polymeric materials can be used (see Medical Applications of Controlled Release, Langer and Wise (eds.), CRC Pres., Boca Raton, Florida (1974); Controlled Dmg Bioavailability, Dmg Product Design and Performance, Smolen and Ball (eds.), Wiley, New York (1984); Ranger and Peppas, J., Macromol. Sci. Rev. Macromol. Chem. 23:61 (1983); see also Levy et al., Science 228:190 (1985); During et al., Ann. Neurol. 25:351 (1989); Howard et al., J.Neurosurg. 71:105 (1989)).
  • a controlled release system can be placed in proximity of the therapeutic target, e.g., the brain, thus requiring only a fraction of the systemic dose (see, e.g., Goodson, in Medical Applications of Controlled Release, supra, vol. 2, pp. 115-138 (1984)).
  • the nucleic acid in a specific embodiment where the compound of the invention is a nucleic acid encoding a protein, can be administered in vivo to promote expression of its encoded protein, by constmcting it as part of an appropriate nucleic acid expression vector and administering it so that it becomes intracellular, e.g., by use of a retroviral vector (see U.S. Patent No.
  • a nucleic acid can be introduced intracellularly and inco ⁇ orated within host cell DNA for expression, by homologous recombination.
  • compositions comprise a therapeutically effective amount of a compound, and a pharmaceutically acceptable carrier.
  • pharmaceutically acceptable means approved by a regulatory agency of the Federal or a state government or listed in the U.S. Pharmacopeia or other generally recognized pharmacopeia for use in animals, and more particularly in humans.
  • carrier refers to a diluent, adjuvant, excipient, or vehicle with which the therapeutic is administered.
  • Such pharmaceutical carriers can be sterile liquids, such as water and oils, including those of petroleum, animal, vegetable or synthetic origin, such as peanut oil, soybean oil, mineral oil, sesame oil and the like.
  • Water is a prefe ⁇ ed carrier when the pharmaceutical composition is administered intravenously.
  • Saline solutions and aqueous dextrose and glycerol solutions can also be employed as liquid carriers, particularly for injectable solutions.
  • Suitable pharmaceutical excipients include starch, glucose, lactose, sucrose, gelatin, malt, rice, flour, chalk, silica gel, sodium stearate, glycerol monostearate, talc, sodium chloride, dried skim milk, glycerol, propylene, glycol, water, ethanol and the like.
  • the composition if desired, can also contain minor amounts of wetting or emulsifying agents, or pH buffering agents.
  • compositions can take the form of solutions, suspensions, emulsion, tablets, pills, capsules, powders, sustained-release formulations and the like.
  • the composition can be formulated as a suppository, with traditional binders and carriers such as triglycerides.
  • Oral formulation can include standard carriers such as pharmaceutical grades of mannitol, lactose, starch, magnesium stearate, sodium saccharine, cellulose, magnesium carbonate, etc. Examples of suitable pharmaceutical carriers are described in "Remington's Pharmaceutical Sciences" by E.W. Martin.
  • Such compositions will contain a therapeutically effective amount of the compound, preferably in purified form, together with a suitable amount of carrier so as to provide the form for proper administration to the patient.
  • the formulation should suit the mode of administration.
  • the composition is formulated in accordance with routine procedures as a pharmaceutical composition adapted for intravenous administration to human beings.
  • compositions for intravenous administration are solutions in sterile isotonic aqueous buffer.
  • the composition may also include a solubilizing agent and a local anesthetic such as lignocaine to ease pain at the site of the injection.
  • the ingredients are supplied either separately or mixed together in unit dosage form, for example, as a dry lyophilized powder or water free concentrate in a hermetically sealed container such as an ampoule or sachette indicating the quantity of active agent.
  • composition is to be administered by infusion, it can be dispensed with an infusion bottle containing sterile pharmaceutical grade water or saline.
  • an ampoule of sterile water for injection or saline can be provided so that the ingredients may be mixed prior to administration.
  • the compounds of the invention can be formulated as neutral or salt forms.
  • Pharmaceutically acceptable salts include those formed with anions such as those derived from hydrochloric, phosphoric, acetic, oxalic, tartaric acids, etc., and those formed with cations such as those derived from sodium, potassium, ammonium, calcium, ferric hydroxides, isopropylamine, triethylamine, 2-ethylamino ethanol, histidine, procaine, etc.
  • the amount of the compound of the invention which will be effective in the treatment, inhibition and prevention of a disease or disorder associated with abe ⁇ ant expression and/or activity of a polypeptide of the invention can be determined by standard clinical techniques. In addition, in vitro assays may optionally be employed to help identify optimal dosage ranges.
  • Effective doses may be extrapolated from dose-response curves derived from in vitro or animal model test systems.
  • the dosage administered to a patient is typically 0.1 mg/kg to 100 mg/kg of the patient's body weight.
  • the dosage administered to a patient is between 0.1 mg/kg and 20 mg/kg of the patient's body weight, more preferably 1 mg/kg to 10 mg/kg of the patient's body weight.
  • human antibodies have a longer half-life within the human body than antibodies from other species due to the immune response to the foreign polypeptides. Thus, lower dosages of human antibodies and less frequent administration is often possible.
  • the dosage and frequency of administration of antibodies of the invention may be reduced by enhancing uptake and tissue penetration (e.g., into the brain) of the antibodies by modifications such as, for example, lipidation.
  • 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.
  • Optionally associated with such container(s) can be a notice in the form prescribed by a governmental agency regulating the manufacture, use or sale of pharmaceuticals or biological products, which notice reflects approval by the agency of manufacture, use or sale for human administration.
  • Labeled antibodies, and derivatives and analogs thereof, which specifically bind to a polypeptide of interest can be used for diagnostic purposes to detect, diagnose, or monitor diseases, disorders, and/or conditions associated with the abe ⁇ ant expression and/or activity of a polypeptide of the invention.
  • the invention provides for the detection of abe ⁇ ant expression of a polypeptide of interest, comprising (a) assaying the expression of the polypeptide of interest in cells or body fluid of an individual using one or more antibodies specific to the polypeptide interest and (b) comparing the level of gene expression with a standard gene expression level, whereby an increase or decrease in the assayed polypeptide gene expression level compared to the standard expression level is indicative of abe ⁇ ant expression.
  • the invention provides a diagnostic assay for diagnosing a disorder, comprising (a) assaying the expression of the polypeptide of interest in cells or body fluid of an individual using one or more antibodies specific to the polypeptide interest and (b) comparing the level of gene expression with a standard gene expression level, whereby an increase or decrease in the assayed polypeptide gene expression level compared to the standard expression level is indicative of a particular disorder.
  • a diagnostic assay for diagnosing a disorder comprising (a) assaying the expression of the polypeptide of interest in cells or body fluid of an individual using one or more antibodies specific to the polypeptide interest and (b) comparing the level of gene expression with a standard gene expression level, whereby an increase or decrease in the assayed polypeptide gene expression level compared to the standard expression level is indicative of a particular disorder.
  • the presence of a relatively high amount of transcript in biopsied tissue from an individual may indicate a predisposition for the development of the disease, or may provide a means for detecting the disease prior
  • Antibodies of the invention can be used to assay protein levels in a biological sample using classical immunohistological methods known to those of skill in the art (e.g., see Jalkanen et al., J. Cell. Biol. 101 :976-985 (1985); Jalkanen et al., J. Cell . Biol. 105:3087- 3096 (1987)).
  • Other antibody-based methods useful for detecting protein gene expression include immunoassays, such as the enzyme linked immunosorbent assay (ELISA) and the radioimmunoassay (RIA).
  • Suitable antibody assay labels include enzyme labels, such as, glucose oxidase; radioisotopes, such as iodine (1251, 1211), carbon (14C), sulfur (35S), tritium (3H), indium (112In), and technetium (99Tc); luminescent labels, such as luminol; and fluorescent labels, such as fluorescein and rhodamine, and biotin.
  • enzyme labels such as, glucose oxidase
  • radioisotopes such as iodine (1251, 1211), carbon (14C), sulfur (35S), tritium (3H), indium (112In), and technetium (99Tc)
  • luminescent labels such as luminol
  • fluorescent labels such as fluorescein and rhodamine, and biotin.
  • diagnosis comprises: a) administering (for example, parenterally, subcutaneously, or intraperitoneally) to a subject an effective amount of a labeled molecule which specifically binds to the polypeptide of interest; b) waiting for a time interval following the administering for permitting the labeled molecule to preferentially concentrate at sites in the subject where the polypeptide is expressed (and for unbound labeled molecule to be cleared to background level); c) determining background level; and d) detecting the labeled molecule in the subject, such that detection of labeled molecule above the background level indicates that the subject has a particular disease or disorder associated with abe ⁇ ant expression of the polypeptide of interest.
  • Background level can be determined by. various methods including, comparing the amount of labeled molecule detected to a standard value previously determined for a particular system.

Landscapes

  • Health & Medical Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Organic Chemistry (AREA)
  • Genetics & Genomics (AREA)
  • Zoology (AREA)
  • Engineering & Computer Science (AREA)
  • Wood Science & Technology (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • General Health & Medical Sciences (AREA)
  • Molecular Biology (AREA)
  • Biochemistry (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Medicinal Chemistry (AREA)
  • Biomedical Technology (AREA)
  • Biotechnology (AREA)
  • Biophysics (AREA)
  • Microbiology (AREA)
  • General Engineering & Computer Science (AREA)
  • Immunology (AREA)
  • Analytical Chemistry (AREA)
  • Toxicology (AREA)
  • Gastroenterology & Hepatology (AREA)
  • Cell Biology (AREA)
  • Pathology (AREA)
  • Physics & Mathematics (AREA)
  • Peptides Or Proteins (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)
  • Micro-Organisms Or Cultivation Processes Thereof (AREA)
  • Medicines Containing Antibodies Or Antigens For Use As Internal Diagnostic Agents (AREA)
  • Investigating Or Analysing Biological Materials (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
  • Measuring Or Testing Involving Enzymes Or Micro-Organisms (AREA)
  • Preparation Of Compounds By Using Micro-Organisms (AREA)

Abstract

La présente invention concerne de nouvelles protéines et, plus particulièrement, des molécules d'acide nucléique isolées codant de nouveaux polypeptides, ainsi que de nouveaux polypeptides et des anticorps qui se lient à ces polypeptides. L'invention concerne également des vecteurs, des cellules hôtes, et des méthodes recombinantes et synthétiques de production de polynucléotides et/ou de polypeptides humains et des anticorps. Elle concerne des méthodes diagnostiques et thérapeutiques servant à diagnostiquer, traiter, prévenir et/ou pronostiquer des troubles liés à ces nouveaux polypeptides. Elle concerne en outre des méthodes de criblage permettant d'identifier des agonistes et des antagonistes des polynucléotides et polypeptides de l'invention. L'invention concerne enfin des méthodes et/ou des compositions pouvant inhiber ou améliorer la production et la fonction des polypeptides de l'invention.
PCT/US2001/001358 2000-01-31 2001-01-17 Acides nucleiques, proteines, et anticorps WO2001055163A1 (fr)

Priority Applications (2)

Application Number Priority Date Filing Date Title
EP01918156A EP1254151A1 (fr) 2000-01-31 2001-01-17 Acides nucleiques, proteines, et anticorps
AU2001245262A AU2001245262A1 (en) 2000-01-31 2001-01-17 Nucleic acids, proteins, and antibodies

Applications Claiming Priority (235)

Application Number Priority Date Filing Date Title
US17906500P 2000-01-31 2000-01-31
US60/179,065 2000-01-31
US18062800P 2000-02-04 2000-02-04
US60/180,628 2000-02-04
US18466400P 2000-02-24 2000-02-24
US60/184,664 2000-02-24
US18635000P 2000-03-02 2000-03-02
US60/186,350 2000-03-02
US18987400P 2000-03-16 2000-03-16
US60/189,874 2000-03-16
US19007600P 2000-03-17 2000-03-17
US60/190,076 2000-03-17
US19812300P 2000-04-18 2000-04-18
US60/198,123 2000-04-18
US20551500P 2000-05-19 2000-05-19
US60/205,515 2000-05-19
US20946700P 2000-06-07 2000-06-07
US60/209,467 2000-06-07
US21488600P 2000-06-28 2000-06-28
US60/214,886 2000-06-28
US21513500P 2000-06-30 2000-06-30
US60/215,135 2000-06-30
US21688000P 2000-07-07 2000-07-07
US21664700P 2000-07-07 2000-07-07
US60/216,880 2000-07-07
US60/216,647 2000-07-07
US21749600P 2000-07-11 2000-07-11
US21748700P 2000-07-11 2000-07-11
US60/217,487 2000-07-11
US60/217,496 2000-07-11
US21829000P 2000-07-14 2000-07-14
US60/218,290 2000-07-14
US22096400P 2000-07-26 2000-07-26
US22096300P 2000-07-26 2000-07-26
US60/220,963 2000-07-26
US60/220,964 2000-07-26
US22575700P 2000-08-14 2000-08-14
US22451800P 2000-08-14 2000-08-14
US22575800P 2000-08-14 2000-08-14
US22451900P 2000-08-14 2000-08-14
US22526700P 2000-08-14 2000-08-14
US22527000P 2000-08-14 2000-08-14
US22521400P 2000-08-14 2000-08-14
US22575900P 2000-08-14 2000-08-14
US22544700P 2000-08-14 2000-08-14
US22521300P 2000-08-14 2000-08-14
US22526600P 2000-08-14 2000-08-14
US22526800P 2000-08-14 2000-08-14
US60/225,267 2000-08-14
US60/225,213 2000-08-14
US60/225,268 2000-08-14
US60/224,518 2000-08-14
US60/225,266 2000-08-14
US60/225,447 2000-08-14
US60/224,519 2000-08-14
US60/225,270 2000-08-14
US60/225,759 2000-08-14
US60/225,214 2000-08-14
US60/225,758 2000-08-14
US60/225,757 2000-08-14
US22627900P 2000-08-18 2000-08-18
US60/226,279 2000-08-18
US22686800P 2000-08-22 2000-08-22
US22718200P 2000-08-22 2000-08-22
US22668100P 2000-08-22 2000-08-22
US60/226,681 2000-08-22
US60/227,182 2000-08-22
US60/226,868 2000-08-22
US22700900P 2000-08-23 2000-08-23
US60/227,009 2000-08-23
US22892400P 2000-08-30 2000-08-30
US60/228,924 2000-08-30
US22934300P 2000-09-01 2000-09-01
US22928700P 2000-09-01 2000-09-01
US22934500P 2000-09-01 2000-09-01
US22934400P 2000-09-01 2000-09-01
US60/229,287 2000-09-01
US60/229,343 2000-09-01
US60/229,345 2000-09-01
US60/229,344 2000-09-01
US22951300P 2000-09-05 2000-09-05
US22950900P 2000-09-05 2000-09-05
US60/229,513 2000-09-05
US60/229,509 2000-09-05
US23043700P 2000-09-06 2000-09-06
US23043800P 2000-09-06 2000-09-06
US60/230,437 2000-09-06
US60/230,438 2000-09-06
US23124400P 2000-09-08 2000-09-08
US23124300P 2000-09-08 2000-09-08
US23141300P 2000-09-08 2000-09-08
US23124200P 2000-09-08 2000-09-08
US23208100P 2000-09-08 2000-09-08
US23208000P 2000-09-08 2000-09-08
US23141400P 2000-09-08 2000-09-08
US60/231,244 2000-09-08
US60/231,413 2000-09-08
US60/232,080 2000-09-08
US60/231,414 2000-09-08
US60/231,242 2000-09-08
US60/231,243 2000-09-08
US60/232,081 2000-09-08
US23196800P 2000-09-12 2000-09-12
US60/231,968 2000-09-12
US23239700P 2000-09-14 2000-09-14
US23240000P 2000-09-14 2000-09-14
US23240100P 2000-09-14 2000-09-14
US23239800P 2000-09-14 2000-09-14
US23306500P 2000-09-14 2000-09-14
US23306400P 2000-09-14 2000-09-14
US23306300P 2000-09-14 2000-09-14
US23239900P 2000-09-14 2000-09-14
US60/232,400 2000-09-14
US60/233,064 2000-09-14
US60/232,397 2000-09-14
US60/232,401 2000-09-14
US60/232,399 2000-09-14
US60/232,398 2000-09-14
US60/233,065 2000-09-14
US60/233,063 2000-09-14
US23427400P 2000-09-21 2000-09-21
US23422300P 2000-09-21 2000-09-21
US60/234,223 2000-09-21
US60/234,274 2000-09-21
US23499700P 2000-09-25 2000-09-25
US23499800P 2000-09-25 2000-09-25
US60/234,998 2000-09-25
US60/234,997 2000-09-25
US23548400P 2000-09-26 2000-09-26
US60/235,484 2000-09-26
US23583600P 2000-09-27 2000-09-27
US23583400P 2000-09-27 2000-09-27
US60/235,836 2000-09-27
US60/235,834 2000-09-27
US23636700P 2000-09-29 2000-09-29
US23636800P 2000-09-29 2000-09-29
US23637000P 2000-09-29 2000-09-29
US23632700P 2000-09-29 2000-09-29
US23636900P 2000-09-29 2000-09-29
US60/236,367 2000-09-29
US60/236,327 2000-09-29
US60/236,368 2000-09-29
US60/236,370 2000-09-29
US60/236,369 2000-09-29
US23703800P 2000-10-02 2000-10-02
US23680200P 2000-10-02 2000-10-02
US23703700P 2000-10-02 2000-10-02
US23704000P 2000-10-02 2000-10-02
US23703900P 2000-10-02 2000-10-02
US60/237,038 2000-10-02
US60/237,040 2000-10-02
US60/237,037 2000-10-02
US60/237,039 2000-10-02
US60/236,802 2000-10-02
US23993500P 2000-10-13 2000-10-13
US23993700P 2000-10-13 2000-10-13
US60/239,937 2000-10-13
US60/239,935 2000-10-13
US24122100P 2000-10-20 2000-10-20
US24182600P 2000-10-20 2000-10-20
US24180900P 2000-10-20 2000-10-20
US24178500P 2000-10-20 2000-10-20
US24180800P 2000-10-20 2000-10-20
US24178700P 2000-10-20 2000-10-20
US24178600P 2000-10-20 2000-10-20
US24096000P 2000-10-20 2000-10-20
US60/241,809 2000-10-20
US60/241,786 2000-10-20
US60/241,808 2000-10-20
US60/241,826 2000-10-20
US60/241,785 2000-10-20
US60/241,221 2000-10-20
US60/240,960 2000-10-20
US60/241,787 2000-10-20
US24461700P 2000-11-01 2000-11-01
US60/244,617 2000-11-01
US24652600P 2000-11-08 2000-11-08
US24652500P 2000-11-08 2000-11-08
US24652700P 2000-11-08 2000-11-08
US24647600P 2000-11-08 2000-11-08
US24661100P 2000-11-08 2000-11-08
US24647500P 2000-11-08 2000-11-08
US24647400P 2000-11-08 2000-11-08
US24652800P 2000-11-08 2000-11-08
US24647700P 2000-11-08 2000-11-08
US24661000P 2000-11-08 2000-11-08
US24653200P 2000-11-08 2000-11-08
US60/246,525 2000-11-08
US60/246,475 2000-11-08
US60/246,526 2000-11-08
US60/246,609 2000-11-08
US60/246,474 2000-11-08
US60/246,611 2000-11-08
US60/246,527 2000-11-08
US60/246,613 2000-11-08
US60/246,478 2000-11-08
US60/246,477 2000-11-08
US60/246,476 2000-11-08
US60/246,532 2000-11-08
US60/246,528 2000-11-08
US60/246,610 2000-11-08
US60/246,524 2000-11-08
US60/246,523 2000-11-08
US60/249,211 2000-11-17
US60/249,244 2000-11-17
US60/249,212 2000-11-17
US60/249,214 2000-11-17
US60/249,265 2000-11-17
US60/249,213 2000-11-17
US60/249,217 2000-11-17
US60/249,264 2000-11-17
US60/249,216 2000-11-17
US60/249,208 2000-11-17
US60/249,245 2000-11-17
US60/249,207 2000-11-17
US60/249,300 2000-11-17
US60/249,299 2000-11-17
US60/249,218 2000-11-17
US60/249,215 2000-11-17
US60/249,297 2000-11-17
US60/249,209 2000-11-17
US60/249,210 2000-11-17
US60/250,160 2000-12-01
US60/250,391 2000-12-01
US60/251,988 2000-12-05
US60/251,030 2000-12-05
US60/256,719 2000-12-05
US60/251,479 2000-12-06
US60/251,869 2000-12-08
US60/251,856 2000-12-08
US60/251,990 2000-12-08
US60/251,989 2000-12-08
US60/251,868 2000-12-08
US60/254,097 2000-12-11
US60/259,678 2001-01-05

Publications (2)

Publication Number Publication Date
WO2001055163A1 true WO2001055163A1 (fr) 2001-08-02
WO2001055163A8 WO2001055163A8 (fr) 2001-09-07

Family

ID=27587117

Family Applications (48)

Application Number Title Priority Date Filing Date
PCT/US2001/001351 WO2001055355A1 (fr) 2000-01-31 2001-01-17 Acides nucleiques, proteines et anticorps
PCT/US2001/001340 WO2001055321A2 (fr) 2000-01-31 2001-01-17 Acides nucleiques, proteines et anticorps
PCT/US2001/001309 WO2001055308A2 (fr) 2000-01-31 2001-01-17 Acides nucleiques, proteines et anticorps
PCT/US2001/001333 WO2001055448A1 (fr) 2000-01-31 2001-01-17 Acides nucleiques, proteines et anticorps
PCT/US2001/001348 WO2001055368A1 (fr) 2000-01-31 2001-01-17 Acides nucleiques, proteines et anticorps
PCT/US2001/001343 WO2001055323A2 (fr) 2000-01-31 2001-01-17 Acides ncleiques, proteines et anticorps
PCT/US2001/001355 WO2001055207A1 (fr) 2000-01-31 2001-01-17 Acides nucleiques, proteines et anticorps
PCT/US2001/001322 WO2001055343A1 (fr) 2000-01-31 2001-01-17 Acides nucleiques, proteines et anticorps
PCT/US2001/001327 WO2001055203A1 (fr) 2000-01-31 2001-01-17 Acides nucleiques, proteines et anticorps
PCT/US2001/001312 WO2001054733A1 (fr) 2000-01-31 2001-01-17 Acides nucleiques, proteines et anticorps
PCT/US2001/001314 WO2001055310A2 (fr) 2000-01-31 2001-01-17 Acides nucleiques, proteines et anticorps
PCT/US2001/001326 WO2001055315A2 (fr) 2000-01-31 2001-01-17 Acides nucleiques, proteines, et anticorps
PCT/US2001/001316 WO2001054473A2 (fr) 2000-01-31 2001-01-17 Acides nucleiques, proteines et anticorps
PCT/US2001/001325 WO2001055202A1 (fr) 2000-01-31 2001-01-17 Acides nucleiques, proteines et anticorps
PCT/US2001/001359 WO2001055328A2 (fr) 2000-01-31 2001-01-17 Acides nucleiques, proteines et anticorps
PCT/US2001/001301 WO2001055303A2 (fr) 2000-01-31 2001-01-17 Acides nucleiques, proteines et anticorps
PCT/US2001/001302 WO2001055304A2 (fr) 2000-01-31 2001-01-17 Acides nucleiques, proteines et anticorps
PCT/US2001/001240 WO2001055302A2 (fr) 2000-01-31 2001-01-17 Acides nucleiques, proteines et anticorps
PCT/US2001/001353 WO2001055206A1 (fr) 2000-01-31 2001-01-17 Acides nucleiques, proteines et anticorps
PCT/US2001/001352 WO2001055327A2 (fr) 2000-01-31 2001-01-17 Acides nucleiques, proteines et anticorps
PCT/US2001/001356 WO2001055173A2 (fr) 2000-01-31 2001-01-17 Acides nucleiques, proteines et anticorps
PCT/US2001/001349 WO2001054474A2 (fr) 2000-01-31 2001-01-17 Acides nucleiques, proteines et anticorps
PCT/US2001/001338 WO2001055367A1 (fr) 2000-01-31 2001-01-17 Acides nucleiques, proteines et antigenes
PCT/US2001/001335 WO2001055319A2 (fr) 2000-01-31 2001-01-17 Acides nucleiques, proteines et anticorps
PCT/US2001/001344 WO2001055324A2 (fr) 2000-01-31 2001-01-17 Acides nucleiques, proteines et anticorps
PCT/US2001/001315 WO2001055311A2 (fr) 2000-01-31 2001-01-17 Acides nucleiques, proteines et anticorps
PCT/US2001/001317 WO2001055201A1 (fr) 2000-01-31 2001-01-17 Acides nucleiques, proteines et anticorps
PCT/US2001/001306 WO2001055307A2 (fr) 2000-01-31 2001-01-17 Acides nucleiques, proteines et anticorps
PCT/US2001/001336 WO2001055204A1 (fr) 2000-01-31 2001-01-17 Acides nucleiques, proteines et anticorps
PCT/US2001/001313 WO2001055200A1 (fr) 2000-01-31 2001-01-17 Acides nucleiques, proteines et anticorps
PCT/US2001/001332 WO2001055318A2 (fr) 2000-01-31 2001-01-17 Acides nucleiques, proteines et anticorps
PCT/US2001/001357 WO2001055208A1 (fr) 2000-01-31 2001-01-17 Acides nucleiques, proteines et anticorps
PCT/US2001/001342 WO2001059064A2 (fr) 2000-01-31 2001-01-17 Acides nucleiques, proteines et anticorps
PCT/US2001/001311 WO2001055309A2 (fr) 2000-01-31 2001-01-17 Acides nucleiques, proteines et anticorps
PCT/US2001/001358 WO2001055163A1 (fr) 2000-01-31 2001-01-17 Acides nucleiques, proteines, et anticorps
PCT/US2001/001330 WO2001055447A1 (fr) 2000-01-31 2001-01-17 Acides nucléiques, proteines et anticorps
PCT/US2001/001354 WO2001057182A2 (fr) 2000-01-31 2001-01-17 Acides nucleiques, proteines et anticorps
PCT/US2001/001310 WO2001055387A1 (fr) 2000-01-31 2001-01-17 Acides nucleiques, proteines et anticorps
PCT/US2001/001321 WO2001055312A2 (fr) 2000-01-31 2001-01-17 Acides nucleiques, proteines et anticorps
PCT/US2001/001308 WO2001055364A2 (fr) 2000-01-31 2001-01-17 Acides nucléiques, protéines et anticorps
PCT/US2001/001341 WO2001055322A2 (fr) 2000-01-31 2001-01-17 Acides nucleiques, proteines et anticorps
PCT/US2001/001239 WO2001055301A2 (fr) 2000-01-31 2001-01-17 Acides nucleiques, proteines et anticorps
PCT/US2001/001328 WO2001055316A2 (fr) 2000-01-31 2001-01-17 Acides nucleiques, proteines et anticorps
PCT/US2001/001350 WO2001055350A1 (fr) 2000-01-31 2001-01-17 Acides nucleiques, proteines et anticorps
PCT/US2001/001324 WO2001055314A2 (fr) 2000-01-31 2001-01-17 Acides nucleiques, proteines et anticorps
PCT/US2001/001337 WO2001055205A1 (fr) 2000-01-31 2001-01-17 Acides nucleiques, proteines et anticorps
PCT/US2001/001345 WO2001055325A2 (fr) 2000-01-31 2001-01-17 Acides nucleiques, proteines et anticorps
PCT/US2001/001339 WO2001055320A2 (fr) 2000-01-31 2001-01-17 Acides nucleiques, proteines et anticorps

Family Applications Before (34)

Application Number Title Priority Date Filing Date
PCT/US2001/001351 WO2001055355A1 (fr) 2000-01-31 2001-01-17 Acides nucleiques, proteines et anticorps
PCT/US2001/001340 WO2001055321A2 (fr) 2000-01-31 2001-01-17 Acides nucleiques, proteines et anticorps
PCT/US2001/001309 WO2001055308A2 (fr) 2000-01-31 2001-01-17 Acides nucleiques, proteines et anticorps
PCT/US2001/001333 WO2001055448A1 (fr) 2000-01-31 2001-01-17 Acides nucleiques, proteines et anticorps
PCT/US2001/001348 WO2001055368A1 (fr) 2000-01-31 2001-01-17 Acides nucleiques, proteines et anticorps
PCT/US2001/001343 WO2001055323A2 (fr) 2000-01-31 2001-01-17 Acides ncleiques, proteines et anticorps
PCT/US2001/001355 WO2001055207A1 (fr) 2000-01-31 2001-01-17 Acides nucleiques, proteines et anticorps
PCT/US2001/001322 WO2001055343A1 (fr) 2000-01-31 2001-01-17 Acides nucleiques, proteines et anticorps
PCT/US2001/001327 WO2001055203A1 (fr) 2000-01-31 2001-01-17 Acides nucleiques, proteines et anticorps
PCT/US2001/001312 WO2001054733A1 (fr) 2000-01-31 2001-01-17 Acides nucleiques, proteines et anticorps
PCT/US2001/001314 WO2001055310A2 (fr) 2000-01-31 2001-01-17 Acides nucleiques, proteines et anticorps
PCT/US2001/001326 WO2001055315A2 (fr) 2000-01-31 2001-01-17 Acides nucleiques, proteines, et anticorps
PCT/US2001/001316 WO2001054473A2 (fr) 2000-01-31 2001-01-17 Acides nucleiques, proteines et anticorps
PCT/US2001/001325 WO2001055202A1 (fr) 2000-01-31 2001-01-17 Acides nucleiques, proteines et anticorps
PCT/US2001/001359 WO2001055328A2 (fr) 2000-01-31 2001-01-17 Acides nucleiques, proteines et anticorps
PCT/US2001/001301 WO2001055303A2 (fr) 2000-01-31 2001-01-17 Acides nucleiques, proteines et anticorps
PCT/US2001/001302 WO2001055304A2 (fr) 2000-01-31 2001-01-17 Acides nucleiques, proteines et anticorps
PCT/US2001/001240 WO2001055302A2 (fr) 2000-01-31 2001-01-17 Acides nucleiques, proteines et anticorps
PCT/US2001/001353 WO2001055206A1 (fr) 2000-01-31 2001-01-17 Acides nucleiques, proteines et anticorps
PCT/US2001/001352 WO2001055327A2 (fr) 2000-01-31 2001-01-17 Acides nucleiques, proteines et anticorps
PCT/US2001/001356 WO2001055173A2 (fr) 2000-01-31 2001-01-17 Acides nucleiques, proteines et anticorps
PCT/US2001/001349 WO2001054474A2 (fr) 2000-01-31 2001-01-17 Acides nucleiques, proteines et anticorps
PCT/US2001/001338 WO2001055367A1 (fr) 2000-01-31 2001-01-17 Acides nucleiques, proteines et antigenes
PCT/US2001/001335 WO2001055319A2 (fr) 2000-01-31 2001-01-17 Acides nucleiques, proteines et anticorps
PCT/US2001/001344 WO2001055324A2 (fr) 2000-01-31 2001-01-17 Acides nucleiques, proteines et anticorps
PCT/US2001/001315 WO2001055311A2 (fr) 2000-01-31 2001-01-17 Acides nucleiques, proteines et anticorps
PCT/US2001/001317 WO2001055201A1 (fr) 2000-01-31 2001-01-17 Acides nucleiques, proteines et anticorps
PCT/US2001/001306 WO2001055307A2 (fr) 2000-01-31 2001-01-17 Acides nucleiques, proteines et anticorps
PCT/US2001/001336 WO2001055204A1 (fr) 2000-01-31 2001-01-17 Acides nucleiques, proteines et anticorps
PCT/US2001/001313 WO2001055200A1 (fr) 2000-01-31 2001-01-17 Acides nucleiques, proteines et anticorps
PCT/US2001/001332 WO2001055318A2 (fr) 2000-01-31 2001-01-17 Acides nucleiques, proteines et anticorps
PCT/US2001/001357 WO2001055208A1 (fr) 2000-01-31 2001-01-17 Acides nucleiques, proteines et anticorps
PCT/US2001/001342 WO2001059064A2 (fr) 2000-01-31 2001-01-17 Acides nucleiques, proteines et anticorps
PCT/US2001/001311 WO2001055309A2 (fr) 2000-01-31 2001-01-17 Acides nucleiques, proteines et anticorps

Family Applications After (13)

Application Number Title Priority Date Filing Date
PCT/US2001/001330 WO2001055447A1 (fr) 2000-01-31 2001-01-17 Acides nucléiques, proteines et anticorps
PCT/US2001/001354 WO2001057182A2 (fr) 2000-01-31 2001-01-17 Acides nucleiques, proteines et anticorps
PCT/US2001/001310 WO2001055387A1 (fr) 2000-01-31 2001-01-17 Acides nucleiques, proteines et anticorps
PCT/US2001/001321 WO2001055312A2 (fr) 2000-01-31 2001-01-17 Acides nucleiques, proteines et anticorps
PCT/US2001/001308 WO2001055364A2 (fr) 2000-01-31 2001-01-17 Acides nucléiques, protéines et anticorps
PCT/US2001/001341 WO2001055322A2 (fr) 2000-01-31 2001-01-17 Acides nucleiques, proteines et anticorps
PCT/US2001/001239 WO2001055301A2 (fr) 2000-01-31 2001-01-17 Acides nucleiques, proteines et anticorps
PCT/US2001/001328 WO2001055316A2 (fr) 2000-01-31 2001-01-17 Acides nucleiques, proteines et anticorps
PCT/US2001/001350 WO2001055350A1 (fr) 2000-01-31 2001-01-17 Acides nucleiques, proteines et anticorps
PCT/US2001/001324 WO2001055314A2 (fr) 2000-01-31 2001-01-17 Acides nucleiques, proteines et anticorps
PCT/US2001/001337 WO2001055205A1 (fr) 2000-01-31 2001-01-17 Acides nucleiques, proteines et anticorps
PCT/US2001/001345 WO2001055325A2 (fr) 2000-01-31 2001-01-17 Acides nucleiques, proteines et anticorps
PCT/US2001/001339 WO2001055320A2 (fr) 2000-01-31 2001-01-17 Acides nucleiques, proteines et anticorps

Country Status (3)

Country Link
AU (16) AU2001241409A1 (fr)
CA (37) CA2395666A1 (fr)
WO (48) WO2001055355A1 (fr)

Cited By (14)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2002016587A2 (fr) * 2000-08-21 2002-02-28 Incyte Genomics, Inc. Proteines associees aux microtubules et tubulines
WO2002090543A2 (fr) * 2001-05-09 2002-11-14 Bayer Aktiengesellschaft Régulation de la protéine semblable à celle de type 2c de la phosphatase de l'acide phosphatidique humain
WO2002020765A3 (fr) * 2000-09-08 2003-03-06 Millennium Pharm Inc 38646, nouveau facteur d'echange de guanine nucleotide et ses utilisations
WO2002031132A3 (fr) * 2000-10-11 2003-03-13 Millennium Pharm Inc Nouvel element de la famille des phosphatases humaines a double specificite, appele 8843, et utilisation de ce dernier
WO2003024276A1 (fr) * 2000-08-02 2003-03-27 Millennium Pharmaceuticals,Inc. Membre de la famille du facteur de liberation du gtp humain (15368) et ses utilisations
WO2003057870A1 (fr) * 2002-01-07 2003-07-17 Bayer Healthcare Ag Proteine humaine analogue a la phosphatase de l'acide phosphatidique de type 2
WO2003080664A1 (fr) * 2002-03-21 2003-10-02 The Babraham Institute Proteine p-rex1, facteur d'echange guanine-nucleotide pour rac regule par ptdins (3, 4, 5) p3 et par g-beta-gamma-
US6774223B2 (en) 2000-06-28 2004-08-10 Diadexus, Inc. Method of diagnosing, monitoring, staging, imaging and treating colon cancer
EP1540014A2 (fr) * 2002-08-27 2005-06-15 Epigenomics AG Procede et acides nucleiques servant a l'analyse de troubles lies a la proliferation des cellules mammaires
WO2008030559A2 (fr) * 2006-09-08 2008-03-13 Corixa Corporation Procédés, compositions, et kits de détection et de surveillance d'un cancer du colon
EP1907589A2 (fr) * 2005-07-26 2008-04-09 Siemens Medical Solutions Diagnostics Polymorphismes de nucleotides uniques a susceptibilite par rapport a une maladie cardio-vasculaire
WO2009013484A1 (fr) * 2007-07-20 2009-01-29 Cancer Research Technology Limited Modulation immunitaire par lectine de type c
US10144934B2 (en) 2012-04-30 2018-12-04 Biocon Limited Targeted/TGF-ßRII fusion proteins and methods for making same
WO2021228999A1 (fr) * 2020-05-12 2021-11-18 Institut Curie Épitopes néo-antigéniques associés à des mutations sf3b1

Families Citing this family (257)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2002008288A2 (fr) * 2000-07-20 2002-01-31 Genentech, Inc. Polypeptides secretes et transmembranaires et acides nucleiques codant ces polypeptides
US20020137890A1 (en) 1997-03-31 2002-09-26 Genentech, Inc. Secreted and transmembrane polypeptides and nucleic acids encoding the same
US7579160B2 (en) 1998-03-18 2009-08-25 Corixa Corporation Methods for the detection of cervical cancer
US6960570B2 (en) 1998-03-18 2005-11-01 Corixa Corporation Compositions and methods for the therapy and diagnosis of lung cancer
US7258860B2 (en) 1998-03-18 2007-08-21 Corixa Corporation Compositions and methods for the therapy and diagnosis of lung cancer
US6706262B1 (en) 1998-03-18 2004-03-16 Corixa Corporation Compounds and methods for therapy and diagnosis of lung cancer
CA2328496C (fr) 1998-05-15 2016-01-05 Genentech, Inc. Polypeptides homologues de il-17 et utilisations therapeutiques de ceux-ci
US7771719B1 (en) 2000-01-11 2010-08-10 Genentech, Inc. Pharmaceutical compositions, kits, and therapeutic uses of antagonist antibodies to IL-17E
US6929938B2 (en) 2001-08-15 2005-08-16 Millennium Pharmaceuticals, Inc. 25501, a human transferase family member and uses therefor
WO2000050565A2 (fr) * 1999-02-25 2000-08-31 Thomas Jefferson University Compositions, kits et procedes concernant le gene fez1 de l'homme, nouveau gene suppresseur des tumeurs
US7141417B1 (en) 1999-02-25 2006-11-28 Thomas Jefferson University Compositions, kits, and methods relating to the human FEZ1 gene, a novel tumor suppressor gene
EP1157038A4 (fr) * 1999-02-26 2005-01-19 Smithkline Beecham Corp Clonage du recepteur 7tm (axor 17) du type p2y
WO2000056881A1 (fr) * 1999-03-23 2000-09-28 Human Genome Sciences, Inc. 48 proteines humaines secretees
CA2365601A1 (fr) 1999-03-29 2000-10-05 Kansai Technology Licensing Organization Co., Ltd. Nouvelle cytidine desaminase
EP1637541B1 (fr) * 1999-09-01 2010-03-03 Genentech, Inc. Polypeptides sécrétés et transmembranaires ainsi que les acides nucléiques codant pour ceux-ci
US20020151047A1 (en) * 2001-01-08 2002-10-17 Yi Hu Novel human protease and polynucleotides encoding the same
AU1238201A (en) * 1999-10-27 2001-05-08 Millennium Pharmaceuticals, Inc. Novel molecules and the card-related protein family and uses thereof
US6893818B1 (en) 1999-10-28 2005-05-17 Agensys, Inc. Gene upregulated in cancers of the prostate
US7005499B1 (en) * 1999-11-18 2006-02-28 Genentech, Inc. Wnt-regulated cytokine-like polypeptide and nucleic acids encoding same
EP1878795A3 (fr) * 1999-11-30 2008-01-23 Genentech, Inc. Compositions et procédés pour le traitement de maladies liées au système immunitaire
EP1250426A2 (fr) * 1999-12-01 2002-10-23 Genentech Inc. Polypeptides transmembranaires et secretes et les acides nucleiques codant ceux-ci
CN1300781A (zh) * 1999-12-22 2001-06-27 上海博德基因开发有限公司 一种新的多肽-人shc蛋白43和编码这种多肽的多核苷酸
US20040043397A1 (en) 2000-01-11 2004-03-04 Genentech, Inc. IL-17 homologous polypeptides and therapeutic uses thereof
AU2001237973B2 (en) 2000-01-26 2006-09-07 Agensys, Inc. 84p2a9: a prostate and testis specific protein highly expressed in prostate cancer
US6953682B2 (en) 2000-02-10 2005-10-11 Millennium Pharmaceuticals, Inc. Nucleic acid sequences encoding adenylate kinase, phospholipid scramblase-like, DNA fragmentation factor-like, phosphatidylserine synthase-like, and atpase-like molecules and uses therefor
US7078205B2 (en) 2000-02-17 2006-07-18 Millennium Pharmaceuticals, Inc. Nucleic acid sequences encoding melanoma associated antigen molecules, aminotransferase molecules, atpase molecules, acyltransferase molecules, pyridoxal-phosphate dependent enzyme molecules and uses therefor
US20020082212A1 (en) * 2000-07-20 2002-06-27 Millennium Pharmaceuticals, Inc. 7716, a novel human ATPase and uses therefor
US20020090705A1 (en) * 2000-07-14 2002-07-11 Rachel Meyers 62088, a novel human nucleoside phosphatase family member and uses thereof
US20020055159A1 (en) * 2000-06-15 2002-05-09 Meyers Rachel A. 23680,a novel human aminotransferase and uses therefor
US7718397B2 (en) 2000-03-21 2010-05-18 Genentech, Inc. Nucleic acids encoding receptor for IL-17 homologous polypeptides and uses thereof
AU2001250980A1 (en) * 2000-03-23 2001-10-03 Carmela Beger Brca-1 regulators and methods of use
AU2001245974A1 (en) 2000-03-24 2001-10-08 Millennium Pharmaceuticals, Inc. 3714, 16742, 23546, and 13887 novel protein kinase molecules and uses therefor
US20030198953A1 (en) * 2000-03-30 2003-10-23 Spytek Kimberly A. Novel proteins and nucleic acids encoding same
US7632929B2 (en) 2000-04-20 2009-12-15 The Board Of Trustees Of The University Of Arkansas Methamphetamine-like hapten compounds, linkers, carriers and compositions and uses thereof
JP2004511205A (ja) 2000-04-25 2004-04-15 レキシコン・ジェネティクス・インコーポレーテッド 新規ヒトキナーゼプロテインおよびそれをコードするポリヌクレオチド
AU2001257407A1 (en) * 2000-04-26 2001-11-07 Millennium Pharmaceuticals, Inc. 21657, a human short-chain dehydrogenase and uses thereof
US6808876B1 (en) * 2000-05-02 2004-10-26 Immusol, Inc. Cellular regulators of infectious agents and methods of use
WO2001085921A2 (fr) * 2000-05-12 2001-11-15 Merck Patent Gmbh Nouvelle serine-threonine kinase-4
EP1158001B1 (fr) * 2000-05-26 2007-11-14 F. Hoffmann-La Roche Ag Un acide nucléique produit excessivement dans des cellules tumorales humaines, une protéine codée par ledit acide nucléique et un procédé de diagnose de tumeurs
AU7634301A (en) * 2000-05-26 2001-12-03 Bayer Aktiengesellschaft Regulation of human p78-like serine/threonine kinase
DE10027170A1 (de) * 2000-05-31 2001-12-13 Schering Ag Humanes PEM als Target für die Fertilitätskontrolle
US7098311B2 (en) * 2000-06-02 2006-08-29 Brigham And Women's Hospital Fusion of jAZF1 and jjAZ1 genes in endometrial stromal tumors
US20030096969A1 (en) 2000-06-02 2003-05-22 Genentech, Inc. Secreted and transmembrane polypeptides and nucleic acids encoding the same
AU6531101A (en) * 2000-06-02 2001-12-17 Genentech Inc Secreted and transmembrane polypeptides and nucleic acids encoding the same
AU6604901A (en) * 2000-06-05 2001-12-17 Bayer Aktiengesellschaft Regulation of human hm74-like g protein coupled receptor
US6323016B1 (en) 2000-06-09 2001-11-27 Pe Corporation (Ny) Isolated human kinase proteins, nucleic acid molecules encoding human kinase proteins, and uses thereof
EP1287029A2 (fr) * 2000-06-09 2003-03-05 Corixa Corporation Compositions et procedes pour la therapie et le diagnostic du cancer du colon
WO2001096546A2 (fr) * 2000-06-16 2001-12-20 Incyte Genomics, Inc. Proteine-phosphatases
EP2075253A1 (fr) * 2000-06-23 2009-07-01 Genentech, Inc. Méthodes et composés pour la diagnose et le traitement de troubles associés à l'angiogenèse
WO2002000861A2 (fr) * 2000-06-26 2002-01-03 Millennium Pharmaceuticals, Inc. 46619, une nouvelle synthase beta-cetoacyle et utilisations de cette derniere
AU2001270215A1 (en) * 2000-06-27 2002-01-08 Curagen Corporation Novel polynucleotides and polypeptides encoded thereby
US7094587B2 (en) 2000-06-27 2006-08-22 Millennium Pharmaceuticals, Inc. 16002 Molecules and uses therefor
WO2002002761A2 (fr) * 2000-06-30 2002-01-10 Millennium Pharmaceuticals, Inc. 57658, nouvelle uridine kinase humaine et utilisations correspondantes
AU2001271843A1 (en) * 2000-07-07 2002-01-21 Incyte Genomics, Inc. Gtp-binding proteins
AU2001273239A1 (en) * 2000-07-07 2002-01-21 Incyte Genomics, Inc. Transporters and ion channels
EP1303620A2 (fr) * 2000-07-18 2003-04-23 Bayer Aktiengesellschaft Regulation de protease a serine humaine de type desc1
US6740751B2 (en) 2000-07-18 2004-05-25 Board Of Regents, The University Of Texas System Methods and compositions for stabilizing microtubules and intermediate filaments in striated muscle cells
AU8762901A (en) * 2000-07-25 2002-02-05 Merck Patent Gmbh Novel protein containing ring finger domaine r1p4
AU2001277202A1 (en) * 2000-07-26 2002-02-05 Applied Genomics, Inc. Bstp-ras/rerg protein and related reagents and methods of use thereof
AU2001277200A1 (en) * 2000-07-26 2002-02-05 Applied Genomics, Inc. Bstp-cad protein and related reagents and methods of use thereof
EP1657254A3 (fr) * 2000-08-01 2006-06-07 Genentech, Inc. Polypeptide, acides nucléiques le codant, et leur utilisation pour le diagnostic du cancer
US7335732B2 (en) 2000-08-01 2008-02-26 Genentech, Inc. PRO9799 polypeptides
WO2002010217A2 (fr) 2000-08-02 2002-02-07 The Johns Hopkins University Profils d'expression de cellules endotheliales
WO2002012330A2 (fr) * 2000-08-04 2002-02-14 Zymogenetics, Inc. Zzp1, proteine humaine secretee
US6943245B2 (en) 2000-08-10 2005-09-13 Board Of Regents, The University Of Texas System Tumor suppressor CAR-1
JP2004506422A (ja) * 2000-08-11 2004-03-04 メルク パテント ゲゼルシャフト ミット ベシュレンクテル ハフトング 新規マイトジェン活性化キナーゼ
EP1326985A1 (fr) * 2000-08-18 2003-07-16 MERCK PATENT GmbH Identification d'un gene d'acetyltransferase n-terminale humaine
US6706513B2 (en) 2000-08-21 2004-03-16 Bristol-Myers Squibb Company Adenosine deaminase homolog
WO2002016579A2 (fr) * 2000-08-24 2002-02-28 Eli Lilly And Company Acides nucleiques, vecteurs, cellules hotes, polypeptides et utilisations de ces derniers
CA2705366A1 (fr) * 2000-08-28 2002-03-07 Agensys, Inc. Acide nucleique et proteine correspondante denommes 85p1b3 utilises dans le traitement et la detection de cancers
CA2420902A1 (fr) * 2000-08-28 2002-03-07 Craig A. Rosen 18 proteines humaines secretees
AU2001284176A1 (en) * 2000-08-28 2002-03-13 Astrazeneca Ab Molecules involved in the regulation of insulin in resistance syndrome (irs)
AU2002210464A1 (en) * 2000-08-30 2002-03-13 Bayer Aktiengesellschaft Regulation of human aminotransferase-like enzyme
WO2002018577A2 (fr) * 2000-08-30 2002-03-07 Millennium Pharmaceuticals, Inc. Nouveau facteur de terminaison du gtp humain 48921 et ses utilisations
WO2002018552A2 (fr) * 2000-08-30 2002-03-07 Bayer Aktiengesellschaft Regulation d'enzyme humaine de type aminotransferase
AU2001286714A1 (en) * 2000-08-30 2002-03-13 Millennium Pharmaceuticals, Inc. 54370, a novel human sulfate transporter and uses therefor
WO2002018582A2 (fr) * 2000-08-31 2002-03-07 Millennium Pharmaceuticals, Inc. 62112, nouvelle deshydrogenase humaine et ses utilisations
US7153669B2 (en) 2000-09-05 2006-12-26 Amgen Inc. Nucleic acids encoding MK61 receptors
CA2421265A1 (fr) * 2000-09-05 2002-03-14 Incyte Genomics, Inc. Molecules utilisees a des fins diagnostiques et therapeutiques
US6372468B1 (en) * 2000-09-14 2002-04-16 Pe Corporation (Ny) Isolated human kinase proteins, nucleic acid molecules encoding human kinase proteins, and uses thereof
US6391606B1 (en) * 2000-09-14 2002-05-21 Pe Corporation Isolated human phospholipase proteins, nucleic acid molecules encoding human phospholipase proteins, and uses thereof
US20040048245A1 (en) * 2000-10-05 2004-03-11 Shimkets Richard A. Novel human proteins, polynucleotides encoding them and methods of using the same
WO2002031126A2 (fr) * 2000-10-11 2002-04-18 Bayer Aktiengesellschaft Regulation d'une peptidyl-prolyl cis-trans isomerase de type cyclophiline humaine
US20020064831A1 (en) * 2000-10-12 2002-05-30 The Texas A&M University System Nucleic acid sequences encoding CMG proteins, CMG proteins, and methods for their use
EP1474528A4 (fr) * 2000-10-13 2006-06-14 Protein Design Labs Inc Procedes de diagnostic du cancer de la prostate, compositions et procedes de criblage de modulateurs du cancer de la prostate
US6531297B2 (en) * 2000-10-20 2003-03-11 Applera Corporation Isolated human drug-metabolizing proteins, nucleic acid molecules encoding human drug-metabolizing proteins, and uses thereof
WO2002062945A2 (fr) 2000-10-25 2002-08-15 Diadexus, Inc. Compositions et techniques relatives a des genes et a des proteines specifiques du poumon
WO2002036781A2 (fr) * 2000-10-31 2002-05-10 Bayer Aktiengesellschaft Regulation de la glutathione-s-transferase humaine
WO2002038743A2 (fr) * 2000-11-09 2002-05-16 Glaxo Group Limited Enzyme
EP1349934A2 (fr) 2000-11-20 2003-10-08 Diadexus, Inc. Compositions et methodes relatives a des genes et des proteines specifiques aux seins
AU2001297765A1 (en) * 2000-12-05 2002-10-21 Incyte Genomics, Inc. Ligases
US7776523B2 (en) 2000-12-07 2010-08-17 Novartis Vaccines And Diagnostics, Inc. Endogenous retroviruses up-regulated in prostate cancer
EP1339845A2 (fr) 2000-12-07 2003-09-03 ZymoGenetics, Inc. Proteine associee au complement adipocyte zacrp3x2
WO2002048328A2 (fr) * 2000-12-14 2002-06-20 Pe Corporation (Ny) Proteines kinases humaines isolees, molecules d'acide nucleique codant des proteines kinases humaines et utilisations associees
US20020142376A1 (en) * 2000-12-20 2002-10-03 Gennady Merkulov Isolated human transporter proteins, nucleic acid molecules encoding human transporter proteins, and uses thereof
IL155687A0 (en) 2000-12-22 2003-11-23 Bristol Myers Squibb Co Polynucleotides encoding hlrrsi1 polypeptides
US7892730B2 (en) 2000-12-22 2011-02-22 Sagres Discovery, Inc. Compositions and methods for cancer
US6423521B1 (en) 2000-12-28 2002-07-23 Pe Corporation (Ny) Isolated human kinase proteins, nucleic acid molecules encoding human kinase proteins, and uses thereof
WO2002053591A1 (fr) * 2000-12-30 2002-07-11 Lion Bioscience Ag Cofacteur de recepteur nucleaire mammalien cf12 et ses procedes d'utilisation
US20030219739A1 (en) * 2001-01-30 2003-11-27 Glass David J. Novel nucleic acid and polypeptide molecules
WO2002098917A2 (fr) * 2001-02-12 2002-12-12 Curagen Corporation Proteines, polynucleotides codant pour lesdites proteines et methodes d'utilisation desdites proteines
EP1637601A3 (fr) * 2001-02-21 2006-03-29 Curagen Corporation Protéines , polynucléotides codant pour ces protéines et procédés d'utilisation
US7271240B2 (en) 2001-03-14 2007-09-18 Agensys, Inc. 125P5C8: a tissue specific protein highly expressed in various cancers
AU2002250143A1 (en) * 2001-03-16 2002-10-03 Eli Lilly And Company Lp mammalian proteins; related reagents
US6913904B2 (en) * 2001-03-27 2005-07-05 Applera Corporation Isolated human Ras-like proteins, nucleic acid molecules encoding these human Ras-like proteins, and uses thereof
AU2002258626B2 (en) 2001-04-10 2007-01-18 Agensys, Inc. Nucleid acid and corresponding protein entitled 158P3D2 useful in treatment and detection of cancer
EP2280030A3 (fr) 2001-04-10 2011-06-15 Agensys, Inc. Acides nucléiques et protéines correspondantes utiles dans la détection et le traitement des différents cancers
EP1573022B1 (fr) 2001-04-10 2011-06-08 Agensys, Inc. Acide nucleique et proteine correspondante intitulee 184p1e2, utilises dans le traitement et la detection de cancers
WO2002086113A2 (fr) * 2001-04-24 2002-10-31 Isis Innovation Ltd Enzyme et marqueur snp pour le depistage d'une maladie
JPWO2002088175A1 (ja) * 2001-04-24 2004-08-19 大塚製薬株式会社 クローン病抗体結合性ペプチド及びクローン病の検査方法
KR100882762B1 (ko) 2001-05-07 2009-02-09 시오노기세이야쿠가부시키가이샤 혈관신생 마커로서 사용되는 폴리펩티드 및 이의 dna
US6905827B2 (en) 2001-06-08 2005-06-14 Expression Diagnostics, Inc. Methods and compositions for diagnosing or monitoring auto immune and chronic inflammatory diseases
US7235358B2 (en) * 2001-06-08 2007-06-26 Expression Diagnostics, Inc. Methods and compositions for diagnosing and monitoring transplant rejection
WO2003000901A2 (fr) * 2001-06-26 2003-01-03 Decode Genetics Ehf. Acides nucleiques codant des proteines kinases
US20030120040A1 (en) 2001-06-29 2003-06-26 Genentech, Inc. Secreted and Transmembrane polypeptides and nucleic acids encoding the same
GB2399086A (en) * 2001-08-02 2004-09-08 Aeomica Inc Human zinc finger containing gene MDZ4
WO2003031568A2 (fr) * 2001-08-17 2003-04-17 Incyte Genomics, Inc. Molecules de signalisation intracellulaire
US7270960B2 (en) 2001-08-29 2007-09-18 Pacific Northwest Research Institute Diagnosis of ovarian carcinomas
WO2003023063A1 (fr) * 2001-09-07 2003-03-20 Sankyo Company, Limited Methode d'estimation du risque d'apparition de diabetes
WO2003025175A2 (fr) * 2001-09-17 2003-03-27 Molecular Engines Laboratories Sequences impliquees dans les phenomenes de suppression tumorale, reversion tumorale, apoptose et/ou resistance aux virus et leur utilisation comme medicaments
WO2003040369A2 (fr) * 2001-09-17 2003-05-15 Molecular Engines Laboratories Sequences impliquees dans les phenomenes de suppression tumorale, reversion tumorale, apoptose et/ou resistance aux virus et leur utilisation comme medicaments
CA2417455C (fr) 2001-09-28 2008-01-29 Brigham Young University Variants nouveaux de la cyclooxygenase et methodes d'utilisation
WO2003038087A1 (fr) 2001-10-04 2003-05-08 Kansai Technology Licensing Organization Co., Ltd. Promoteur du gene dr5 et promoteur du gene siah-1
US7084257B2 (en) 2001-10-05 2006-08-01 Amgen Inc. Fully human antibody Fab fragments with human interferon-gamma neutralizing activity
WO2003031607A1 (fr) * 2001-10-10 2003-04-17 Bayer Healthcare Ag Regulation de deshydrogenase/reductase des chaines courtes humaine
DE60220621T2 (de) 2001-11-09 2008-03-06 Proteologics, Inc. Posh Nukleinsäure, Polypeptide und darauf bezogene Verfahren
AU2002365162A1 (en) 2001-11-14 2003-07-09 Northwestern University Self-assembly and mineralization of peptide-amphiphile nanofibers
CA2468145A1 (fr) * 2001-11-21 2003-05-30 Japan Science And Technology Agency Facteurs de transcription de gene de la choree de huntington
US7151162B2 (en) 2001-12-06 2006-12-19 The University Of Children's Hospital Of Both Cantons Of Basel Nuclear protein
AU2002339697A1 (en) * 2001-12-19 2003-06-30 Genset S.A. Gmg-5 polynucleotides and polypeptides and uses thereof
JP2003245084A (ja) * 2001-12-20 2003-09-02 Morinaga Milk Ind Co Ltd 脳梁又は精子の形成不全の診断及び治療に有用な新規遺伝子、並びにその用途
DE10163467A1 (de) * 2001-12-21 2003-11-27 Axaron Bioscience Ag Protein 24B2 und zugrundliegende DNA-Sequenz
US6759222B2 (en) 2002-01-02 2004-07-06 Millennium Pharmaceuticals, Inc. 14815, a human kinase family member and uses therefor
AU2003235631A1 (en) * 2002-01-15 2003-07-30 Bayer Aktiengesellschaft Human subtilisin/kexin-like convertase
AU2003205611A1 (en) * 2002-01-15 2003-07-30 Medigene Ag Dilated cardiomyopathy associated gene-2 (dcmag-2): a cytoplasmatic inducer of sarcomeric remodeling in cardiomyocytes
WO2003062429A1 (fr) * 2002-01-23 2003-07-31 Yamanouchi Pharmaceutical Co., Ltd. Nouvelle serine protease
US7371719B2 (en) 2002-02-15 2008-05-13 Northwestern University Self-assembly of peptide-amphiphile nanofibers under physiological conditions
EP1338609A1 (fr) * 2002-02-21 2003-08-27 MEMOREC Stoffel GmbH-Medizinisch-Molekulare Entwicklung, Köln Un outre récepteur KDR et son utilisation
CA2478271A1 (fr) * 2002-03-05 2003-09-18 Applera Corporation Proteines transporteuses humaines isolees, molecules d'acide nucleique codant celles-ci, et utilisations associees
GB2388113A (en) * 2002-03-06 2003-11-05 Oxford Glycosciences B-cell malignancy-associated protein
DK1918386T3 (da) 2002-03-13 2012-01-02 Genomic Health Inc Genekspressionsprofiler i biopsier af tumorvæv
AU2003220913A1 (en) * 2002-03-19 2003-09-29 Tanabe Seiyaku Co., Ltd. Novel g protein-coupled recepotrs and genes thereof
US7527935B2 (en) 2002-03-19 2009-05-05 Mitsubishi Tanabe Pharma Corporation G-protein coupled receptor having eicosanoid as ligand and gene thereof
US7193069B2 (en) 2002-03-22 2007-03-20 Research Association For Biotechnology Full-length cDNA
US7329502B2 (en) 2002-04-25 2008-02-12 The United States Of America As Represented By The Department Of Health And Human Services ZAP-70 expression as a marker for chronic lymphocytic leukemia/small lymphocytic lymphoma (CLL/SLL)
JP2004041003A (ja) * 2002-05-17 2004-02-12 Takeda Chem Ind Ltd 新規タンパク質、そのdnaおよびその用途
EP2275118A3 (fr) 2002-05-29 2011-10-19 DeveloGen Aktiengesellschaft Proteines specifiques du pancreas
AU2003237367A1 (en) 2002-06-03 2003-12-19 Chiron Corporation Use of nrg4, or inhibitors thereof, in the treatment of colon and pancreatic cancer
DK1513934T3 (da) 2002-06-06 2011-05-02 Oncotherapy Science Inc Gener og polypeptider relateret til humane coloncancersygdomme
JP2005535308A (ja) 2002-06-13 2005-11-24 カイロン コーポレイション Hml−2ポリペプチド発現用ベクター
AU2003281626A1 (en) * 2002-07-22 2004-02-09 Masayoshi Harigai Novel gene associated with rheumatoid arthritis
JP4186004B2 (ja) 2002-08-14 2008-11-26 独立行政法人産業技術総合研究所 新規n−アセチルガラクトサミン転移酵素およびそれをコードする核酸
AU2003243151A1 (en) * 2002-08-16 2004-03-03 Agensys, Inc. Nucleic acid and corresponding protein entitled 251p5g2 useful in treatment and detection of cancer
WO2004018518A1 (fr) * 2002-08-23 2004-03-04 Japan Science And Technology Agency Peptides antigeniques de tumeur cancereuse solide humains, polynucleotides codant pour ceux-ci et utilisation de ceux-ci
US7452969B2 (en) 2002-08-30 2008-11-18 Licentia Ltd Neurotrophic factor protein and uses thereof
CN1703524B (zh) * 2002-09-30 2010-04-07 肿瘤疗法科学股份有限公司 与人胰腺癌相关的基因和多肽
EP1552310A2 (fr) * 2002-10-17 2005-07-13 EVOTEC Neurosciences GmbH L'utilisation de la proteine et du gene de la nicotinamide mononucleotide adenylyltransferase 2 (nmnat-2) pour le diagnostique et la therapie des maladies neurodegeneratives
AU2002952216A0 (en) * 2002-10-23 2002-11-07 The Walter And Eliza Hall Institute Of Medical Research Novel therapeutic molecules
US7554021B2 (en) 2002-11-12 2009-06-30 Northwestern University Composition and method for self-assembly and mineralization of peptide amphiphiles
EP1570080A4 (fr) 2002-11-15 2006-03-01 Genomic Health Inc Etablissement de profils d'expressions genetique du cancer a recepteur de facteur de croissance epidermique positif
DE10254601A1 (de) 2002-11-22 2004-06-03 Ganymed Pharmaceuticals Ag Differentiell in Tumoren exprimierte Genprodukte und deren Verwendung
AU2002953341A0 (en) * 2002-12-13 2003-01-09 The Walter And Eliza Hall Institute Of Medical Research A novel phosphoprotein
AU2003287764B2 (en) * 2002-12-13 2010-01-21 The Walter And Eliza Hall Institute Of Medical Research A novel phosphoprotein
US20040231909A1 (en) 2003-01-15 2004-11-25 Tai-Yang Luh Motorized vehicle having forward and backward differential structure
WO2004070025A2 (fr) * 2003-02-05 2004-08-19 Juan Saus Nouvelles isoformes de la proteine liant l'antigene de goodpasture et troubles induits par une proteine incorrectement repliee
EP1445614A1 (fr) * 2003-02-06 2004-08-11 Institut National De La Sante Et De La Recherche Medicale (Inserm) Méthodes d'évaluation in vitro de l'état de progression d'un virus HIV chez un individu
US7074891B2 (en) * 2003-02-07 2006-07-11 Posco Leukocyte stimulating peptides
AU2004213871B9 (en) 2003-02-20 2009-09-03 Genomic Health, Inc. Use of intronic RNA to measure gene expression
WO2004078784A1 (fr) * 2003-03-04 2004-09-16 Astellas Pharma Inc. Nouveau gene associe a des conditions fibreuses
PL378582A1 (pl) 2003-03-19 2006-05-02 Biogen Idec Ma Inc. Białko wiążące receptor Nogo
ITRM20030149A1 (it) 2003-04-02 2004-10-03 Giuliani Spa Oligonucleotidi (odn) antisenso per smad7 e loro usi in campo medico
EP3470535B1 (fr) 2003-06-24 2020-04-01 Genomic Health, Inc. Prédiction de probabilité de récurrence de cancer
US7526387B2 (en) 2003-07-10 2009-04-28 Genomic Health, Inc. Expression profile algorithm and test for cancer prognosis
ATE528397T1 (de) 2003-08-08 2011-10-15 Perseus Proteomics Inc Bei krebs überexprimiertes gen
US20050136434A1 (en) * 2003-08-12 2005-06-23 Mai Xu Isolated heat-inducible cell surface protein and hyperthermia-based tumor immunotargeting therapy
US20070178458A1 (en) * 2003-09-05 2007-08-02 O'brien Philippa Methods of diagnosis and prognosis of ovarian cancer II
TW200517503A (en) * 2003-09-10 2005-06-01 Japan Science & Tech Agency Group of genes differentially expressed in peripheral blood cells and diagnostic method and assay method using the same
EP1667712B1 (fr) * 2003-10-02 2010-07-21 GlaxoSmithKline Biologicals S.A. Antigenes de b. pertussis et leur utilisation dans la vaccination
CA2541006C (fr) 2003-10-03 2015-02-17 Brigham And Women's Hospital Ligands de tim-3 et methodes associees
WO2005040791A2 (fr) * 2003-10-21 2005-05-06 Bayer Healthcare Ag Diagnostics et therapeutiques pour maladies liees au recepteur de peptides similaire a la somatostatine et a l'angiogenine (salpr)
US20050255560A1 (en) * 2003-11-21 2005-11-17 Zeren Gao Ztnf11, a tumor necrosis factor
CA2549164A1 (fr) 2003-12-05 2005-06-23 Northwestern University Amphiphiles peptidiques a assemblage automatique et procedes associes d'administration de facteur de croissance
JP4718490B2 (ja) 2003-12-23 2011-07-06 ジェノミック ヘルス, インコーポレイテッド フラグメント化rnaの全般的増幅
US20050186577A1 (en) 2004-02-20 2005-08-25 Yixin Wang Breast cancer prognostics
PT3556866T (pt) 2004-03-02 2021-04-30 Univ Johns Hopkins Mutações do gene de pik3ca em cancros humanos
TW200539890A (en) 2004-03-12 2005-12-16 Brigham & Womens Hospital Methods of modulating immune responses by modulating tim-1, tim-2 and tim-4 function
WO2005090569A1 (fr) * 2004-03-24 2005-09-29 The Council Of The Queensland Institute Of Medical Research Cancer et acides nucleiques et proteines de testicule vsm1 et vsm2, et leurs utilisations
WO2005095450A2 (fr) 2004-03-30 2005-10-13 Nsgene A/S Utilisation therapeutique d'un facteur de croissance, nsg33
DK2163650T3 (en) 2004-04-09 2015-11-02 Genomic Health Inc Genekspressionsmarkører for prediction of response to chemotherapy
DE102004024617A1 (de) 2004-05-18 2005-12-29 Ganymed Pharmaceuticals Ag Differentiell in Tumoren exprimierte Genprodukte und deren Verwendung
WO2005118832A2 (fr) * 2004-06-01 2005-12-15 Bayer Healthcare Ag Agents diagnostiques et therapeutiques pour des maladies associees a une proteine de type kinase regulee par serum/glucocorticoide (sgkl)
AU2005258335B2 (en) 2004-06-24 2011-03-17 Biogen Ma Inc. Treatment of conditions involving demyelination
US7587279B2 (en) 2004-07-06 2009-09-08 Genomic Health Method for quantitative PCR data analysis system (QDAS)
US7985831B2 (en) * 2004-08-05 2011-07-26 Toagosei Co., Ltd. Crohn's disease antibody epitope peptide and reagent for testing Crohn's disease
HUE051605T2 (hu) 2004-11-05 2021-03-01 Nsabp Found Inc Kemoterápiára adott válasz elõrejelzése génexpressziós markereket alkalmazva
CA2585561C (fr) 2004-11-05 2018-07-17 Genomic Health, Inc. Resultat de groupe esr1, pgr, bcl2 et scube2 comme indicateurs de pronostic de cancer du sein et de prediction de la reponse au traitement
EP2845865A1 (fr) 2004-11-12 2015-03-11 Xencor Inc. Variantes Fc avec liaison altérée en FcRn
US8367805B2 (en) 2004-11-12 2013-02-05 Xencor, Inc. Fc variants with altered binding to FcRn
CA2598251A1 (fr) 2005-03-04 2006-09-14 Northwestern University Epitopes angiogeniques de liaison a l'heparine, amphiphiles peptidiques, compositions auto-assemblees et procedes d'utilisation associes
WO2006120019A2 (fr) * 2005-05-13 2006-11-16 Centre National De La Recherche Scientifique Utilisation de nouveau codage genique pour nouvel element de la famille mcm2-8 dans les compositions pharmaceutiques
MX2008000253A (es) 2005-07-08 2008-04-02 Biogen Idec Inc Anticuerpos de sp35 y usos de los mismos.
WO2007037601A1 (fr) * 2005-09-28 2007-04-05 Knu-Industry Cooperation Foundation Polypeptide inhibant la transmigration de leucocytes ou la croissance et/ou la métastase de cellules cancéreuses, et une protéine de fusion de celui-ci
EP1790664A1 (fr) 2005-11-24 2007-05-30 Ganymed Pharmaceuticals AG Anticorps monoclonaux contre claudin-18 pour le traitement du cancer
DE602005023550D1 (de) 2005-12-14 2010-10-21 Licentia Ltd Verwendungen eines neurotrophischen Faktors
EP2044216A2 (fr) * 2006-07-03 2009-04-08 Exonhit Therapeutics SA Produits de la transcription spécifiques de la prostate et leur utilisation pour des thérapeutiques et des diagnostics du cancer de la prostate
US8076295B2 (en) 2007-04-17 2011-12-13 Nanotope, Inc. Peptide amphiphiles having improved solubility and methods of using same
US8999634B2 (en) * 2007-04-27 2015-04-07 Quest Diagnostics Investments Incorporated Nucleic acid detection combining amplification with fragmentation
ES2553270T3 (es) * 2007-07-27 2015-12-07 Immatics Biotechnologies Gmbh Nuevo epítopo inmunogénico para inmunoterapia
US7863021B2 (en) 2007-09-05 2011-01-04 Celera Corporation Genetic polymorphisms associated with rheumatoid arthritis, methods of detection and uses thereof
WO2009034661A1 (fr) * 2007-09-12 2009-03-19 Toppan Printing Co., Ltd. Procédé de diagnostic et induction d'une résistance à un virus
ES2401233T3 (es) 2007-10-17 2013-04-18 Universidad de Córdoba Isoformas del receptor tipo 5 de somatostatina humana producidas por tratamiento alternativo y pares de oligonucleótidos para la detección de las mismas por PCR
HUE024903T2 (en) 2007-12-26 2016-02-29 Xencor Inc FC variants with modified binding to FCRN
GB0803352D0 (en) * 2008-02-22 2008-04-02 Ntnu Technology Transfer As Oligopeptidic compounds and uses thereof
ES2584438T3 (es) 2008-03-21 2016-09-27 Universiteit Hasselt Biomarcadores para artritis reumatoide
EP2315779A2 (fr) 2008-07-09 2011-05-04 Biogen Idec MA Inc. Compositions comprenant des anticorps anti-lingo ou leurs fragments
ATE531383T1 (de) 2008-07-24 2011-11-15 Nsgene As Therapeutische verwendung eines wachstumsfaktors, metrnl
ES2536465T3 (es) 2008-10-01 2015-05-25 Immatics Biotechnologies Gmbh Composición de péptidos tumor-asociados y relacionados con la vacuna contra el cáncer para el tratamiento de glioblastoma (GBM) y otros cánceres
CA2758687A1 (fr) 2008-11-14 2010-05-20 Gen-Probe Incorporated Compositions, kits et procedes pour la detection d'acide nucleique de campylobacter
WO2010062960A2 (fr) 2008-11-26 2010-06-03 Cedars-Sinai Medical Center Méthodes de détermination d'une réceptivité à une thérapie par anti-tnfα lors d’une maladie intestinale inflammatoire
WO2010066018A1 (fr) * 2008-12-09 2010-06-17 Alethia Biotherapeutics Inc. Nouveau variant rétroviral endogène humain d’erv3 et utilisations de celui-ci pour diagnostiquer le cancer des ovaires
WO2010120830A1 (fr) 2009-04-13 2010-10-21 Northwestern University Nouvelles structures peptidiques pour la régénération des cartilages et procédés pour les utiliser
WO2011028952A1 (fr) 2009-09-02 2011-03-10 Xencor, Inc. Compositions et procédés pour une co-liaison bivalente et monovalente simultanée d'antigènes
ES2802623T3 (es) 2010-01-27 2021-01-20 Massachusetts Inst Technology Agentes polipeptídicos técnicamente diseñados para la neutralización dirigida de amplio espectro de la gripe
AU2011312417B2 (en) 2010-09-29 2015-08-20 Agensys, Inc. Antibody drug conjugates (ADC) that bind to 191P4D12 proteins
WO2012041328A1 (fr) 2010-10-01 2012-04-05 Nsgene A/S Traitement de l'allodynie, de l'hyperalgésie, de la douleur spontanée et de la douleur illusionnelle
DK2651430T3 (en) * 2010-12-14 2018-08-27 Hananja Ehf Biological activity of placental protein 13
US9474786B2 (en) 2011-09-05 2016-10-25 Nsgene A/S Treatment of allodynia
WO2013068445A1 (fr) * 2011-11-09 2013-05-16 Sanofi Diacylglycérol lipase et applications associées
WO2013167153A1 (fr) 2012-05-09 2013-11-14 Ganymed Pharmaceuticals Ag Anticorps utiles dans le diagnostic du cancer
CN104470541A (zh) 2012-05-14 2015-03-25 比奥根艾迪克Ma公司 用于治疗涉及运动神经元的疾患的lingo-2拮抗剂
WO2013174404A1 (fr) 2012-05-23 2013-11-28 Ganymed Pharmaceuticals Ag Polythérapie impliquant des anticorps dirigés contre la claudine 18,2 pour le traitement du cancer
CN105073776B (zh) 2012-11-13 2019-04-12 拜恩科技股份公司 用于治疗表达紧密连接蛋白的癌症疾病的制剂
WO2014127785A1 (fr) 2013-02-20 2014-08-28 Ganymed Pharmaceuticals Ag Polythérapie impliquant des anticorps dirigés contre la claudine 18,2 pour le traitement du cancer
US9023353B2 (en) * 2013-03-13 2015-05-05 The Board Of Trustees Of The University Of Arkansas Anti-(+)—methamphetamine monoclonal antibodies
WO2014146672A1 (fr) 2013-03-18 2014-09-25 Ganymed Pharmaceuticals Ag Thérapie comprenant des anticorps dirigés contre cldn 18.2 pour le traitement du cancer
CA2908553A1 (fr) 2013-03-27 2014-10-02 Cedars-Sinai Medical Center Attenuation ou regression de la fibrose et de l'inflammation par inhibition de la fonction tl1a et des voies de signalisation connexes
JP7019233B2 (ja) 2013-07-11 2022-02-15 モデルナティエックス インコーポレイテッド CRISPR関連タンパク質をコードする合成ポリヌクレオチドおよび合成sgRNAを含む組成物ならびに使用方法
WO2015010108A1 (fr) 2013-07-19 2015-01-22 Cedars-Sinai Medical Center Signature de la voie de signalisation de tl1a (tnfsf15)
JP6915987B2 (ja) 2013-09-25 2021-08-11 シトムクス セラピューティクス,インコーポレイティド マトリックスメタロプロテイナーゼ基質及び他の切断可能部分並びにそれらの使用方法
CA2925658C (fr) 2013-10-01 2022-12-06 Mie University Antigene contenant une sequence d'interepitope favorisant la presentation d'antigene aux cellules t
IL302642A (en) 2014-01-31 2023-07-01 Cytomx Therapeutics Inc Polypeptide substrates that activate matriptase and U-plasminogen and other cleavable groups, preparations containing them and their uses
JP2018504400A (ja) 2015-01-08 2018-02-15 バイオジェン・エムエイ・インコーポレイテッドBiogen MA Inc. Lingo‐1拮抗薬及び脱髄障害の治療のための使用
MA41374A (fr) 2015-01-20 2017-11-28 Cytomx Therapeutics Inc Substrats clivables par métalloprotéase matricielle et clivables par sérine protéase et procédés d'utilisation de ceux-ci
EP3307070A4 (fr) * 2015-06-12 2019-01-02 Emory University Compositions de croissance et de survie pour des cellules capables de produire des anticorps et procédés associés
CN108138198B (zh) * 2015-09-04 2022-10-28 合成基因组股份有限公司 为提高的产率而改造的微生物
KR20180127416A (ko) 2016-03-17 2018-11-28 세다르스-신나이 메디칼 센터 Rnaset2를 통한 염증성 장 질환의 진단 방법
US20200330606A1 (en) * 2016-03-29 2020-10-22 Valkyrie Therapeutics Inc. Modulation of structural maintenance of chromosome-1 expression
US9611297B1 (en) 2016-08-26 2017-04-04 Thrasos Therapeutics Inc. Compositions and methods for the treatment of cast nephropathy and related conditions
US11125757B2 (en) 2017-05-26 2021-09-21 Emory University Methods of culturing and characterizing antibody secreting cells
EP3508499A1 (fr) 2018-01-08 2019-07-10 iOmx Therapeutics AG Anticorps ciblant et d'autres modulateurs d'un gène d'immunoglobuline associé à une résistance contre des réponses immunitaires antitumorales et leurs utilisations
AU2020261435A1 (en) * 2019-04-26 2021-11-25 Sangamo Therapeutics, Inc. Engineering AAV
JP2022538688A (ja) 2019-07-05 2022-09-05 イオックス セラピューティクス アーゲー IGSF11(VSIG3)のIgC2に結合する抗体及びその使用
WO2021083958A1 (fr) * 2019-10-29 2021-05-06 Specialites Pet Food Nouveaux peptides induisant la satiété
WO2022008027A1 (fr) 2020-07-06 2022-01-13 Iomx Therapeutics Ag Anticorps de liaison à l'igv d'igsf11 (vsig3) et leurs utilisations
CN117529492A (zh) * 2021-01-21 2024-02-06 鹿特丹伊拉斯谟大学医疗中心 用于治疗的t细胞
US20220260597A1 (en) * 2021-02-18 2022-08-18 Beren Therapeutics P.B.C. Methods for the treatment of familial heterozygous and homozygous hypercholesterolemia with cyclodextrins
WO2023225602A1 (fr) * 2022-05-20 2023-11-23 Medikine, Inc. Polypeptides de liaison au récepteur de l'interleukine-18 et leurs utilisations

Family Cites Families (25)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4446240A (en) * 1981-01-30 1984-05-01 Nerenberg Samuel T Pancreas specific protein systems
US5457038A (en) * 1988-11-10 1995-10-10 Genetics Institute, Inc. Natural killer stimulatory factor
US6080540A (en) * 1990-04-20 2000-06-27 Cold Spring Harbor Laboratory Cloning of mammalian genes in microbial organisms and methods for pharmacological screening
JP2845623B2 (ja) * 1990-11-26 1999-01-13 アメリカ合衆国 細胞のストレス転写因子
US5721352A (en) * 1991-02-19 1998-02-24 University Of Florida Research Foundation Entomopoxvirus expression system
US5840870A (en) * 1995-12-29 1998-11-24 Incyte Pharmaceuticals, Inc. Polynucleotides PANC1A and PANC1B associated with pancreatic cancer
WO1999014234A2 (fr) * 1997-09-17 1999-03-25 Genentech, Inc. Stimulation ou inhibition de l'angiogenese et de la cardiovascularisation
US5912160A (en) * 1995-08-22 1999-06-15 Thomas Jefferson University Gab1, Grb2 binding protein, and compositions for making and methods of using the same
JP3462313B2 (ja) * 1995-08-24 2003-11-05 キッコーマン株式会社 変異型ウリカーゼ、変異型ウリカーゼ遺伝子、新規な組み換え体dna及び変異型ウリカーゼの製造法
US5998165A (en) * 1995-12-29 1999-12-07 Incyte Pharmaceuticals, Inc. Polynucleotides encoding a protein associated with pancreatic cancer
JP2000511410A (ja) * 1996-04-29 2000-09-05 ザ ジョーンズ ホプキンス ユニバーシティー スクール オブ メディシン ナンセンス介在rna崩壊の哺乳動物の調節因子
US6500934B1 (en) * 1996-07-24 2002-12-31 Michael Rush Lerner Bivalent agonists for G-protein coupled receptors
US5976834A (en) * 1997-01-09 1999-11-02 Smithkline Beecham Corporation cDNA clone HNFJD15 that encodes a novel human 7-transmembrane receptor
US5925521A (en) * 1997-03-31 1999-07-20 Incyte Pharmaceuticals, Inc. Human serine carboxypeptidase
CA2232743A1 (fr) * 1997-04-02 1998-10-02 Smithkline Beecham Corporation Tl5, homologue tnf
WO1998045435A2 (fr) * 1997-04-10 1998-10-15 Genetics Institute, Inc. MARQUEURS SECRETES DE SEQUENCE EXPRIMEE (sEST)
IT1291110B1 (it) * 1997-04-15 1998-12-29 Istituto Europ Di Oncologia S Interattori intracellulari e specificita' di legame del dominio eh
US5948641A (en) * 1997-05-29 1999-09-07 Incyte Pharmaceuticals, Inc. Polynucleotides encoding a metal response element binding protein
WO1998055620A1 (fr) * 1997-06-06 1998-12-10 Regeneron Pharmaceuticals, Inc. Element ntn-2 de la famille des ligands du tnf
US5932442A (en) * 1997-09-23 1999-08-03 Incyte Pharmaceuticals, Inc. Human regulatory molecules
US5972660A (en) * 1997-10-22 1999-10-26 Incyte Pharmaceuticals, Inc. Human hydroxypyruvate reductase
DE19818598A1 (de) * 1998-04-19 1999-10-21 Metagen Gesellschaft Fuer Genomforschung Mbh Menschliche Nukleinsäuresequenzen aus Pankreasnormalgewebe
CA2327317A1 (fr) * 1998-04-29 1999-11-04 Genesis Research And Development Corporation Limited Polynucleotides isoles de cellules de la peau et leurs procedes d'utilisation
US6262249B1 (en) * 1998-06-23 2001-07-17 Chiron Corporation Pancreatic cancer genes
CA2296792A1 (fr) * 1999-02-26 2000-08-26 Genset S.A. Sequences marqueurs exprimees et proteines humaines codees

Non-Patent Citations (5)

* Cited by examiner, † Cited by third party
Title
DATABASE GENBANK [online] 1 November 1998 (1998-11-01), TAKATSU ET AL.: "Gamma-adaptin", XP002941826, Database accession no. O75842 *
DATABASE GENBANK [online] 22 September 1998 (1998-09-22), NAKAYAMA K.: "Homo sapiens mRNA for gamma1-adaptin, complete cds", XP002941824, Database accession no. AB015317 *
J. BIOL. CHEM., vol. 273, 1998, pages 24693 - 24700 *
J. BIOL. CHEM., vol. 273, no. 38, 1998, pages 24693 - 24700 *
TAKATSU ET AL.: "Identification and characterization of novel clathrin adapter-related proteins", J. OF BIOLOGICAL CHEMISTRY, vol. 273, no. 38, 18 September 1998 (1998-09-18), pages 24693 - 24700, XP002941825 *

Cited By (25)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6774223B2 (en) 2000-06-28 2004-08-10 Diadexus, Inc. Method of diagnosing, monitoring, staging, imaging and treating colon cancer
WO2003024276A1 (fr) * 2000-08-02 2003-03-27 Millennium Pharmaceuticals,Inc. Membre de la famille du facteur de liberation du gtp humain (15368) et ses utilisations
US6962799B2 (en) * 2000-08-21 2005-11-08 Incyte Corporation Microtubule-associated proteins and tubulins
WO2002016587A3 (fr) * 2000-08-21 2003-01-16 Incyte Genomics Inc Proteines associees aux microtubules et tubulines
WO2002016587A2 (fr) * 2000-08-21 2002-02-28 Incyte Genomics, Inc. Proteines associees aux microtubules et tubulines
WO2002020765A3 (fr) * 2000-09-08 2003-03-06 Millennium Pharm Inc 38646, nouveau facteur d'echange de guanine nucleotide et ses utilisations
WO2002031132A3 (fr) * 2000-10-11 2003-03-13 Millennium Pharm Inc Nouvel element de la famille des phosphatases humaines a double specificite, appele 8843, et utilisation de ce dernier
WO2002090543A2 (fr) * 2001-05-09 2002-11-14 Bayer Aktiengesellschaft Régulation de la protéine semblable à celle de type 2c de la phosphatase de l'acide phosphatidique humain
WO2002090543A3 (fr) * 2001-05-09 2003-12-18 Bayer Ag Régulation de la protéine semblable à celle de type 2c de la phosphatase de l'acide phosphatidique humain
WO2003057870A1 (fr) * 2002-01-07 2003-07-17 Bayer Healthcare Ag Proteine humaine analogue a la phosphatase de l'acide phosphatidique de type 2
JP2006504398A (ja) * 2002-03-21 2006-02-09 ザ・バブラハム・インスティテュート P−Rex1、RACのためのPTDINS(3,4,5)P3−G−β−γにより制御されたグアニンヌクレオチド交換因子
WO2003080664A1 (fr) * 2002-03-21 2003-10-02 The Babraham Institute Proteine p-rex1, facteur d'echange guanine-nucleotide pour rac regule par ptdins (3, 4, 5) p3 et par g-beta-gamma-
EP1540014A2 (fr) * 2002-08-27 2005-06-15 Epigenomics AG Procede et acides nucleiques servant a l'analyse de troubles lies a la proliferation des cellules mammaires
EP1907589A2 (fr) * 2005-07-26 2008-04-09 Siemens Medical Solutions Diagnostics Polymorphismes de nucleotides uniques a susceptibilite par rapport a une maladie cardio-vasculaire
EP1907589A4 (fr) * 2005-07-26 2010-11-24 Siemens Healthcare Diagnostics Polymorphismes de nucleotides uniques a susceptibilite par rapport a une maladie cardio-vasculaire
WO2008030559A2 (fr) * 2006-09-08 2008-03-13 Corixa Corporation Procédés, compositions, et kits de détection et de surveillance d'un cancer du colon
WO2008030559A3 (fr) * 2006-09-08 2008-12-11 Corixa Corp Procédés, compositions, et kits de détection et de surveillance d'un cancer du colon
JP2010534200A (ja) * 2007-07-20 2010-11-04 キャンサー・リサーチ・テクノロジー・リミテッド C型レクチンを介する免疫
WO2009013484A1 (fr) * 2007-07-20 2009-01-29 Cancer Research Technology Limited Modulation immunitaire par lectine de type c
US8580266B2 (en) 2007-07-20 2013-11-12 Cancer Research Technology Limited Immune modulation via C-type lectin
US9205153B2 (en) 2007-07-20 2015-12-08 Cancer Research Technology Limited Immune modulation via C-type lectin
US10144934B2 (en) 2012-04-30 2018-12-04 Biocon Limited Targeted/TGF-ßRII fusion proteins and methods for making same
US11028399B2 (en) 2012-04-30 2021-06-08 Biocon Limited CTLA4/PD1 targeted/immunomodulatory fusion proteins
US11060097B2 (en) 2012-04-30 2021-07-13 Biocon Limited Immunotherapy methods using anti-PD-L1 antibodies in combination with EGFR1 targeted-TGF-beta immunomodulatory fusion proteins
WO2021228999A1 (fr) * 2020-05-12 2021-11-18 Institut Curie Épitopes néo-antigéniques associés à des mutations sf3b1

Also Published As

Publication number Publication date
CA2392428A1 (fr) 2001-08-02
WO2001055308A2 (fr) 2001-08-02
WO2001055303A3 (fr) 2001-12-20
AU2001241414A1 (en) 2001-08-07
AU2001241410A1 (en) 2001-08-07
WO2001055207A8 (fr) 2001-09-07
WO2001055321A8 (fr) 2001-09-07
WO2001054473A2 (fr) 2001-08-02
WO2001055303A8 (fr) 2001-09-07
WO2001055323A2 (fr) 2001-08-02
CA2395849A1 (fr) 2001-08-02
WO2001055447A1 (fr) 2001-08-02
WO2001055322A8 (fr) 2001-09-07
CA2392450A1 (fr) 2001-08-16
WO2001055314A2 (fr) 2001-08-02
CA2393652A1 (fr) 2001-08-02
WO2001055163A8 (fr) 2001-09-07
WO2001055308A3 (fr) 2002-07-04
AU2001250770A1 (en) 2001-08-07
WO2001055343A8 (fr) 2001-09-07
WO2001055368A1 (fr) 2001-08-02
WO2001055315A8 (fr) 2001-09-07
CA2395693A1 (fr) 2001-08-02
CA2395816A1 (fr) 2001-08-02
WO2001055304A3 (fr) 2002-07-18
WO2001055310A8 (fr) 2001-09-07
AU2001241416A1 (en) 2001-08-07
WO2001054733A8 (fr) 2001-09-07
WO2001055387A8 (fr) 2001-12-06
WO2001055205A1 (fr) 2001-08-02
WO2001055309A3 (fr) 2002-07-18
CA2395403A1 (fr) 2001-08-02
CA2395738A1 (fr) 2002-08-02
AU4313701A (en) 2001-08-14
AU2001241415A1 (en) 2001-08-07
WO2001055202A8 (fr) 2001-09-07
WO2001055208A8 (fr) 2001-09-07
WO2001055314A3 (fr) 2002-05-02
WO2001055318A3 (fr) 2002-07-04
WO2001055367A8 (fr) 2001-12-20
WO2001055307A8 (fr) 2001-09-07
WO2001055204A8 (fr) 2001-09-07
WO2001055173A8 (fr) 2001-11-29
WO2001055207A1 (fr) 2001-08-02
CA2397407A1 (fr) 2001-08-02
WO2001055301A8 (fr) 2001-09-07
WO2001055319A2 (fr) 2001-08-02
WO2001055364A2 (fr) 2001-08-02
WO2001055316A3 (fr) 2002-04-11
WO2001055311A8 (fr) 2001-09-07
WO2001055325A2 (fr) 2001-08-02
CA2395178A1 (fr) 2001-08-02
WO2001057182A3 (fr) 2002-03-28
WO2001059064A3 (fr) 2002-03-14
CA2395666A1 (fr) 2001-08-02
WO2001055200A8 (fr) 2001-09-07
WO2001055324A8 (fr) 2001-09-07
WO2001054474A8 (fr) 2001-12-20
WO2001055320A3 (fr) 2002-04-11
WO2001055203A8 (fr) 2001-09-07
CA2392422A1 (fr) 2001-08-02
WO2001055307A3 (fr) 2002-01-10
CA2395724A1 (fr) 2001-08-02
AU2001241417A1 (en) 2001-08-07
CA2398411A1 (fr) 2001-08-02
CA2395295A1 (fr) 2001-08-02
WO2001055206A8 (fr) 2001-12-13
WO2001055316A8 (fr) 2001-09-07
CA2395885A1 (fr) 2001-08-02
WO2001055324A2 (fr) 2001-08-02
CA2392751A1 (fr) 2001-08-02
WO2001055315A2 (fr) 2001-08-02
WO2001055350A8 (fr) 2001-09-07
WO2001055202A1 (fr) 2001-08-02
WO2001055327A2 (fr) 2001-08-02
WO2001055327A3 (fr) 2002-02-28
WO2001055312A3 (fr) 2002-03-14
AU2001241406A1 (en) 2001-08-07
WO2001055200A1 (fr) 2001-08-02
WO2001055448A1 (fr) 2001-08-02
AU2001241407A1 (en) 2001-08-07
WO2001055327A8 (fr) 2001-09-07
AU4141101A (en) 2001-08-20
CA2393002A1 (fr) 2001-08-02
WO2001055350A1 (fr) 2001-08-02
WO2001055319A3 (fr) 2002-02-21
CA2394841A1 (fr) 2001-08-02
WO2001055364A3 (fr) 2002-07-04
WO2001055325A3 (fr) 2002-07-04
WO2001055316A2 (fr) 2001-08-02
WO2001055322A3 (fr) 2002-07-04
WO2001055323A8 (fr) 2001-09-07
WO2001057182A2 (fr) 2001-08-09
WO2001055310A2 (fr) 2001-08-02
CA2395787A1 (fr) 2001-08-02
WO2001055311A3 (fr) 2002-07-04
CA2395699A1 (fr) 2001-08-02
WO2001054474A2 (fr) 2001-08-02
WO2001055203A1 (fr) 2001-08-02
WO2001055304A2 (fr) 2001-08-02
CA2393618A1 (fr) 2001-08-02
WO2001055387A1 (fr) 2001-08-02
WO2001055307A2 (fr) 2001-08-02
AU2001241412A1 (en) 2001-08-07
WO2001054733A1 (fr) 2001-08-02
CA2393912A1 (fr) 2001-08-02
WO2001055447A8 (fr) 2001-09-07
AU2001241405A1 (en) 2001-08-07
WO2001055364A8 (fr) 2001-09-07
WO2001055205A8 (fr) 2001-12-13
WO2001055309A8 (fr) 2001-09-07
CA2398877A1 (fr) 2001-08-09
CA2392438A1 (fr) 2001-08-02
WO2001055367A1 (fr) 2001-08-02
WO2001055204A1 (fr) 2001-08-02
WO2001055328A3 (fr) 2002-07-04
WO2001055301A2 (fr) 2001-08-02
WO2001055312A8 (fr) 2001-09-07
WO2001055308A8 (fr) 2001-09-07
WO2001055309A2 (fr) 2001-08-02
WO2001055355A8 (fr) 2001-09-07
WO2001055320A2 (fr) 2001-08-02
WO2001055321A3 (fr) 2002-07-04
AU2001241418A1 (en) 2001-08-07
WO2001055173A2 (fr) 2001-08-02
WO2001055321A2 (fr) 2001-08-02
CA2395398A1 (fr) 2001-08-02
WO2001055318A2 (fr) 2001-08-02
WO2001055343A1 (fr) 2001-08-02
WO2001055304A8 (fr) 2001-09-07
WO2001055302A2 (fr) 2001-08-02
WO2001055312A2 (fr) 2001-08-02
WO2001055320A8 (fr) 2001-09-07
WO2001055201A1 (fr) 2001-08-02
CA2395654A1 (fr) 2001-08-02
CA2395729A1 (fr) 2001-08-02
WO2001055301A3 (fr) 2009-06-04
WO2001055319A8 (fr) 2001-09-07
WO2001055302A8 (fr) 2001-09-07
WO2001055311A2 (fr) 2001-08-02
WO2001055315A3 (fr) 2002-02-07
WO2001055303A2 (fr) 2001-08-02
WO2001055201A8 (fr) 2001-09-07
WO2001055328A2 (fr) 2001-08-02
WO2001055355A1 (fr) 2001-08-02
WO2001055368A8 (fr) 2001-09-07
WO2001055310A3 (fr) 2001-12-27
WO2001055206A1 (fr) 2001-08-02
CA2394039A1 (fr) 2001-08-02
CA2395827A1 (fr) 2001-08-02
WO2001055208A1 (fr) 2001-08-02
CA2392398A1 (fr) 2001-08-02
WO2001055318A8 (fr) 2001-09-07
WO2001055448A8 (fr) 2001-09-07
CA2395858A1 (fr) 2001-08-02
AU2001241409A1 (en) 2001-08-07
WO2001055328A8 (fr) 2001-09-07
WO2001055324A3 (fr) 2002-01-24
WO2001059064A2 (fr) 2001-08-16
CA2395671A1 (fr) 2001-08-02
CA2392757A1 (fr) 2001-08-02
WO2001055322A2 (fr) 2001-08-02
WO2001054473A8 (fr) 2001-09-07
WO2001055302A3 (fr) 2002-02-14
WO2001055323A3 (fr) 2002-05-10
CA2395872A1 (fr) 2001-08-02
CA2395734A1 (fr) 2001-08-02
WO2001055314A8 (fr) 2001-09-07
AU2001241408A1 (en) 2001-08-07
CA2397839A1 (fr) 2001-08-02
WO2001055325A8 (fr) 2001-12-13
AU2001241413A1 (en) 2001-08-07

Similar Documents

Publication Publication Date Title
WO2001055163A1 (fr) Acides nucleiques, proteines, et anticorps
WO2001090304A2 (fr) Acides nucleiques, proteines et anticorps
WO2001055317A2 (fr) Acides nucleiques, proteines et anticorps
WO2001055326A2 (fr) Acides nucleiques, proteines et anticorps
WO2001055441A2 (fr) Acides nucléiques, protéines et anticorps
EP1254151A1 (fr) Acides nucleiques, proteines, et anticorps
EP1254157A1 (fr) Acides nucleiques, proteines et anticorps
WO2002057420A2 (fr) 50 proteines secretees humaines
WO2001055162A1 (fr) Acides nucleiques, proteines et anticorps
WO2003072761A1 (fr) 20 proteines humaines secretees
EP1268527A2 (fr) Acides nucleiques, proteines et anticorps
EP1254217A2 (fr) Acides nucl iques, prot ines et anticorps
EP1254228A1 (fr) Acides nucleiques, proteines et anticorps

Legal Events

Date Code Title Description
AK Designated states

Kind code of ref document: A1

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

AL Designated countries for regional patents

Kind code of ref document: A1

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

AK Designated states

Kind code of ref document: C1

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

AL Designated countries for regional patents

Kind code of ref document: C1

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

CFP Corrected version of a pamphlet front page
CR1 Correction of entry in section i

Free format text: PAT. BUL. 31/2001 UNDER "PUBLISHED", ADD "WITH SEQUENCE LISTING PART OF DESCRIPTION PUBLISHED SEPARATELY IN ELECTRONIC FORM AND AVAILABLE UPON REQUEST FROM THE INTERNATIONAL BUREAU."

121 Ep: the epo has been informed by wipo that ep was designated in this application
REG Reference to national code

Ref country code: DE

Ref legal event code: 8642

WWE Wipo information: entry into national phase

Ref document number: 2395398

Country of ref document: CA

DFPE Request for preliminary examination filed prior to expiration of 19th month from priority date (pct application filed before 20040101)
WWE Wipo information: entry into national phase

Ref document number: 2001918156

Country of ref document: EP

WWP Wipo information: published in national office

Ref document number: 2001918156

Country of ref document: EP

WWW Wipo information: withdrawn in national office

Ref document number: 2001918156

Country of ref document: EP

NENP Non-entry into the national phase

Ref country code: JP