US20070141590A1 - Modulators of tissue regeneration - Google Patents

Modulators of tissue regeneration Download PDF

Info

Publication number
US20070141590A1
US20070141590A1 US11/463,239 US46323906A US2007141590A1 US 20070141590 A1 US20070141590 A1 US 20070141590A1 US 46323906 A US46323906 A US 46323906A US 2007141590 A1 US2007141590 A1 US 2007141590A1
Authority
US
United States
Prior art keywords
kim
seq
subject
polypeptide
protein
Prior art date
Legal status (The legal status 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 status listed.)
Abandoned
Application number
US11/463,239
Inventor
Michele Sanicola-Nadel
Joseph Bonventre
Catherine Hession
Takaharu Ichimura
Henry Wei
Richard Cate
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
General Hospital Corp
Biogen MA Inc
Original Assignee
Individual
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
Family has litigation
First worldwide family litigation filed litigation Critical https://patents.darts-ip.com/?family=26690881&utm_source=google_patent&utm_medium=platform_link&utm_campaign=public_patent_search&patent=US20070141590(A1) "Global patent litigation dataset” by Darts-ip is licensed under a Creative Commons Attribution 4.0 International License.
Application filed by Individual filed Critical Individual
Priority to US11/463,239 priority Critical patent/US20070141590A1/en
Assigned to THE GENERAL HOSPITAL CORPORATION reassignment THE GENERAL HOSPITAL CORPORATION ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: ICHIMURA, TAKAHARU, BONVENTRE, JOSEPH V.
Assigned to BIOGEN, INC. reassignment BIOGEN, INC. ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: CATE, RICHARD L., HESSION, CATHERINE, SANICOLA-NADEL, MICHELE, WEI, HENRY
Assigned to BIOGEN IDEC MA INC. reassignment BIOGEN IDEC MA INC. CHANGE OF NAME (SEE DOCUMENT FOR DETAILS). Assignors: BIOGEN, INC.
Publication of US20070141590A1 publication Critical patent/US20070141590A1/en
Assigned to NATIONAL INSTITUTES OF HEALTH - DIRECTOR DEITR reassignment NATIONAL INSTITUTES OF HEALTH - DIRECTOR DEITR CONFIRMATORY LICENSE (SEE DOCUMENT FOR DETAILS). Assignors: MASSACHUSETTS GENERAL HOSPITAL
Abandoned legal-status Critical Current

Links

Images

Classifications

    • 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
    • C07K16/28Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants
    • C07K16/2803Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants against the immunoglobulin superfamily
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/50Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates
    • A61K47/51Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent
    • A61K47/68Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an antibody, an immunoglobulin or a fragment thereof, e.g. an Fc-fragment
    • A61K47/6835Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an antibody, an immunoglobulin or a fragment thereof, e.g. an Fc-fragment the modifying agent being an antibody or an immunoglobulin bearing at least one antigen-binding site
    • A61K47/6849Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an antibody, an immunoglobulin or a fragment thereof, e.g. an Fc-fragment the modifying agent being an antibody or an immunoglobulin bearing at least one antigen-binding site the antibody targeting a receptor, a cell surface antigen or a cell surface determinant
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K51/00Preparations containing radioactive substances for use in therapy or testing in vivo
    • A61K51/02Preparations containing radioactive substances for use in therapy or testing in vivo characterised by the carrier, i.e. characterised by the agent or material covalently linked or complexing the radioactive nucleus
    • A61K51/04Organic compounds
    • A61K51/08Peptides, e.g. proteins, carriers being peptides, polyamino acids, proteins
    • A61K51/10Antibodies or immunoglobulins; Fragments thereof, the carrier being an antibody, an immunoglobulin or a fragment thereof, e.g. a camelised human single domain antibody or the Fc fragment of an antibody
    • A61K51/1027Antibodies or immunoglobulins; Fragments thereof, the carrier being an antibody, an immunoglobulin or a fragment thereof, e.g. a camelised human single domain antibody or the Fc fragment of an antibody against receptors, cell-surface antigens or cell-surface determinants
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P13/00Drugs for disorders of the urinary system
    • A61P13/12Drugs for disorders of the urinary system of the kidneys
    • 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
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/5005Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving human or animal cells
    • G01N33/5008Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving human or animal cells for testing or evaluating the effect of chemical or biological compounds, e.g. drugs, cosmetics
    • G01N33/5082Supracellular entities, e.g. tissue, organisms
    • 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
    • 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

Definitions

  • the invention relates to proteins which are upregulated in injured or regenerating tissues, as well as to the DNA encoding these proteins.
  • the invention further relates to therapeutic compositions and methods of treatment encompassing these proteins.
  • a dynamic remodeling of tissue architecture occurs during development and during tissue repair after injury.
  • results of studies of this process using a model of kidney injury caused by an ischemia-reperfusion insult.
  • the kidney is able to repair damage to the proximal tubule epithelium through a complex series of events involving cell death, proliferation of surviving proximal tubule epithelial cells, formation of poorly differentiated regenerative epithelium over the denuded basement membrane, and differentiation of the regenerative epithelium to form fully functional proximal tubule epithelial cells (Wallin et al., Lab. Invest. 66:474-484, 1992; Witzgall et al., Mol. Cell. Biol. 13:1933-1942, 1994; Ichimura et al., Am. J. Physiol. 269:F653-662, 1995; Thadhani et al., N. Engl.
  • RDA representational difference analysis
  • the invention generally provides Kidney Injury-related Molecules (each of which is henceforth called a “KIM”) which are upregulated in renal tissue after injury to the kidney.
  • KIM proteins and peptides of the invention as well as their agonists and antagonists, and their corresponding nucleic acids are useful in a variety of therapeutic interventions.
  • the invention provides a purified and isolated DNA molecule having a nucleotide sequence set forth in SEQ ID NO:1, SEQ ID NO:2, SEQ ID NO:4 or SEQ ID NO:6.
  • the invention also includes the complementary strands of these sequences, DNA molecules which hybridize under stringent conditions to the aforementioned DNA molecules, and DNA molecules which, but for the degeneracy of the genetic code, would hybridize to any of the DNA molecules defined above.
  • These DNA molecules may be recombinant, and may be operably linked to an expression control sequence.
  • the invention further provides a vector comprising a purified and isolated DNA molecule having a nucleotide sequence set forth in SEQ ID NO:1, SEQ ID NO:2, SEQ ID NO:4 or SEQ ID NO:6, or one of the other DNA molecules defined above.
  • This vector may be a biologically functional plasmid or viral DNA vector.
  • One embodiment of the invention provides a prokaryotic or eukaryotic host cell stably transformed or transfected by a vector comprising a DNA molecule of SEQ ID NO:1, SEQ ID NO:2, SEQ ID NO:4 or SEQ ID NO:6.
  • a process for the production of a KIM polypeptide product encoded by a DNA molecule as described above; the process involves growing, under suitable culture conditions, prokaryotic or eukaryotic host cells transformed or transfected with the DNA molecule in a manner allowing expression of the DNA molecule, and recovering the polypeptide product of said expression.
  • KIM proteins of the invention may have an amino acid sequence which comprises SEQ ID NO:3, SEQ ID NO:5, or SEQ ID NO:7, or may be a variant of SEQ ID NO:3, SEQ ID NO:5 or SEQ ID NO:7, or a purified and isolated protein encoded by the DNA of SEQ ID NO:1, SEQ ID NO:2, SEQ ID NO:4 or SEQ ID NO:6. These proteins can be provided substantially free of other human proteins.
  • the invention further includes variants of these proteins, such as soluble variants or fusion proteins.
  • KIM fusion proteins of the invention may comprise an immunoglobulin, a toxin, an imageable compound or a radionuclide.
  • the invention also provides a specific antibody, such as a monoclonal antibody (MAb), to the KIM proteins described above.
  • the present antibody binds to any epitope that is unique to a KIM protein disclosed herein. In some embodiments, the epitope is displayed on the surface of a cell expressing KIM.
  • Antigen-binding fragments of the antibody are also provided herein, preferably Fab, Fab2, Fab′ and Fv fragments, whether produced by chemical or enzymatic cleavage, or by molecular engineering techniques.
  • Engineered versions of the present antibody include chimeric, humanized and human antibodies, and antibody fusion proteins.
  • Monoclonal antibodies (MAbs) of the present invention can be of mouse, rat, hamster or human origin.
  • An exemplary MAb of the invention is the murine AKG7 MAb disclosed herein, which binds specifically to the human KIM polypeptide disclosed herein.
  • Anti-KIM antibodies of the present invention may be conjugated or fused to a therapeutic agent, toxin, imageable compound or radionuclide.
  • therapeutic agents include cytokines, lymphokines, trophic factors, survival factors, chemokines and chemoattractants.
  • Exemplary toxins include ricin and diphtheria toxin.
  • Exemplary imageable compounds include luminescent proteins (e.g., luciferin), fluorescent proteins (e.g., green fluorescent protein), haptens (e.g., biotin), and radioactively labeled proteins.
  • Exemplary radionuclides include any radionuclide used for medical imaging purposes.
  • the invention further encompasses all hybridoma cell lines and engineered host cells which produce antibodies of the invention.
  • compositions are also within the scope of the invention.
  • a pharmaceutical composition of the invention may comprise a therapeutically effective amount of a KIM protein or anti-KIM antibody of the invention, along with a pharmacologically acceptable carrier.
  • Diagnostic methods are within the invention, such as assessing the presence or course of resolution of renal injury by measuring the concentration of KIM in urine, serum, or urine sediment of patients who have or who are at risk of developing renal disease.
  • Other diagnostic methods that are within the invention include assessing KIM expression level in kidney tissue (e.g., in kidney biopsy tissue) of patients who have, are suspected of having, or are at risk of developing renal cancer (e.g., renal carcinoma).
  • the present methods involve contacting an appropriate tissue or fluid sample derived from the patient being diagnosed, with a KIM antibody or a KIM probe (as the case may be), under binding conditions. Complexes formed by the binding of antibody or probe to KIM protein or nucleic acid (e.g., RNA) in the sample are detected by standard techniques.
  • the presence or abnormal elevation of KIM protein in urine or serum is expected to correlate with renal failure or renal disease.
  • the presence or abnormal elevation of KIM gene expression in renal cells or tissue is expected to correlate with disease processes, particularly carcinogenesis. Such correlations are expected to be useful in the prognostication, staging and clinical management of diseases or other conditions deleterious to renal tissue and/or renal function.
  • Methods of treatment of the invention include treating patients with therapeutically effective amounts of KIM, KIM variants, KIM analogs, KIM fusion proteins, KIM agonists, and antibodies to KIM or to KIM ligands.
  • Other therapeutic compounds of the invention include KIM ligands, anti-KIM antibodies, and fusions proteins of KIM ligands. These compounds can be useful in therapeutic methods which either stimulate or inhibit cellular responses that are dependent on KIM function.
  • Further methods of the invention inhibit the growth of KIM-expressing tumor cells by contacting the cells with a fusion protein of a KIM ligand and either a toxin or radionuclide, or with an anti-KIM antibody conjugated to a toxin or to a radionuclide.
  • growth of tumor cells which express KIM ligand may be inhibited by contacting the cells with a fusion protein of a KIM and either a toxin or radionuclide, or with an anti-KIM ligand antibody conjugated to a toxin or to a radionuclide.
  • the invention also encompasses methods of gene therapy. These include a method of treating a subject with a renal disorder, a method of promoting growth of new tissue in a subject, and a method of promoting survival of damaged tissue in a subject, wherein each method comprises administering to the subject a vector which includes DNA comprising the nucleotide sequence of SEQ ID NO:1, SEQ ID NO:2, SEQ ID NO:4 or SEQ ID NO:6.
  • the compounds of the invention are also useful for imaging tissues, either in vitro or in vivo.
  • One such method involves targeting an imageable compound to a cell expressing a protein of SEQ ID NO:3, SEQ ID NO:5 or SEQ ID NO:7, comprising contacting the cell with either a monoclonal antibody of the invention or a fusion protein comprising a protein as described above, fused to an imageable compound.
  • the cell is within a subject, and the protein or the monoclonal antibody is administered to the subject.
  • the invention also includes diagnostic methods, such as a method of identifying damage or regeneration of renal cells in a subject, comprising comparing the level of expression of either SEQ ID NO:1,SEQ ID NO:2, SEQ ID NO:4 or SEQ ID NO:6 in renal cells of the subject to a control level of expression of the sequence in control renal cells.
  • diagnostic methods such as a method of identifying damage or regeneration of renal cells in a subject, comprising comparing the level of expression of either SEQ ID NO:1,SEQ ID NO:2, SEQ ID NO:4 or SEQ ID NO:6 in renal cells of the subject to a control level of expression of the sequence in control renal cells.
  • Another method of the invention includes identifying upregulation of SEQ ID NO:1, SEQ ID NO:2, SEQ ID NO:4 or SEQ ID NO:6 in cells comprising contacting the cells with an antisense probe and measuring hybridization to RNA within the cell.
  • a further embodiment of the diagnostic methods of the invention includes assessing the presence or concentration of a molecule of the invention either in urine, serum, or other body fluids, or in urine sediment or tissue samples.
  • the measured injury-related molecule can be correlated with the presence, extent or course of a pathologic process. This correlation can also be used to assess the efficacy of a therapeutic regime.
  • FIG. 1 is a text depiction of the nucleotide sequence of rat clone cDNA 3-2, with putative protein reading frame of 615 to 1535.
  • FIG. 2 is a text depiction of the cDNA sequence of rat clone 1-7, with putative protein reading frame of 145 to 1065.
  • FIG. 3 is a text depiction of the cDNA sequence of rat clone 4-7, with putative protein reading frame of 107 to 1822.
  • FIG. 4 is a text depiction of the cDNA and deduced amino acid sequences of human clone HI3-10-85, with putative protein reading frame of 1 to 1002.
  • the upper line of the listing is the cDNA sequence (SEQ ID NO:6), and the lower line is the deduced amino acid sequence (SEQ ID NO:7).
  • FIG. 5 is a text depiction of a BESTFIT comparison of the nucleotide sequence of human clone HI3-10-85 with that of rat clone 3-2.
  • FIG. 6 is a digitized image of a Northern blot of human renal tissue samples isolated from renal carcinomas and from normal renal tissue samples.
  • RDA representational difference analysis
  • KIM protein is a protein encoded by mRNA which is selectively upregulated following injury to a kidney.
  • One group of KIM proteins of interest includes those coded for by mRNA which is selectively upregulated at any time within one week following any insult which results in injury to renal tissue. Examples of times at which such upregulation might be identified include 10 hours, 24 hours, 48 hours or 96 hours following an insult. Examples of types of insults include those resulting in ischemic, toxic or other types of injury.
  • a “KIM agonist” is a molecule which can specifically trigger a cellular response normally triggered by the interaction of KIM with a KIM ligand.
  • a KIM agonist can be a KIM variant, or a specific antibody to KIM, or a soluble form of the KIM ligand.
  • KIM antagonist is a molecule which can specifically associate with a KIM ligand or KIM, thereby blocking or otherwise inhibiting KIM binding to the KIM ligand.
  • the antagonist binding blocks or inhibits cellular responses which would otherwise be triggered by ligation of the KIM ligand with KIM or with a KIM agonist.
  • KIM antagonists include certain KIM variants, KIM fusion proteins and specific antibodies to a KIM ligand or KIM.
  • a “KIM ligand” is any molecule which noncovalently and specifically binds to a KIM protein.
  • a ligand can be a protein, peptide, steroid, antibody, amino acid derivative, or other type molecule, in any form, including naturally-occurring, recombinantly produced, or otherwise synthetic.
  • a KIM ligand can be in any form, including soluble, membrane-bound, or part of a fusion construct with immunoglobulin, fatty acid, or other moieties.
  • the KIM ligand may be an integrin.
  • a membrane-bound KIM ligand can act as a receptor which, when bound to or associated with KIM, triggers a cellular response.
  • KIM may associate with more than a single KIM ligand, or may associate with a KIM ligand as part of a complex with one or more other molecules or cofactors.
  • the KIM may associate and react with KIM ligand which is bound to the same cell as the KIM, or it may associate and react with KIM ligand be bound to a second cell.
  • the KIM ligation occurs between molecules bound to different cells, the two cells may be the same or different with respect to cellular type or origin, phenotypic or metabolic condition, or type or degree of cellular response (e.g., growth, differentiation or apoptosis) to a given stimulus.
  • KIM ligation refers to the contact and binding of KIM with a KIM ligand.
  • alignment of sequences is meant the positioning of one sequence, either nucleotide or amino acid, with that of another, to allow a comparison of the sequence of relevant portions of one with that of the other.
  • An example of one method of this procedure is given in Needleman et al. (J. Mol. Biol. 48:443-453, 1970). The method may be implemented conveniently by computer programs such as the Align program (DNAstar, Inc.).
  • homologous or functionally equivalent sequences include functionally equivalent arrangements of the cysteine residues within the conserved cysteine skeleton, including amino acid insertions or deletions which alter the linear arrangement of these cysteines, but do not materially impair their relationship in the folded structure of the protein.
  • Antisense DNA refers to the sequence of chromosomal DNA that is transcribed.
  • an “antisense probe” is a probe which comprises at least a portion of the antisense DNA for a nucleic acid portion of interest.
  • cloning is meant the use of in vitro recombination techniques to insert a particular gene or other DNA sequence into a vector molecule.
  • in vitro recombination techniques to insert a particular gene or other DNA sequence into a vector molecule.
  • it is necessary to employ methods for generating DNA fragments, for joining the fragments to vector molecules, for introducing the composite DNA molecule into a host cell in which it can replicate, and for selecting the clone having the target gene from amongst the recipient host cells.
  • cDNA is meant complementary or copy DNA produced from an RNA template by the action of RNA-dependent DNA polymerase (reverse transcriptase).
  • a “cDNA clone” means a duplex DNA sequence complementary to an RNA molecule of interest, carried in a cloning vector.
  • cDNA library is meant a collection of recombinant DNA molecules containing cDNA inserts which together comprise a representation of the mRNA molecules present in an entire organism or tissue, depending on the source of the RNA templates.
  • a cDNA library may be prepared by methods known to those of skill, and described, for example, in Maniatis et al., Molecular Cloning: A Laboratory Manual, supra.
  • RNA is first isolated from the cells of an organism from whose genome it is desired to clone a particular gene. Preferred for the purposes of the present invention are mammalian, and particularly human, cell lines.
  • RNA may be isolated from a tumor cell, derived from an animal tumor, and preferably from a human tumor.
  • a library may be prepared from, for example, a human adrenal tumor, but any tumor may be used.
  • DNA polymorphism refers to the condition in which two or more different nucleotide sequences can exist at a particular site in DNA.
  • “Expression vector” includes vectors which are capable of expressing DNA sequences contained therein, i.e., the coding sequences are operably linked to other sequences capable of effecting their expression. It is implied, although not always explicitly stated, that these expression vectors must be replicable in the host organisms either as episomes or as an integral part of the chromosomal DNA.
  • a useful, but not a necessary, element of an effective expression vector is a marker encoding sequence, which is a sequence encoding a protein which results in a phenotypic property (such as tetracycline resistance) of the cells containing the protein which permits those cells to be readily identified.
  • expression vector is given a functional definition, and any DNA sequence which is capable of effecting expression of a specified contained DNA code is included in this term, as it is applied to the specified sequence.
  • vectors are frequently in the form of plasmids, thus “plasmid” and “expression vector” are often used interchangeably.
  • the invention is intended to include such other forms of expression vectors which serve equivalent functions and which may, from time to time become known in the art.
  • a “functional derivative” is meant the “fragments”, “variants”, “analogs”, or “chemical derivatives” of a molecule.
  • a “fragment” of a molecule, such as any of the antigens of the present invention is meant to refer to any polypeptide subset of the molecule.
  • a “variant” of such molecules is meant to refer to a naturally occurring molecule substantially similar to either the entire molecule, or a fragment thereof.
  • An “analog” of a molecule is meant to refer to a non-natural molecule substantially similar to either the entire molecule or a fragment thereof.
  • gene means a polynucleotide sequence encoding a peptide.
  • homogeneous is meant, when referring to a peptide or DNA sequence, that the primary molecular structure (i.e., the sequence of amino acids or nucleotides) of substantially all molecules present in the composition under consideration is identical.
  • isolated refers to a protein of the present invention, or any gene encoding any such protein, which is essentially free of other proteins or genes, respectively, or of other contaminants with which it might normally be found in nature, and as such exists in a form not found in nature.
  • label refers to a molecular moiety capable of detection including, by way of example, without limitation, radioactive isotopes, enzymes, luminescent agents, and dyes.
  • probe refers to a ligand of known qualities capable of selectively binding to a target antiligand.
  • probe refers to a strand of nucleic acid having a base sequence complementary to a target strand.
  • Recombinant host cells refers to cells which have been transformed with vectors constructed using recombinant DNA techniques. As defined herein, the antibody or modification thereof produced by a recombinant host cell is by virtue of this transformation, rather than in such lesser amounts, or more commonly, in such less than detectable amounts, as would be produced by the untransformed host.
  • substantially pure is meant any protein of the present invention, or any gene encoding any such protein, which is essentially free of other proteins or genes, respectively, or of other contaminants with which it might normally be found in nature, and as such exists in a form not found in nature.
  • a molecule is said to be “substantially similar” to another molecule if the sequence of amino acids in both molecules is substantially the same, and if both molecules possess a similar biological activity. Thus, provided that two molecules possess a similar activity, they are considered variants as that term is used herein even if one of the molecules contains additional amino acid residues not found in the other, or if the sequence of amino acid residues is not identical.
  • a molecule is said to be a “chemical derivative” of another molecule when it contains additional chemical moieties not normally a part of the molecule. Such moieties may improve the molecule's solubility, absorption, biological half life, etc.
  • the moieties may alternatively decrease the toxicity of the molecule, eliminate or attenuate any undesirable side effect of the molecule, etc.
  • Moieties capable of mediating such effects are disclosed, for example, in Remington's Pharmaceutical Sciences. 16th ed., Mack Publishing Co., Easton, Pa. (1980).
  • vector is meant a DNA molecule, derived from a plasmid or bacteriophage, into which fragments of DNA may be inserted or cloned.
  • a vector will contain one or more unique restriction sites, and may be capable of autonomous replication in a defined host or vehicle organism such that the cloned sequence is reproducible.
  • the invention includes the cDNA of SEQ ID NO: 1, SEQ ID NO:2, SEQ ID NO:4 or SEQ ID NO:6, as well as sequences which include the sequence of SEQ ID NO: 1, SEQ ID NO:2, SEQ ID NO:4, or SEQ ID NO:6, and derivatives of these sequences.
  • the invention also includes vectors, liposomes and other carrier vehicles which encompass these sequence or derivatives of them.
  • the invention further includes proteins transcribed from SEQ ID NO:1, SEQ ID NO:2, SEQ ID NO:4, or SEQ ID NO:6, including but not limited to SEQ ID NO:3, SEQ ID NO:5 or SEQ ID NO:7, and their derivatives and variants.
  • One embodiment of the invention includes soluble variants of a KIM protein that is usually synthesized as a membrane associated protein, and which is upregulated after injury.
  • Soluble variants lack at least a portion of the transmembrane or intra-membrane section of a native KIM protein.
  • the soluble variant lacks the entire transmembrane or intra-membrane section of a native KIM protein.
  • the present soluble variants can be derived from intact or native KIM proteins by any suitable means, including chemical or enzymatic cleavage, or through molecular engineering techniques such by expression of a truncated KIM nucleic acid.
  • the present soluble variants should be understood to be derivatives of an intact KIM.
  • Soluble variants include fusion proteins which encompass derivatives of KIM proteins that lack at least a portion of the transmembrane or intra-membrane section of a native KIM protein. All types of KIM fusion proteins are included, particularly those which incorporate his-tag, Ig-tag, and myc-tag forms of the molecule. These KIM fusions may have characteristics which are therapeutically advantageous, such as the increased half-life conferred by the Ig-tag. Also included are fusion proteins which incorporate portions of selected domains of the KIM protein.
  • Variants can differ from naturally occurring KIM protein in amino acid sequence or in ways that do not involve sequence, or both. Variants in amino acid sequence are produced when one or more amino acids in naturally occurring KIM protein is substituted with a different natural amino acid, an amino acid derivative or non-native amino acid. Particularly preferred variants include naturally occurring KIM protein, or biologically active fragments of naturally occurring KIM protein, whose sequences differ from the wild type sequence by one or more conservative amino acid substitutions, which typically have minimal influence on the secondary structure and hydrophobic nature of the protein or peptide. Variants may also have sequences which differ by one or more non-conservative amino acid substitutions, deletions or insertions which do not abolish the KIM protein biological activity.
  • Conservative substitutions typically include the substitution of one amino acid for another with similar characteristics such as substitutions within the following groups: valine, glycine; glycine, alanine; valine, isoleucine; aspartic acid, glutamic acid; asparagine, glutamine; serine, threonine; lysine, arginine; and phenylalanine, tyrosine.
  • the non-polar (hydrophobic) amino acids include alanine, leucine, isoleucine, valine, proline, phenylalanine, tryptophan and methionine.
  • the polar neutral amino acids include glycine, serine, threonine, cysteine, tyrosine, asparagine and glutamine.
  • the positively charged (basic) amino acids include arginine, lysine and histidine.
  • the negatively charged (acidic) amino acids include aspartic acid and glutamic acid.
  • variants within the invention are those with modifications which increase peptide stability.
  • Such variants may contain, for example, one or more non-peptide bonds (which replace the peptide bonds) in the peptide sequence.
  • variants that include residues other than naturally occurring L-amino acids such as D-amino acids or non-naturally occurring or synthetic amino acids such as beta or gamma amino acids and cyclic variants. Incorporation of D- instead of L-amino acids into the polypeptide may increase its resistance to proteases. See, e.g., U.S. Pat. No. 5,219,990.
  • substitutions that may be expected to induce changes in the functional properties of KIM polypeptides are those in which: (I) a hydrophilic residue, e.g., serine or threonine, is substituted by a hydrophobic residue, e.g., leucine, isoleucine, phenylalanine, or alanine; (ii) a cysteine residue is substituted for (or by) any other residue; (iii) a residue having an electropositive side chain, e.g., lysine, arginine or histidine, is substituted for (or by) a residue having an electronegative charge, e.g., glutamic acid or aspartic acid; or (iv) a residue having a bulky side chain, e.g., phenylalanine, is substituted for (or by) one not having such a side chain, e.g., glycine.
  • a hydrophilic residue e.g., serine or
  • the peptides of this invention may also be modified by various changes such as insertions, deletions and substitutions, either conservative or nonconservative where such changes might provide for certain advantages in their use. Splice variants are specifically included in the invention.
  • variants with amino acid substitutions which are less conservative may also result in desired derivatives, e.g., by causing changes in charge, conformation and other biological properties.
  • substitutions would include for example, substitution of hydrophilic residue for a hydrophobic residue, substitution of a cysteine or proline for another residue, substitution of a residue having a small side chain for a residue having a bulky side chain or substitution of a residue having a net positive charge for a residue having a net negative charge.
  • the derivatives may be readily assayed according to the methods disclosed herein to determine the presence or absence of the desired characteristics.
  • the scope of the invention includes proteins and peptides with variant amino acid sequences (sequence variants) having at least eighty percent similarity with a KIM protein. More preferably the sequence similarity is at least ninety percent, or at least ninety-five percent.
  • a sequence variant within the scope of the invention is any which shares at least eighty percent similarity with a KIM selected from rat KIM1 (SEQ ID NO:3) and human KIM1 (SEQ ID NO: 7).
  • the percentage of similarity between a variant sequence and a reference sequence is determined after the variant is aligned with the reference, preferably according to the method of Needelman et al.
  • the percentage of sequence “identity” of the variant to the reference is the percentage of amino acid or nucleic acid residues of the variant sequence that are identical to the aligned (corresponding) residues of the reference.
  • the percentage of sequence “similarity” is the sum of the percentage of identical corresponding residues and the percentage of variant residues that are conservative substitutions for the aligned (corresponding) reference residues.
  • a number of algorithms and computer software programs can be utilized to determine whether a sequence of interest is a KIM sequence variant within the scope of this invention.
  • One such is the algorithm of Karlin and Altschul (1990) Proc. Natl. Acad. Sci. USA 87:2264-68, as modified in Karlin and Altschul (1993), Proc. Natl. Acad. Sci. USA 90:5873-77. This algorithm is incorporated into the NBLAST and XBLAST programs of Altschul et al. (1990) J. Mol. Biol. 215:403-10.
  • BLAST nucleotide searches can be performed with NBLAST.
  • BLAST protein searches can be performed with XBLAST.
  • gapped BLAST can be utilized as described in Altschul et al. (1997) Nucl. Acids Res. 25(17):3389-3402. All of the foregoing are available through http: ⁇ www.ncbi.nlm.nih.gov. Another suitable algorithm is that of Myers and Miller, CABIOS (1989), which is incorporated into the ALIGN program (version 2.0), which is a part of the Wisconsin Sequence Analysis package of the Genetics Computing Group (GCG).
  • GCG Genetics Computing Group
  • variants within the invention include any polypeptide or nucleic acid sequence which is identified through BLAST or CABIOS analysis as being related to a KIM nucleic acid or polypeptide sequence disclosed herein (e.g., rat KIM1, SEQ ID NO:3 or human KIM1, SEQ ID NO:7).
  • KIM nucleic acid or polypeptide sequence disclosed herein e.g., rat KIM1, SEQ ID NO:3 or human KIM1, SEQ ID NO:7.
  • “Related” sequences are those for which, in the judgement of an ordinarily skilled practitioner, the results of sequence analysis indicate a biologically significant relationship with the reference sequence (e.g., rat KIM1 or human KIM1).
  • sequence variants of the KIM proteins disclosed herein are those having an amino acid sequence which is at least forty (40) percent identical overall (i.e., when aligned) to a disclosed KIM polypeptide sequence, and sharing at least 80% of the aligned KIM cysteine residues.
  • Particularly preferred sequence variants are those which meet the foregoing analysis criteria when analyzed, relative to a KIM reference sequence disclosed herein (e.g., rat KIM1, SEQ ID NO:3 or human KIM1, SEQ ID NO:7), using the program GAP, which is a part of the Wisconsin analysis package of the GCG.
  • the gap creation penalty (gap weight) should be set to 3.0
  • the gap extension penalty (gap length) should be set to 0.1.
  • GAP uses the algorithm of Needelman et al. (1970), 48 J. Mol. Biol. 443-453, to create an alignment with the largest number of matched (aligned identical) residues and the fewest number of sequence gaps.
  • Non-sequence modifications may include, for example, in vivo or in vitro chemical derivatization of portions of naturally occurring KIM protein, as well as changes in acetylation, methylation, phosphorylation, carboxylation or glycosylation.
  • agents which specifically bind to the protein, or a fragment of the protein include ligands and antibodies (including monoclonal, single chain, double chain, Fab fragments, and others, whether native, human, humanized, primatized, or chimeric). Additional descriptions of these categories of agents are in PCT application 95/16709, the specification of which is herein incorporated by reference.
  • Ischemic injured rat kidneys are generated as described by Witzgall et al. (J. Clin Invest. 93: 2175-2188, 1994). Briefly, the renal artery and vein from one kidney of an adult Sprague-Dawley rat are clamped for 40 minutes and then reperfused. Injured kidneys are harvested from the rats at 24 hours and at 48 hours after reperfusion. Kidneys from sham-operated, normal adult Sprague-Dawley rats are also harvested.
  • RNA is prepared from the organs based on the protocol by Glisin et al. (Biochemistry 13: 2633, 1974). Briefly, the harvested organs are placed immediately into GNC buffer (4M guanidine thiocyanate, 0.5% SDS, 25 mM sodium citrate, 0.1% Sigma anti foam) and disrupted. on ice with a polytron. Cell debris is removed with a low speed spin in a clinical centrifuge and the supernatant fluid is placed on a 5.7 M CsCl, 25 mM sodium acetate, 1 mM EDTA cushion. RNA is pelleted through the cushion in a SW40Ti rotor at 22K for 15 hrs.
  • GNC buffer M guanidine thiocyanate, 0.5% SDS, 25 mM sodium citrate, 0.1% Sigma anti foam
  • RNA is resuspended in sterile DEPC-treated water, precipitated twice with 1/10 volume 3M sodium acetate and 2.5 volumes of EtOH.
  • Poly A+ RNA is isolated using an mRNA purification kit (Pharmacia, catalog No.27-9258-02).
  • Double stranded cDNA is synthesized from sham-operated and from 48 hr post-ischemic kidney poly A+ RNA using Gibco BRL “Superscript ChoiceTM System cDNA Synthesis Kit”, catalog No. 18090.
  • First strand is synthesized by priming with oligo dT and using Superscript IITM reverse transcriptase.
  • Second strand is generated using E. coli DNA polymerase I and RNase H followed by T4 DNA polymerase using BRL recommended conditions.
  • RDA analysis is performed essentially as described by Hubank and Schatz (Nucleic Acid Research 22: 5640-48, 1994). Briefly, 48 hr post-ischemic kidney cDNA is digested with the restriction enzyme Dpn II, and ligated to R-Bgl-12/24 oligonucleotides (see reference for exact sequence). PCR amplification (performed with Perkin-Elmer Taq polymerase and their corresponding PCR buffer) of the linker ligated cDNA is used to generate the initial representation. This PCR product is designated “tester amplicon.” The same procedure is used to generate “driver amplicon” from sham-operated rat kidney cDNA.
  • Hybridization of tester and driver amplicons followed by selective amplification are performed three times to generate Differential Product One (DP1), Two (DP2) and Three (DP3).
  • Generation of the DP1 product is performed as described by Hubank and Schatz (Nucleic Acid Research 22: 5640-48, 1994).
  • the DP2 and DP3 products are also generated as described by Hubank and Schatz (id.), except that the driver:tester ratios are changed to 5,333:1 for DP2 and to 40,000:1 or 4,000:1 for DP3.
  • RDA product 1-7 (252 bp) when the DP3 was generated using a ratio of 40,000:1
  • product RDA 3-2 (445 bp) and 4-7 (483 bp) when the DP3 was generated using a ratio of 4,000:1.
  • the DNA fragments are subcloned using the Pharmacia SurecloneTM kit (catalog No. 27-9300-01) to repair the ends of the PCR fragments with Klenow enzyme and to facilitate blunt end ligation of the fragments into the pUC18 vector.
  • RNA Poly A+ RNA (2.5 ⁇ g) from rat normal adult kidney (sham operated), from 48 hr post-ischemic injured adult kidney, and from day 18 embryonic kidney is electrophoresed and Northern blotted (Cate, Cell 45:685, 1986) to a GeneScreenTM membrane (Dupont).
  • Hybridization in PSB buffer 50 mM Tris 7.5, 1M NaCl, 0.1% Na pyrophosphate, 0.2% PVP, 0.2% Ficoll, 0.2% BSA, 1% SDS
  • containing 10% dextran sulphate and 100 ⁇ g/ml tRNA is performed at 65 C using three different probes: 1-7 RDA product, 3-2 RDA product and 4-7 RDA product.
  • RDA products 1-7, 3-2 and 4-7 hybridize to mRNAs present in all three samples, but most intensely to mRNAs in the 48 hr post-ischemic kidney RNA samples.
  • a Northern blot analysis of adult rat tissues indicates that the 1-7 gene is expressed at very low levels in normal adult kidney, testis, spleen and lung.
  • the 3-2 gene is expressed in liver, kidney, spleen, and brain.
  • the 4-7 gene is expressed in spleen, kidney, lung, testis, heart, brain, liver, and skeletal muscle.
  • the presence of different sized mRNAs in some tissues in the 1-7 and 3-2 blot indicates that the primary transcription product of the 1-7 gene and of the 3-2 gene may undergo alternate splicing and/or polyadenylation.
  • a cDNA library is generated from 4 ⁇ g of polyA+ RNA from 48 hr post-ischemic injured kidney using reagents from BRL Superscript ChoiceTM System for cDNA synthesis, and StratageneTM Lambda ZapII cloning-kit (catalog No. 236201), according to protocols recommended by the manufacturers.
  • coli K-12, SOLR/p3-2#5-1) has been deposited as ATCC No. 98061.
  • the sequence of cDNA clone 3-2 is identical to that of clone 1-7 cDNA (SEQ ID NO: 2), except that nucleotides 136-605 of SEQ ID NO:1 represent an insertion.
  • SEQ ID NO:2 represents a splice variant form of SEQ ID NO: 1.
  • the clone for 1-7 E. coli K-12, SOLR/p1-7#3-1) has been deposited as ATCC No. 98060.
  • the sequence of 3-2 cDNA ( FIG. 1 ; SEQ ID NO:1) contains an open reading frame of 307 amino acids ( FIG. 1 ; SEQ ID NO:3).
  • a signal sequence of 21 amino acids is inferred from Von Heijne analysis (Von Heijne et al., Nucl. Acid Res. 14:14683 (1986)), and a transmembrane region spanning approximately aa 235-257 indicates that the 3-2 product is a cell surface protein.
  • the sequence of 1-7 cDNA ( FIG. 2 ; SEQ ID NO:2) contains an open reading frame of 307 amino acids, which is identical to the open reading frame contained in the 3-2 cDNA (SEQ ID NO: 3). This 307 amino acid polypeptide is herein designated rat KIM1.
  • sequence of 4-7 cDNA ( FIG. 3 ; SEQ ID NO:4) contains an open reading frame of 572 amino acids (SEQ ID NO:5). A transmembrane region is located at approximately amino acids 501-521. This 572 amino acid polypeptide is related to the polypeptide known as nmb. Weterman et al. (1995) 60 Int. J. Cancer 73-81.
  • In situ hybridization is carried out according to the method described by Finch et al., Dev. Dynamics 203: 223-240, 1995. Briefly, both ischemic and contralateral kidneys are perfusion fixed with 4% paraformaldehyde in PBS. Kidneys are further fixed overnight at 4 C and processed. Paraffin sections are deparaffinized and rehydrated, fixed with 4% paraformaldehyde in PBS, digested with proteinase K, refixed, then acetylated with acetic anhydride in triethanolamine buffer. Sections are then dehydrated and hybridized with 32 P-labeled riboprobes at 55° C.
  • RNA expression pattern also reveals abundant expression of this gene in the injured ischemic kidney compared to the normal adult kidney. The site of expression appears to be infiltrating cells.
  • a 32 P-labeled DNA probe comprising nucleotides 546-969 of the insert of clone 3-2 shown in FIG. 1 is generated and used to screen a human embryonic liver lambda gt10 cDNA library (Clontech Catalog #HL5003a). 1 ⁇ 10 6 plaques are screened in duplicate using standard conditions as described above but temperature for screening was 55 C. For the high stringency wash, the filters are washed in 2 ⁇ SSC at 55 C. Fifty positive phage are identified and plaque purified, and DNA is prepared. The phage DNAs are subjected to Southern analysis using the same probe as above. The Southern blot filter is subjected to a final wash with 0.5 ⁇ SSC at 55 C. Two clones are identified as positive. The insert of clone H13-10-85 is sequenced and a region is found that encodes a protein with a high level of identity to the 3-2 protein shown in FIG. 3 .
  • the nucleotide sequence (SEQ ID NO:6) and predicted amino acid sequence (SEQ ID NO:7) of the human 3-2 related protein are shown in FIG. 4 .
  • the human 3-2 related protein is 43.8% identical and 59.1% similar to the rat 3-2 protein. Both contain IgG, mucin, transmembrane, and cytoplasmic domains. The six cysteines within the IgG domains of both proteins are conserved. Accordingly, the polypeptide of SEQ D NO:7 is designated human KIM1 (or KIM-1) herein. It is believed that the mammalian genome (e.g., the human genome) includes genes encoding a family of KIM-1 related proteins. Thus, the human KIM-1 disclosed herein is more precisely designated human KIM-1a.
  • a fusion protein of the extracellular, domain of KIM and the Fc region of immunoglobulin (Ig) is a useful tool for the study of the molecular and cellular biology of the injured/regenerating kidney and as a therapeutic molecule.
  • a fragment of the extracellular domain of KIM-1 cDNA was amplified by PCR and cloned in the Biogen expression vector, pCA125, for transient expression in COS cells.
  • the expression vector pCA125 produces a fusion protein which has a structure from gene cloned at N-terminus and a human Ig Fc region at the C-terminus.
  • COS cells were transfected with the plasmids SJR 103 or 104; these plasmids express a fusion protein which contains the human KIM sequences 263-1147 (SEQ ID NO:6; SJR 103) or rat KIM sequences 599-1319 (SEQ ID NO:1; SJR 104) of the extracellular domain fused to human Ig Fc region.
  • the cells were grown in 10% FBS in DMEM in the cell factory (Nunc, Naperville, Ill.). Two to three days post-transfection, medium was harvested, concentrated using Amicon concentrator, and fusion protein was purified using Protein-A Sepharose column. After purification, purity of fusion protein was evaluated by SDS-PAGE.
  • Antibody production involves administration of one or more immunogenic doses of a KIM polypeptide preparation (whether isolated, or as part of a cell suspension, fraction or extract) to an appropriate non-human animal, such as a mouse, rat, rabbit, gunea pig, turkey, goat, sheep, pig or horse.
  • a conventional adjuvant e.g., Complete Freund's Adjuvant, CFA.
  • Serum immunoglobulins (Ig) using peripheral blood samples withdrawn at intervals (e.g., weekly) after initial or a subsequent immunization, are monitored to detect the onset and/or maturation of a humoral immune response to KIM.
  • any conventional technique e.g., ELISA, RIA, Western blotting, or the like, can be used to detect and/or quantitate (titer) the KIM-specific Ig.
  • High titer immune sera are preferred for use as anti-KIM polyclonal antibodies herein.
  • High titer sera e.g., having a titer of at least 1:1000
  • Animals e.g., mice, rats, guinea pigs, hamsters having a high serum titer of anti-KIM Ig also can be used for the conventional production of monoclonal antibodies (MAb) with binding specificity for KIM.
  • Splenocytes are isolated from such animals and are fused with a myeloma cell line according to standard techniques to produce hybridomas.
  • hybridomas are cloned by limiting dilution, fluorescence-activated cell sorting, or any other suitable cell isolation technique, to produce monoclonal hybridoma cell lines of the present invention.
  • Hybridoma lines of the present invention are useful for the production of anti-KIM MAbs, and also as sources from which nucleic acids encoding the present anti-KIM MAbs can be routinely isolated.
  • nucleic acids, encoding heavy and/or light chain polypeptides of the present MAbs can be used to produce recombinant, engineered versions of the present MAbs, including truncated MAb polypeptides, chimeric or humanized MAbs, and MAb fusion proteins.
  • mice have been immunized with a human KIM1-Ig fusion protein as disclosed herein, which was immobilized on Protein A Sepharose beads and emulsified in CFA.
  • the fusion protein included the extracellular domain of human KIM1 (amino acids 1 to 291) fused to a truncated human IgG1 polypeptide comprising hinge-C2-C3 domains of the IgG1 heavy chain polypeptide.
  • the onset and maturation of a humoral immune response was monitored using a conventional ELISA, in which another Ig fusion protein was used as a blocking agent to subtract serum Ig's having undesired specificity for the IgG1 portion of the KIM1 fusion protein.
  • MAbs were obtained with desirable binding specificity for human KIM1.
  • AKG7 is preferred herein.
  • the AKG7 antibody is useful in ELISA, Western blot and immunohistochemistry analysis of human KIM1 polypeptides.
  • the present technique can be repeated and/or adapted routinely to produce any number of MAbs to domains (e.g., the extracellular domain) of KIM proteins, that have the KIM-specific binding properties of MAb AKG7 and are useful in the analytical, diagnostic and therapeutic methods described herein.
  • MAbs of the present invention are obtained from cell culture media conditioned by the present hybridoma lines, or from ascites fluid, or from media conditioned by an engineered host cell expressing a heavy and/or light chain nucleic acid construct encoding an anti-KIM MAb of the present invention.
  • Such MAbs are purified using conventional immunoaffinity chromatography, and used in the various diagnostic, analytical and therapeutic methods described herein.
  • Anti-KIM antibodies of the invention which specifically bind to the protein of SEQ ID NO:3, SEQ ID NO:5 or SEQ D NO:7 or a fragment thereof, are useful in several diagnostic methods. These agents may be labeled with detectable markers, such as fluoroscopically or radiographically opaque substances, and administered to a subject to allow imaging of tissues which express KIM protein. The agents may also be bound to substances, such as horseradish peroxidase, which can be used as immunocytochemical stains to allow visualization of areas of KIM protein-positive cells on histological sections. A specific antibody could be used alone in this manner, and sites where it is bound can be visualized in a sandwich assay using an anti-immunoglobulin antibody which is itself bound to a detectable marker.
  • detectable markers such as fluoroscopically or radiographically opaque substances
  • the agents may also be bound to substances, such as horseradish peroxidase, which can be used as immunocytochemical stains to allow visualization of areas of KIM protein-positive cells on histological sections
  • the AKG7 MAb can be used to visualize cells producing KIM-1 protein in immunohistochemistry analysis of human renal tissue samples. Immunohistochemistry is carried out on paraformaldehyde-fixed paraffin sections by deparaffinizing the sections, then ablating endogenous peroxidase activity by incubation in 2% hydrogen peroxide in methanol for 20 min. Sections are then heated in a microwave oven in 0.1 M citrate buffer, pH 6.0, for 10 min., then blocked with diluted goat serum (1:67) overnight at 4° C. Thereafter, sections are incubated with affinity purified AKG7 or control IgG at a concentration of 5 ⁇ g/mL.
  • the sections are washed in PBS and incubated with biotinylated goat anti-mouse IgG for 30 min. After further washes with PBS, the sections are incubated with avidin-biotinylated horseradish peroxidase complex (Vectastain Elite ABC kit, Vector Laboratories) for 1 hr.
  • Sections are washed in PBS and developed by the addition of 50 mM sodium phosphate buffer, pH 7.6, containing 0.12% 3,3-diaminobenzidine tetrahydrochloride (Sigma), 0.0075% nickel chloride, 0.0075% cobalt chloride, and 0.0075% hydrogen peroxide for 2-5 min., producing a dark brown stain indicating immunoreactivity. Sections are then counterstained with 0.01% toluidine blue. This technique has been used to visualize KIM-1 immunoreactivity in paraffin sections from two different human patients with renal cell carcinoma. Sections comprising tumor tissue and adjacent normal renal tissue were assessed. In patients, strong KIM-1 immunoreactivity was noted in tumor tissue.
  • the invention includes a method of diagnosing renal injury, or of monitoring a process of renal repair, by measuring the concentration of KIM or of KIM fragments in the urine, plasma or serum of a patient.
  • KIM can be measured in urine sediment, in particular in cellular debris in the urine sediment. Casts of renal tubule cells, which may be present in urine sediment from patients with ongoing renal disease, may contain elevated levels of KIM protein and mRNA.
  • Specific antibodies to KIM protein may also be bound to solid supports, such as beads or dishes, and used to remove the ligand from a solution, either for measurement, or for purification and characterization of the protein or its attributes (such as posttranslational modifications). Such characterization of a patient's KIM protein might be useful in identifying deleterious mutants or processing defects which interfere with KIM function and are associated with abnormal patient phenotypes. Each of these techniques is routine to those of skill in the immunological arts.
  • Additional imaging methods utilize KIM or KIM fragments, fused to imageable moieties, for diagnostic imaging of tissues that express KIM ligands, particularly tumors.
  • kidney homogenates of contralateral and postischemic kidneys were examined 24 and 48 hours following a 40 minute clamping of the renal artery and vein of a single kidney for each rat.
  • the kidney homogenate was assessed for the presence of KIM-1 protein.
  • Western blot analysis identifies three proteins detected by two different antibodies after ischemic injury, which are not detectable in homogenates from contralateral kidneys which were not exposed to ischemic injury. The apparent molecular weights of the bands are approximately 40 kDa, 50 kDa and 70-80 kDa.
  • the three protein species detected by western blotting could represent glycosylated forms of the same protein given the presence of potential N and O linked glycosylation sites.
  • the KIM-1 sequence presents two putative sites for N-glycosylation and a mucin domain where O-glycosylation could cover the,polypeptide chain.
  • the three KIM-1 bands detected in postischemic kidney could correspond to glycosylation variants of the same core protein.
  • De-N-glycosylation with PNGase F resulted in a shift of all three bands to a lower molecular weight, corresponding to a loss of about 3 kDa, indicating that all three proteins are N-glycosylated. Differences in O-glycosylation might explain the differences in sizes of these three bands.
  • RNA samples of interest e.g., kidney biopsy or necropsy samples, or control samples
  • size fractionated by electrophoresis e.g., size fractionated by electrophoresis
  • transferred to a GeneScreen membrane e.g., GeneScreen membrane
  • Hybridization with a KIM-1 probe corresponding to part or all of the open reading frame of the KIM-1 gene of interest is carried out in plaque screening buffer (PSB, 50 mM Tris, pH 7.5, 1 M NaCl, 0.1% sodium pyrophosphate, 0.2% polyvinylpyrrolidone, 0.2% Ficoll, 0.2% bovine serum albumin, 1% SDS) containing 10% dextran sulfate and 100 ⁇ g/mL tRNA at 65° C.
  • the KIM-1 probe is preferably detectably labeled, e.g., radiolabelled.
  • the membrane is then washed to remove unbound probe, and KIM-1 mRNA is visualized using standard membrane development techniques. Thereafter, the KIM-1 probe is stripped from the blot, and the blot is rehybridized with a control probe, such as a ⁇ -actin probe.
  • a control probe such as a ⁇ -actin probe.
  • FIG. 6 The results of an exemplary Northern blot analysis are set forth in FIG. 6 : biopsy samples containing histologically normal renal tissue were compared to renal tumor tissues (renal cell carcinoma) obtained from four different patients afflicted with renal cancers. KIM-1 expression was virtually undetectable in three of the four normal samples and was weakly detectable in the fourth. In contract, two of the four renal cancer samples displayed strong KIM-1 expression. These results are consistent with the utility of KIM-1 probes as reagents for the diagnosis, prognostication or staging of renal cancers. Without being limited hereby, it is believed that aberrant KIM-1 gene expression may correlate with the degree of neoplastic transformation, agressiveness or metastatic propensity of a given renal cell carcinoma. Thus, assessment of KIM-1 gene expression is believed to provide information useful in conjunction with other, conventional diagnostic and/or clinical criteria for characterizing renal cancers.
  • the therapeutic methods of the invention involve selectively promoting or inhibiting cellular responses that are dependent on KIM ligation.
  • the KIM and the KIM ligand are both membrane bound, and expressed by different cells, the signal transduction may occur in the KIM-expressing cell, in the KIM ligand-expressing cell, or in both.
  • KIM ligation-triggered response in a KIM ligand-expressing cell may be generated by contacting the cell with exogenous KIM, KIM fusion proteins or activating antibodies against KIM ligand, either in vitro or in vivo. Further, responses of the KIM ligand-expressing cell that would otherwise be triggered by endogenous KIM could be blocked by contacting the KIM ligand-expressing cell with a KIM ligand antagonist (e.g., an antagonist antibody that binds to KIM ligand), or by contacting the endogenous KIM with an anti-KIM antibody or other KIM-binding molecule which prevents the effective ligation of KIM with a KIM ligand.
  • a KIM ligand antagonist e.g., an antagonist antibody that binds to KIM ligand
  • KIM ligation-triggered response in a KIM-expressing cell may be generated by contacting the cell with a soluble KIM ligand, or certain anti-KIM activating antibodies.
  • responses of the KIM-expressing cell that would otherwise be triggered by interaction with endogenous KIM ligand could be blocked by contacting the KIM-expressing cell with an antagonist to KIM (e.g., a blocking antibody that binds to KIM in a manner that prevents effective, signal-generating KIM ligation), or by contacting the endogenous KIM ligand with an anti-KIM ligand antibody or other KIM ligand-binding molecule which prevents the effective ligation of KIM with the KIM ligand.
  • an antagonist to KIM e.g., a blocking antibody that binds to KIM in a manner that prevents effective, signal-generating KIM ligation
  • an anti-KIM ligand antibody or other KIM ligand-binding molecule which prevents the effective ligation of KIM with the KIM ligand.
  • One therapeutic use of the KIM-related compounds of the invention is for treating a subject with renal disease, promoting growth of new tissue in a subject, or promoting survival of damaged tissue in a subject, and includes the step of administering to the subject a therapeutically effective amount of a KIM protein of the invention, or of a pharmaceutical composition which includes a protein of the invention.
  • the protein used in these methods may be a fragment of a full-length KIM protein, a soluble KIM ligand protein or fusion fragment, or a KIM agonist.
  • These methods may also be practiced by administering to the subject a therapeutically effective amount of an agonist antibody of the invention, or a pharmaceutical composition which includes an agonist antibody of the invention.
  • a KIM protein may be administered concurrently with a therapeutically effective amount of a second compound which exerts a medically desirable adjunct effect.
  • tissues of interest for these methods may include any tissue, preferred tissues include renal tissue, liver, neural tissue, heart, stomach, small intestine, spinal cord, or lung.
  • Particular renal conditions which may be beneficially treated with the compounds of the invention include acute renal failure, acute nephritis, chronic renal failure, nephrotic syndrome, renal tubule defects, kidney transplants, toxic injury, hypoxic injury, and trauma.
  • Renal tubule defects include those of either hereditary or acquired nature, such as polycystic renal disease, medullary cystic disease, and medullary sponge kidney. This list is not limited, and may include many other renal disorders (see, e.g., Harrison's Principles of Internal Medicine, 13th ed., 1994, which is herein incorporated by reference.)
  • the subject of the methods may be human.
  • a therapeutic intervention for inhibiting growth of undesirable, KIM ligand-expressing tissue in a subject includes the step of administering to the subject a therapeutically effective amount of a KIM antagonist (e.g., an antagonist antibody that binds to KIM ligand), or by administering a therapeutically effective amount of an anti-KIM antibody or other KIM-binding molecule which blocks KIM binding to the KIM ligand-expressing tissue.
  • a KIM antagonist e.g., an antagonist antibody that binds to KIM ligand
  • an anti-KIM antibody or other KIM-binding molecule which blocks KIM binding to the KIM ligand-expressing tissue.
  • the KIM antagonist or anti-KIM antibody may be used therapeutically to inhibit or block growth of tumors which depend on KIM protein for growth.
  • Other methods of the invention include killing KIM ligand-expressing tumor cells, or inhibiting their growth, by contacting the cells with a fusion protein of a KIM and a toxin or radionuclide, or an anti-KIM ligand antibody conjugated to a toxin or radionuclide.
  • the cell may be within a subject, and the protein or the conjugated antibody is administered to the subject.
  • Also encompassed within the invention is a method for targeting a toxin or radionuclide to a cell expressing a KIM, comprising contacting the cell with a fusion protein comprising a KIM ligand and a toxin or radionuclide, or an anti-KIM antibody conjugated to a toxin or radionuclide.
  • Another embodiment includes the method of suppressing growth of a tumor cell which expresses KIM, comprising contacting the cell with a fusion protein of KIM ligand and a toxin or radionuclide or with an anti-KIM antibody conjugated to a toxin or radionuclide; the cell may be within a subject, and the protein administered to the subject.
  • subject used herein is taken to mean any mammal to which KIM may be administered.
  • Subjects specifically intended for treatment with the method of the invention include humans, as well as nonhuman primates, sheep, horses, cattle, goats, pigs, dogs, cats, rabbits, guinea pigs, hamsters, gerbils, rats and mice, as well as the organs, tumors, and cells derived or originating from these hosts.
  • the KIM genes of the invention are introduced into damaged tissue, or into tissue where stimulated growth is desirable. Such gene therapy stimulates production of KIM protein by the transfected cells, promoting cell growth and/or survival of cells that express the KIM protein.
  • a gene coding for a KIM protein may be introduced into a renal target tissue.
  • the KIM protein would be stably expressed and stimulate tissue growth, division, or differentiation, or could potentiate cell survival.
  • a KIM gene may be introduced into a target cell using a variety of well-known methods that use either viral or non-viral based strategies.
  • Non-viral methods include electroporation, membrane fusion with liposomes, high velocity bombardment with DNA-coated microprojectiles, incubation with calcium-phosphate-DNA precipitate, DEAE-dextran mediated transfection, and direct micro-injection into single cells.
  • a KIM gene may be introduced into a cell by calcium phosphate coprecipitation (Pillicer et al., Science, 209: 1414-1422 (1980); mechanical microinjection and/or particle acceleration (Anderson et al., Proc. Nat. Acad. Sci.
  • liposome based DNA transfer e.g., LIPOFECTIN-mediated transfection—Fefgner et al., Proc. Nat. Acad. Sci., USA, 84: 471-477,1987; Gao and Huang, Biochim. Biophys. Res. Comm., 179: 280-285, 1991; DEAE Dextran-mediated transfection; electroporation (U.S. Pat. No.
  • Target cells may be transfected with the genes of the invention by direct gene transfer. See, e.g., Wolff et al., “Direct Gene Transfer Into Moose Muscle In Vivo”, Science 247:1465-68, 1990. In many cases, vector-mediated transfection will be desirable. Any of the methods known in the art for the insertion of polynucleotide sequences into a vector may be used. ( See, for example, Sambrook et al., Molecular Cloning: A Laboratory Manual, Cold Spring Harbor Laboratory, Cold Spring Harbor, N.Y., 1989; and Ausubel et al., Current Protocols in Molecular Biology, J.
  • Promoter activation may be tissue specific or inducible by a metabolic product or administered substance.
  • promoters/enhancers include, but are not limited to, the native c-ret ligand protein promoter, the cytomegalovirus immediate-early promoter/enhancer (Karasuyama et al., J. Exp. Med., 169: 13, 1989); the human beta-actin promoter (Gunning et al., Proc. Nat. Acad. Sci.
  • MMTV LTR mouse mammary tumor virus long terminal repeat
  • MoLV LTR Moloney murine leukemia virus
  • SSV Rous sarcoma virus
  • HSV herpes simplex virus
  • the KIM genes may also be introduced by specific viral vectors for use in gene transfer systems which are now well established. See for example: Madzak et al., J. Gen. Virol., 73: 1533-36, 1992 (papovavirus SV40); Berkner et al., Curr. Top. Microbiol. Immunol., 158: 39-61, 1992 (adenovirus); Hofmann et al., Proc. Natl. Acad. Sci. 92: 10099-10103, 1995 (baculovirus); Moss et al., Curr. Top. Microbiol. Immunol., 158: 25-38, 1992 (vaccinia virus); Muzyczka, Curr. Top. Microbiol.
  • Preferred vectors are DNA viruses that include adenoviruses (preferably Ad-2 or Ad-5 based vectors), baculovirus, herpes viruses (preferably herpes simplex virus based vectors), and parvoviruses (preferably “defective” or non-autonomous parvovirus based vectors, more preferably adeno-associated virus based vectors, most preferably AAV-2 based vectors).
  • adenoviruses preferably Ad-2 or Ad-5 based vectors
  • baculovirus preferably herpes viruses
  • parvoviruses preferably “defective” or non-autonomous parvovirus based vectors, more preferably adeno-associated virus based vectors, most preferably AAV-2 based vectors.
  • a particular vector system for transferring for instance, a KIM sequence will depend on a variety of factors.
  • One important factor is the nature of the target cell population.
  • retroviral vectors have been extensively studied and used in a number of gene therapy applications, they are generally unsuited for infecting cells that are not dividing but may be useful in cancer therapy since they only integrate and express their genes in replicating cells. They are useful for ex vivo approaches and are attractive in this regard due to their stable integration into the target cell genome.
  • Adenoviruses are eukaryotic DNA viruses that can be modified to efficiently deliver a therapeutic or reporter transgene to a variety of cell types.
  • the general adenoviruses types 2 and 5 Ad2 and Ad5, respectively
  • Ad2 and Ad5 are currently being developed for gene therapy of Duchenne Muscular Dystrophy (DMD) and Cystic Fibrosis (CF).
  • DMD Duchenne Muscular Dystrophy
  • CF Cystic Fibrosis
  • Both Ad2 and Ad5 belong to a subclass of adenovirus that are not associated with human malignancies.
  • Adenovirus vectors are capable of providing extremely high levels of transgene delivery to virtually all cell types, regardless of the mitotic state.
  • High titers (10 13 plaque forming units/ml) of recombinant virus can be easily generated in 293 cells (an adenovirus-transformed, complementation human embryonic kidney cell line: ATCC CRL1573) and cryo-stored for extended periods without appreciable losses.
  • the efficacy of this system in delivering a therapeutic transgene in vivo that complements a genetic imbalance has been demonstrated in animal models of various disorders. See Watanabe, Atherosclerosis, 36: 261-268, 1986; Tanzawa et al., FEBS Letters 118(1):81-84, 1980; Golasten et al., New Engl. J. Med. 309:288-296, 1983; Ishibashi et al., J. Clin.
  • recombinant replication defective adenovirus encoding a cDNA for the cystic fibrosis transmembrane regulator has been approved for use in at least two human CF clinical trials. See, e.g., Wilson, Nature 365:691-692, 1993. Further support of the safety of recombinant adenoviruses for gene therapy is the extensive experience of live adenovirus vaccines in human populations.
  • the first-generation recombinant, replication-deficient adenoviruses which have been developed for gene therapy of DMD and other inherited disorders contain deletions of the entire E1a and part of the E1b regions.
  • This replication-defective virus is grown in 293 cells containing a functional adenovirus E1a gene which provides a transacting E1a protein.
  • E1-deleted viruses are capable of replicating and producing infectious virus in the 293 cells, which provide E1a and E1b region gene products in trans.
  • the resulting virus is capable of infecting many cell types and can express the introduced gene (providing it carries its own promoter), but cannot replicate in a cell that does not carry the E1 region DNA unless the cell is infected at a very high multiplicity of infection.
  • Adenoviruses have the advantage that they have a broad host range, can infect quiescent or terminally differentiated cells such as neurons, and appear essentially non-oncogenic. Adenoviruses do not appear to integrate into the host genome. Because they exist extrachromasomally, the risk of insertional mutagenesis is greatly reduced. Ali et al., supra, at 373.
  • Recombinant adenoviruses produce very high titers, the viral particles are moderately stable, expression levels are high, and a wide range of cells can be infected. Their natural host cells are airway epithelium, so they are useful for therapy of lung cancers.
  • Baculovirus-mediated transfer has several advantages. Baculoviral gene transfer can occur in replicating and nonreplicating cells, and can occur in renal cells, as well as in hepatocytes, neural cells, spleen, skin, and muscle. Baculovirus is non-replicating and nonpathogenic in mammalian cells. Humans lack pre-existing antibodies to recombinant baculovirus which could block infection. In addition, baculovirus is capable of incorporating and transducing very large DNA inserts.
  • Adeno-associated viruses have also been employed as vectors for somatic gene therapy.
  • AAV is a small, single-stranded (ss) DNA virus with a simple genomic organization (4-7 kb) that makes it an ideal substrate for genetic engineering.
  • Two open reading frames encode a series of rep and cap polypeptides.
  • Rep polypeptides rep78, rep68, rep 62 and rep 40
  • the cap proteins form the virion capsid.
  • ITRs inverted terminal repeats
  • the entire rep and cap domains can be excised and replaced with a therapeutic or reporter transgene. See B. J. Carter, in Handbook of Parvoviruses, ed., P. Tijsser, CRC Press, pp. 155-168 (1990). It has been shown that the ITRs represent the minimal sequence required for replication, rescue, packaging, and integration of the AAV genome.
  • Adeno-associated viruses have significant potential in gene therapy.
  • the viral particles are very stable and recombinant AAVs (rAAV) have “drug-like” characteristics in that rAAV can be purified by pelleting or by CsCl gradient banding. They are heat stable and can be lyophilized to a powder and rehydrated to full activity.
  • rAAV recombinant AAVs
  • Their DNA stably integrates into host chromosomes so expression is long-term. Their host range is broad and AAV causes no known disease so that the recombinant vectors are non-toxic.
  • sequences of interest can be identified by conventional methods such as nucleic acid hybridization using probes comprising sequences that are homologous/complementary to the inserted gene sequences of the vector.
  • sequence(s) may be identified by the presence or absence of a “marker” gene function (e.g, thymidine kinase activity, antibiotic resistance, and the like) caused by introduction of the expression vector into the target cell.
  • a “marker” gene function e.g, thymidine kinase activity, antibiotic resistance, and the like
  • the compounds of the invention are formulated according to standard practice, such as prepared in a carrier vehicle.
  • a carrier vehicle includes sterile saline although other aqueous and non-aqueous isotonic sterile solutions and sterile suspensions known to be pharmaceutically acceptable are known to those of ordinary skill in the art.
  • carrier encompasses liposomes and the HIV-1 tat protein (See Chen et al., Anal. Biochem. 227: 168-175, 1995) as well as any plasmid and viral expression vectors.
  • any of the novel polypeptides of this invention may be used in the form of a pharmaceutically acceptable salt.
  • Suitable acids and bases which are capable of forming salts with the polypeptides of the present invention are well known to those of skill in the art, and include inorganic and organic acids and bases.
  • a compound of the invention is administered to a subject in a therapeutically-effective amount, which means an amount of the compound which produces a medically desirable result or exerts an influence on the particular condition being treated.
  • An effective amount of a compound of the invention is capable of ameliorating or delaying progression of the diseased, degenerative or damaged condition.
  • the effective amount can be determined on an individual basis and will be based, in part, on consideration of the physical attributes of the subject, symptoms to be treated and results sought. An effective amount can be determined by one of ordinary skill in the art employing such factors and using no more than routine experimentation.
  • a liposome delivery system for a compound of the invention may be any of a variety of unilamellar vesicles, multilamellar vesicles, or stable plurilamellar vesicles, and may be prepared and administered according to methods well known to those of skill in the art, for example in accordance with the teachings of U.S. Pat. Nos. 5,169,637, 4,762,915, 5,000,958 or 5,185,154.
  • treatment of human acute renal failure with liposome-encapsulated KIM protein may be performed in vivo by introducing a KIM protein into cells in need of such treatment using liposomes.
  • the liposomes can be delivered via catheter to the renal artery.
  • the recombinant KIM protein is purified, for example, from CHO cells by immunoaffinity chromatography or any other convenient method, then mixed with liposomes and incorporated into them at high efficiency.
  • the encapsulated protein may be tested in vitro for any effect on stimulating cell growth.
  • the compounds of the invention may be administered in any manner which is medically acceptable. This may include injections, by parenteral routes such as intravenous, intravascular, intraarterial, subcutaneous, intramuscular, intratumor, intraperitoneal, intraventricular, intraepidural, or others as well as oral, nasal, ophthalmic, rectal, or topical. Sustained release administration is also specifically included in the invention, by such means as depot injections or erodible implants. Localized delivery is particularly contemplated, by such means as delivery via a catheter to one or more arteries, such as the renal artery or a vessel supplying a localized tumor.

Abstract

Proteins which are upregulated in injured or regenerating tissues, as well as the DNA encoding these proteins, are disclosed, as well as therapeutic compositions and methods of treatment encompassing these compounds.

Description

    RELATED APPLICATIONS
  • This is a continuation-in-part of PCT/US97/09393, filed on May 23, 1997 as a continuation-in-part of U.S. Ser. No. 60/018,228, filed on May 24, 1996 and of U.S. Ser. No. 60/023,442, filed on Aug. 23, 1996. The entire disclosure of each of the aforesaid patent applications are incorporated herein by reference.
  • FIELD OF THE INVENTION
  • The invention relates to proteins which are upregulated in injured or regenerating tissues, as well as to the DNA encoding these proteins. The invention further relates to therapeutic compositions and methods of treatment encompassing these proteins.
  • BACKGROUND OF THE INVENTION
  • A dynamic remodeling of tissue architecture occurs during development and during tissue repair after injury. Herein are presented results of studies of this process, using a model of kidney injury caused by an ischemia-reperfusion insult.
  • The kidney is able to repair damage to the proximal tubule epithelium through a complex series of events involving cell death, proliferation of surviving proximal tubule epithelial cells, formation of poorly differentiated regenerative epithelium over the denuded basement membrane, and differentiation of the regenerative epithelium to form fully functional proximal tubule epithelial cells (Wallin et al., Lab. Invest. 66:474-484, 1992; Witzgall et al., Mol. Cell. Biol. 13:1933-1942, 1994; Ichimura et al., Am. J. Physiol. 269:F653-662, 1995; Thadhani et al., N. Engl. J. Med. 334:1448-1460, 1996). Growth factors such as IGF, EGF, and HGF have been implicated in this process of repair, as has the endothelial cell adhesion molecule ICAM-1. However, the mechanisms by which the tubular epithelial cells are restored are still not understood.
  • To identify molecules involved in processes of injury and repair of the tubular epithelium, differences were analyzed in the mRNA populations between injured/regenerating and normal kidneys using representational difference analysis (RDA). RDA is a PCR-based method for subtraction which yields target tissue or cell specific cDNA fragments by repetitive subtraction and amplification (Hubank and Schutz, Nucl. Acids Res. 22:5640-5648, 1994).
  • SUMMARY OF THE INVENTION
  • The invention generally provides Kidney Injury-related Molecules (each of which is henceforth called a “KIM”) which are upregulated in renal tissue after injury to the kidney. The KIM proteins and peptides of the invention, as well as their agonists and antagonists, and their corresponding nucleic acids are useful in a variety of therapeutic interventions.
  • The invention provides a purified and isolated DNA molecule having a nucleotide sequence set forth in SEQ ID NO:1, SEQ ID NO:2, SEQ ID NO:4 or SEQ ID NO:6. The invention also includes the complementary strands of these sequences, DNA molecules which hybridize under stringent conditions to the aforementioned DNA molecules, and DNA molecules which, but for the degeneracy of the genetic code, would hybridize to any of the DNA molecules defined above. These DNA molecules may be recombinant, and may be operably linked to an expression control sequence.
  • The invention further provides a vector comprising a purified and isolated DNA molecule having a nucleotide sequence set forth in SEQ ID NO:1, SEQ ID NO:2, SEQ ID NO:4 or SEQ ID NO:6, or one of the other DNA molecules defined above. This vector may be a biologically functional plasmid or viral DNA vector. One embodiment of the invention provides a prokaryotic or eukaryotic host cell stably transformed or transfected by a vector comprising a DNA molecule of SEQ ID NO:1, SEQ ID NO:2, SEQ ID NO:4 or SEQ ID NO:6. In another embodiment of the invention, a process is provided for the production of a KIM polypeptide product encoded by a DNA molecule as described above; the process involves growing, under suitable culture conditions, prokaryotic or eukaryotic host cells transformed or transfected with the DNA molecule in a manner allowing expression of the DNA molecule, and recovering the polypeptide product of said expression.
  • A purified and isolated human KIM protein substantially free of other human proteins is specifically within the invention, as is a process for the production of a polypeptide product having part or all of the primary structural conformation and the biological activity of a KIM protein. KIM proteins of the invention may have an amino acid sequence which comprises SEQ ID NO:3, SEQ ID NO:5, or SEQ ID NO:7, or may be a variant of SEQ ID NO:3, SEQ ID NO:5 or SEQ ID NO:7, or a purified and isolated protein encoded by the DNA of SEQ ID NO:1, SEQ ID NO:2, SEQ ID NO:4 or SEQ ID NO:6. These proteins can be provided substantially free of other human proteins. The invention further includes variants of these proteins, such as soluble variants or fusion proteins. KIM fusion proteins of the invention may comprise an immunoglobulin, a toxin, an imageable compound or a radionuclide.
  • The invention also provides a specific antibody, such as a monoclonal antibody (MAb), to the KIM proteins described above. The present antibody binds to any epitope that is unique to a KIM protein disclosed herein. In some embodiments, the epitope is displayed on the surface of a cell expressing KIM. Antigen-binding fragments of the antibody are also provided herein, preferably Fab, Fab2, Fab′ and Fv fragments, whether produced by chemical or enzymatic cleavage, or by molecular engineering techniques. Engineered versions of the present antibody include chimeric, humanized and human antibodies, and antibody fusion proteins. Monoclonal antibodies (MAbs) of the present invention can be of mouse, rat, hamster or human origin. An exemplary MAb of the invention is the murine AKG7 MAb disclosed herein, which binds specifically to the human KIM polypeptide disclosed herein. Anti-KIM antibodies of the present invention may be conjugated or fused to a therapeutic agent, toxin, imageable compound or radionuclide. Exemplary therapeutic agents include cytokines, lymphokines, trophic factors, survival factors, chemokines and chemoattractants. Exemplary toxins include ricin and diphtheria toxin. Exemplary imageable compounds include luminescent proteins (e.g., luciferin), fluorescent proteins (e.g., green fluorescent protein), haptens (e.g., biotin), and radioactively labeled proteins. Exemplary radionuclides include any radionuclide used for medical imaging purposes. The invention further encompasses all hybridoma cell lines and engineered host cells which produce antibodies of the invention.
  • Pharmaceutical compositions are also within the scope of the invention. A pharmaceutical composition of the invention may comprise a therapeutically effective amount of a KIM protein or anti-KIM antibody of the invention, along with a pharmacologically acceptable carrier.
  • Diagnostic methods are within the invention, such as assessing the presence or course of resolution of renal injury by measuring the concentration of KIM in urine, serum, or urine sediment of patients who have or who are at risk of developing renal disease. Other diagnostic methods that are within the invention include assessing KIM expression level in kidney tissue (e.g., in kidney biopsy tissue) of patients who have, are suspected of having, or are at risk of developing renal cancer (e.g., renal carcinoma). The present methods involve contacting an appropriate tissue or fluid sample derived from the patient being diagnosed, with a KIM antibody or a KIM probe (as the case may be), under binding conditions. Complexes formed by the binding of antibody or probe to KIM protein or nucleic acid (e.g., RNA) in the sample are detected by standard techniques. The presence or abnormal elevation of KIM protein in urine or serum is expected to correlate with renal failure or renal disease. The presence or abnormal elevation of KIM gene expression in renal cells or tissue is expected to correlate with disease processes, particularly carcinogenesis. Such correlations are expected to be useful in the prognostication, staging and clinical management of diseases or other conditions deleterious to renal tissue and/or renal function.
  • Methods of treatment of the invention include treating patients with therapeutically effective amounts of KIM, KIM variants, KIM analogs, KIM fusion proteins, KIM agonists, and antibodies to KIM or to KIM ligands. Other therapeutic compounds of the invention include KIM ligands, anti-KIM antibodies, and fusions proteins of KIM ligands. These compounds can be useful in therapeutic methods which either stimulate or inhibit cellular responses that are dependent on KIM function.
  • Further methods of the invention inhibit the growth of KIM-expressing tumor cells by contacting the cells with a fusion protein of a KIM ligand and either a toxin or radionuclide, or with an anti-KIM antibody conjugated to a toxin or to a radionuclide. Likewise, growth of tumor cells which express KIM ligand may be inhibited by contacting the cells with a fusion protein of a KIM and either a toxin or radionuclide, or with an anti-KIM ligand antibody conjugated to a toxin or to a radionuclide.
  • The invention also encompasses methods of gene therapy. These include a method of treating a subject with a renal disorder, a method of promoting growth of new tissue in a subject, and a method of promoting survival of damaged tissue in a subject, wherein each method comprises administering to the subject a vector which includes DNA comprising the nucleotide sequence of SEQ ID NO:1, SEQ ID NO:2, SEQ ID NO:4 or SEQ ID NO:6.
  • The compounds of the invention are also useful for imaging tissues, either in vitro or in vivo. One such method involves targeting an imageable compound to a cell expressing a protein of SEQ ID NO:3, SEQ ID NO:5 or SEQ ID NO:7, comprising contacting the cell with either a monoclonal antibody of the invention or a fusion protein comprising a protein as described above, fused to an imageable compound. For in vivo methods, the cell is within a subject, and the protein or the monoclonal antibody is administered to the subject.
  • The invention also includes diagnostic methods, such as a method of identifying damage or regeneration of renal cells in a subject, comprising comparing the level of expression of either SEQ ID NO:1,SEQ ID NO:2, SEQ ID NO:4 or SEQ ID NO:6 in renal cells of the subject to a control level of expression of the sequence in control renal cells. Another method of the invention includes identifying upregulation of SEQ ID NO:1, SEQ ID NO:2, SEQ ID NO:4 or SEQ ID NO:6 in cells comprising contacting the cells with an antisense probe and measuring hybridization to RNA within the cell.
  • A further embodiment of the diagnostic methods of the invention includes assessing the presence or concentration of a molecule of the invention either in urine, serum, or other body fluids, or in urine sediment or tissue samples. The measured injury-related molecule can be correlated with the presence, extent or course of a pathologic process. This correlation can also be used to assess the efficacy of a therapeutic regime.
  • BRIEF DESCRIPTION OF THE DRAWINGS
  • FIG. 1 is a text depiction of the nucleotide sequence of rat clone cDNA 3-2, with putative protein reading frame of 615 to 1535.
  • FIG. 2 is a text depiction of the cDNA sequence of rat clone 1-7, with putative protein reading frame of 145 to 1065.
  • FIG. 3 is a text depiction of the cDNA sequence of rat clone 4-7, with putative protein reading frame of 107 to 1822.
  • FIG. 4 is a text depiction of the cDNA and deduced amino acid sequences of human clone HI3-10-85, with putative protein reading frame of 1 to 1002. The upper line of the listing is the cDNA sequence (SEQ ID NO:6), and the lower line is the deduced amino acid sequence (SEQ ID NO:7).
  • FIG. 5 is a text depiction of a BESTFIT comparison of the nucleotide sequence of human clone HI3-10-85 with that of rat clone 3-2.
  • FIG. 6 is a digitized image of a Northern blot of human renal tissue samples isolated from renal carcinomas and from normal renal tissue samples.
  • DETAILED DESCRIPTION OF THE INVENTION
  • KIM genes were identified by analyzing differences in mRNA expression between regenerating and normal kidneys using representational difference analysis (RDA). RDA is a PCR-based method for subtraction which yields target tissue or cell-specific cDNA fragments by repetitive subtraction and amplification. The cDNA representation from 48 hr postischemic adult rat kidney RNA is subtracted with the sample from normal (sham-operated) adult rat kidney. In this procedure, sequences which are common to both postischemic and to normal kidney samples are removed, leaving those sequences which are significantly expressed only in the injured kidney tissue. Such genes encode proteins that may be therapeutically beneficial for renal disorders or involved in the injury process. Several clones have been obtained, sequenced and characterized. The clones are then investigated for their expression patterns during kidney repair, development and tissue distribution by northern analysis and RNA in situ hybridization.
  • Sequence Identification Numbers
  • Nucleotide and amino acid sequences referred to in the specification have been given the following sequence identification numbers:
    • SEQ ID NO:1—nucleotide sequence of rat 3-2 cDNA insert
    • SEQ ID NO:2—nucleotide sequence of rat 1-7 cDNA insert
    • SEQ ID NO:3—amino acid sequence of rat KIM-1, encoded by rat 3-2 and 1-7 cDNAs
    • SEQ ID NO:4—nucleotide sequence of rat 4-7 cDNA insert
    • SEQ ID NO:5—amino acid sequence encoded by 4-7 cDNA insert
    • SEQ ID NO:6—nucleotide sequence of human cDNA clone H13-10-85
    • SEQ ID NO:7—amino acid sequence encoded by human cDNA clone H13-10-85
    DEFINITIONS OF TERMS
  • A “KIM protein”, herein used synonymously with “KIM”, is a protein encoded by mRNA which is selectively upregulated following injury to a kidney. One group of KIM proteins of interest includes those coded for by mRNA which is selectively upregulated at any time within one week following any insult which results in injury to renal tissue. Examples of times at which such upregulation might be identified include 10 hours, 24 hours, 48 hours or 96 hours following an insult. Examples of types of insults include those resulting in ischemic, toxic or other types of injury.
  • A “KIM agonist” is a molecule which can specifically trigger a cellular response normally triggered by the interaction of KIM with a KIM ligand. A KIM agonist can be a KIM variant, or a specific antibody to KIM, or a soluble form of the KIM ligand.
  • A “KIM antagonist” is a molecule which can specifically associate with a KIM ligand or KIM, thereby blocking or otherwise inhibiting KIM binding to the KIM ligand. The antagonist binding blocks or inhibits cellular responses which would otherwise be triggered by ligation of the KIM ligand with KIM or with a KIM agonist. Examples of KIM antagonists include certain KIM variants, KIM fusion proteins and specific antibodies to a KIM ligand or KIM.
  • A “KIM ligand” is any molecule which noncovalently and specifically binds to a KIM protein. Such a ligand can be a protein, peptide, steroid, antibody, amino acid derivative, or other type molecule, in any form, including naturally-occurring, recombinantly produced, or otherwise synthetic. A KIM ligand can be in any form, including soluble, membrane-bound, or part of a fusion construct with immunoglobulin, fatty acid, or other moieties. The KIM ligand may be an integrin. A membrane-bound KIM ligand can act as a receptor which, when bound to or associated with KIM, triggers a cellular response. In some interactions, KIM may associate with more than a single KIM ligand, or may associate with a KIM ligand as part of a complex with one or more other molecules or cofactors. In a situation where both the KIM and the KIM ligand are bound to cell membranes, the KIM may associate and react with KIM ligand which is bound to the same cell as the KIM, or it may associate and react with KIM ligand be bound to a second cell. Where the KIM ligation occurs between molecules bound to different cells, the two cells may be the same or different with respect to cellular type or origin, phenotypic or metabolic condition, or type or degree of cellular response (e.g., growth, differentiation or apoptosis) to a given stimulus. “KIM ligation” refers to the contact and binding of KIM with a KIM ligand.
  • By “alignment of sequences” is meant the positioning of one sequence, either nucleotide or amino acid, with that of another, to allow a comparison of the sequence of relevant portions of one with that of the other. An example of one method of this procedure is given in Needleman et al. (J. Mol. Biol. 48:443-453, 1970). The method may be implemented conveniently by computer programs such as the Align program (DNAstar, Inc.). As will be understood by those skilled in the art, homologous or functionally equivalent sequences include functionally equivalent arrangements of the cysteine residues within the conserved cysteine skeleton, including amino acid insertions or deletions which alter the linear arrangement of these cysteines, but do not materially impair their relationship in the folded structure of the protein. Therefore, internal gaps and amino acid insertions in the candidate sequence are ignored for purposes of calculating the level of amino acid sequence similarity or identity between the candidate and reference sequences. One characteristic frequently used in establishing the similarity of proteins is the similarity of the number and location of the cysteine residues between one protein and another.
  • “Antisense DNA” refers to the sequence of chromosomal DNA that is transcribed.
  • An “antisense probe” is a probe which comprises at least a portion of the antisense DNA for a nucleic acid portion of interest.
  • By “cloning” is meant the use of in vitro recombination techniques to insert a particular gene or other DNA sequence into a vector molecule. In order to successfully clone a desired gene, it is necessary to employ methods for generating DNA fragments, for joining the fragments to vector molecules, for introducing the composite DNA molecule into a host cell in which it can replicate, and for selecting the clone having the target gene from amongst the recipient host cells.
  • By “cDNA” is meant complementary or copy DNA produced from an RNA template by the action of RNA-dependent DNA polymerase (reverse transcriptase). Thus a “cDNA clone” means a duplex DNA sequence complementary to an RNA molecule of interest, carried in a cloning vector.
  • By “cDNA library” is meant a collection of recombinant DNA molecules containing cDNA inserts which together comprise a representation of the mRNA molecules present in an entire organism or tissue, depending on the source of the RNA templates. Such a cDNA library may be prepared by methods known to those of skill, and described, for example, in Maniatis et al., Molecular Cloning: A Laboratory Manual, supra. Generally, RNA is first isolated from the cells of an organism from whose genome it is desired to clone a particular gene. Preferred for the purposes of the present invention are mammalian, and particularly human, cell lines. Alternatively, RNA may be isolated from a tumor cell, derived from an animal tumor, and preferably from a human tumor. Thus, a library may be prepared from, for example, a human adrenal tumor, but any tumor may be used.
  • As used herein, the term “DNA polymorphism” refers to the condition in which two or more different nucleotide sequences can exist at a particular site in DNA.
  • “Expression vector” includes vectors which are capable of expressing DNA sequences contained therein, i.e., the coding sequences are operably linked to other sequences capable of effecting their expression. It is implied, although not always explicitly stated, that these expression vectors must be replicable in the host organisms either as episomes or as an integral part of the chromosomal DNA. A useful, but not a necessary, element of an effective expression vector is a marker encoding sequence, which is a sequence encoding a protein which results in a phenotypic property (such as tetracycline resistance) of the cells containing the protein which permits those cells to be readily identified. In sum, “expression vector” is given a functional definition, and any DNA sequence which is capable of effecting expression of a specified contained DNA code is included in this term, as it is applied to the specified sequence. As at present, such vectors are frequently in the form of plasmids, thus “plasmid” and “expression vector” are often used interchangeably. However, the invention is intended to include such other forms of expression vectors which serve equivalent functions and which may, from time to time become known in the art.
  • By “functional derivative” is meant the “fragments”, “variants”, “analogs”, or “chemical derivatives” of a molecule. A “fragment” of a molecule, such as any of the antigens of the present invention is meant to refer to any polypeptide subset of the molecule. A “variant” of such molecules is meant to refer to a naturally occurring molecule substantially similar to either the entire molecule, or a fragment thereof. An “analog” of a molecule is meant to refer to a non-natural molecule substantially similar to either the entire molecule or a fragment thereof.
  • The term “gene” means a polynucleotide sequence encoding a peptide.
  • By “homogeneous” is meant, when referring to a peptide or DNA sequence, that the primary molecular structure (i.e., the sequence of amino acids or nucleotides) of substantially all molecules present in the composition under consideration is identical.
  • “Isolated” refers to a protein of the present invention, or any gene encoding any such protein, which is essentially free of other proteins or genes, respectively, or of other contaminants with which it might normally be found in nature, and as such exists in a form not found in nature.
  • The term “label” refers to a molecular moiety capable of detection including, by way of example, without limitation, radioactive isotopes, enzymes, luminescent agents, and dyes.
  • The term “probe” refers to a ligand of known qualities capable of selectively binding to a target antiligand. As applied to nucleic acids, the term “probe” refers to a strand of nucleic acid having a base sequence complementary to a target strand.
  • “Recombinant host cells” refers to cells which have been transformed with vectors constructed using recombinant DNA techniques. As defined herein, the antibody or modification thereof produced by a recombinant host cell is by virtue of this transformation, rather than in such lesser amounts, or more commonly, in such less than detectable amounts, as would be produced by the untransformed host.
  • By “substantially pure” is meant any protein of the present invention, or any gene encoding any such protein, which is essentially free of other proteins or genes, respectively, or of other contaminants with which it might normally be found in nature, and as such exists in a form not found in nature.
  • A molecule is said to be “substantially similar” to another molecule if the sequence of amino acids in both molecules is substantially the same, and if both molecules possess a similar biological activity. Thus, provided that two molecules possess a similar activity, they are considered variants as that term is used herein even if one of the molecules contains additional amino acid residues not found in the other, or if the sequence of amino acid residues is not identical. As used herein, a molecule is said to be a “chemical derivative” of another molecule when it contains additional chemical moieties not normally a part of the molecule. Such moieties may improve the molecule's solubility, absorption, biological half life, etc. The moieties may alternatively decrease the toxicity of the molecule, eliminate or attenuate any undesirable side effect of the molecule, etc. Moieties capable of mediating such effects are disclosed, for example, in Remington's Pharmaceutical Sciences. 16th ed., Mack Publishing Co., Easton, Pa. (1980).
  • By “vector” is meant a DNA molecule, derived from a plasmid or bacteriophage, into which fragments of DNA may be inserted or cloned. A vector will contain one or more unique restriction sites, and may be capable of autonomous replication in a defined host or vehicle organism such that the cloned sequence is reproducible.
  • COMPOUNDS OF THE INVENTION
  • The invention includes the cDNA of SEQ ID NO: 1, SEQ ID NO:2, SEQ ID NO:4 or SEQ ID NO:6, as well as sequences which include the sequence of SEQ ID NO: 1, SEQ ID NO:2, SEQ ID NO:4, or SEQ ID NO:6, and derivatives of these sequences. The invention also includes vectors, liposomes and other carrier vehicles which encompass these sequence or derivatives of them. The invention further includes proteins transcribed from SEQ ID NO:1, SEQ ID NO:2, SEQ ID NO:4, or SEQ ID NO:6, including but not limited to SEQ ID NO:3, SEQ ID NO:5 or SEQ ID NO:7, and their derivatives and variants.
  • One embodiment of the invention includes soluble variants of a KIM protein that is usually synthesized as a membrane associated protein, and which is upregulated after injury. Soluble variants lack at least a portion of the transmembrane or intra-membrane section of a native KIM protein. In some examples, the soluble variant lacks the entire transmembrane or intra-membrane section of a native KIM protein. The present soluble variants can be derived from intact or native KIM proteins by any suitable means, including chemical or enzymatic cleavage, or through molecular engineering techniques such by expression of a truncated KIM nucleic acid. Thus, the present soluble variants should be understood to be derivatives of an intact KIM. Soluble variants include fusion proteins which encompass derivatives of KIM proteins that lack at least a portion of the transmembrane or intra-membrane section of a native KIM protein. All types of KIM fusion proteins are included, particularly those which incorporate his-tag, Ig-tag, and myc-tag forms of the molecule. These KIM fusions may have characteristics which are therapeutically advantageous, such as the increased half-life conferred by the Ig-tag. Also included are fusion proteins which incorporate portions of selected domains of the KIM protein.
  • Variants can differ from naturally occurring KIM protein in amino acid sequence or in ways that do not involve sequence, or both. Variants in amino acid sequence are produced when one or more amino acids in naturally occurring KIM protein is substituted with a different natural amino acid, an amino acid derivative or non-native amino acid. Particularly preferred variants include naturally occurring KIM protein, or biologically active fragments of naturally occurring KIM protein, whose sequences differ from the wild type sequence by one or more conservative amino acid substitutions, which typically have minimal influence on the secondary structure and hydrophobic nature of the protein or peptide. Variants may also have sequences which differ by one or more non-conservative amino acid substitutions, deletions or insertions which do not abolish the KIM protein biological activity. Conservative substitutions typically include the substitution of one amino acid for another with similar characteristics such as substitutions within the following groups: valine, glycine; glycine, alanine; valine, isoleucine; aspartic acid, glutamic acid; asparagine, glutamine; serine, threonine; lysine, arginine; and phenylalanine, tyrosine. The non-polar (hydrophobic) amino acids include alanine, leucine, isoleucine, valine, proline, phenylalanine, tryptophan and methionine. The polar neutral amino acids include glycine, serine, threonine, cysteine, tyrosine, asparagine and glutamine. The positively charged (basic) amino acids include arginine, lysine and histidine. The negatively charged (acidic) amino acids include aspartic acid and glutamic acid.
  • Other conservative substitutions can be taken from the table below, and yet others are those which meet the criteria for an accepted point mutation in Dayhoff et al. (1978), 5 Atlas of Protein Sequence and Structure, Suppl. 3, ch. 22, Natl. Biomed. Res. Found., Washington, D.C. 20007, the teachings of which are incorporated by reference herein.
    TABLE 1
    CONSERVATIVE AMINO ACID REPLACEMENTS
    For Amino Acid Code Replace with any of
    Alanine A D-Ala, Gly, beta-Ala, L-Cys, D-Cys
    Arginine R D-Arg, Lys, homo-Arg, D-homo-Arg, Met, D-Met, Ile, D-Ile,
    Orn, D-Orn
    Asparagine N D-Asn, Asp, D-Asp, Glu, D-Glu, Gln, D-Gln
    Aspartic Acid D D-Asp, D-Asn, Asn, Glu, D-Glu, Gln, D-Gln
    Cysteine C D-Cys, S—Me-Cys, Met, D-Met, Thr, D-Thr
    Glutamine Q D-Gln, Asn, D-Asn, Glu, D-Glu, Asp, D-Asp
    Glutamic Acid E D-Glu, D-Asp, Asp, Asn, D-Asn, Gln, D-Gln
    Glycine G Ala, D-Ala, Pro, D-Pro, Beta-Ala, Acp
    Isoleucine I D-Ile, Val, D-Val, Leu, D-Leu, Met, D-Met
    Leucine L D-Leu, Val, D-Val, Met, D-Met
    Lysine K D-Lys, Arg, D-Arg, homo-Arg, D-homo-Arg, Met, D-Met, Ile,
    D-Ile, Orn, D-Orn
    Methionine M D-Met, S—Me-Cys, Ile, D-Ile, Leu, D-Leu, Val, D-Val, Norleu
    Phenylalanine F D-Phe, Tyr, D-Thr, L-Dopa, His, D-His, Trp, D-Trp, Trans 3,4
    or 5-phenylproline, cis 3,4 or 5 phenylproline
    Proline P D-Pro, L-I-thioazolidine-4-carboxylic acid, D- or L-1-
    oxazolidine-4-carboxylic acid
    Serine S D-Ser, Thr, D-Thr, allo-Thr, Met, D-Met, Met(O), D-Met(O),
    Val, D-Val
    Threonine T D-Thr, Ser, D-Ser, allo-Thr, Met, D-Met, Met)O, D-Met(O),
    Val, D-Val
    Tyrosine Y D-Tyr, Phe, D-Phe, L-Dopa, His, D-His
    Valine V D-Val, Leu, D-Leu, Ile, D-Ile, Met, D-Met
  • Other variants within the invention are those with modifications which increase peptide stability. Such variants may contain, for example, one or more non-peptide bonds (which replace the peptide bonds) in the peptide sequence. Also included are: variants that include residues other than naturally occurring L-amino acids, such as D-amino acids or non-naturally occurring or synthetic amino acids such as beta or gamma amino acids and cyclic variants. Incorporation of D- instead of L-amino acids into the polypeptide may increase its resistance to proteases. See, e.g., U.S. Pat. No. 5,219,990.
  • Generally, substitutions that may be expected to induce changes in the functional properties of KIM polypeptides are those in which: (I) a hydrophilic residue, e.g., serine or threonine, is substituted by a hydrophobic residue, e.g., leucine, isoleucine, phenylalanine, or alanine; (ii) a cysteine residue is substituted for (or by) any other residue; (iii) a residue having an electropositive side chain, e.g., lysine, arginine or histidine, is substituted for (or by) a residue having an electronegative charge, e.g., glutamic acid or aspartic acid; or (iv) a residue having a bulky side chain, e.g., phenylalanine, is substituted for (or by) one not having such a side chain, e.g., glycine.
  • The peptides of this invention may also be modified by various changes such as insertions, deletions and substitutions, either conservative or nonconservative where such changes might provide for certain advantages in their use. Splice variants are specifically included in the invention.
  • In other embodiments, variants with amino acid substitutions which are less conservative may also result in desired derivatives, e.g., by causing changes in charge, conformation and other biological properties. Such substitutions would include for example, substitution of hydrophilic residue for a hydrophobic residue, substitution of a cysteine or proline for another residue, substitution of a residue having a small side chain for a residue having a bulky side chain or substitution of a residue having a net positive charge for a residue having a net negative charge. When the result of a given substitution cannot be predicted with certainty, the derivatives may be readily assayed according to the methods disclosed herein to determine the presence or absence of the desired characteristics.
  • In accordance with the foregoing, the scope of the invention includes proteins and peptides with variant amino acid sequences (sequence variants) having at least eighty percent similarity with a KIM protein. More preferably the sequence similarity is at least ninety percent, or at least ninety-five percent. For example, a sequence variant within the scope of the invention is any which shares at least eighty percent similarity with a KIM selected from rat KIM1 (SEQ ID NO:3) and human KIM1 (SEQ ID NO: 7). The percentage of similarity between a variant sequence and a reference sequence (such as SEQ ID NO:3 or SEQ ID NO:7) is determined after the variant is aligned with the reference, preferably according to the method of Needelman et al. (1970), 48 J. Mol. Biol. 443-454, or according to an appropriate sequence alignment program (discussed more fully below). The percentage of sequence “identity” of the variant to the reference is the percentage of amino acid or nucleic acid residues of the variant sequence that are identical to the aligned (corresponding) residues of the reference. The percentage of sequence “similarity” is the sum of the percentage of identical corresponding residues and the percentage of variant residues that are conservative substitutions for the aligned (corresponding) reference residues.
  • A number of algorithms and computer software programs can be utilized to determine whether a sequence of interest is a KIM sequence variant within the scope of this invention. One such is the algorithm of Karlin and Altschul (1990) Proc. Natl. Acad. Sci. USA 87:2264-68, as modified in Karlin and Altschul (1993), Proc. Natl. Acad. Sci. USA 90:5873-77. This algorithm is incorporated into the NBLAST and XBLAST programs of Altschul et al. (1990) J. Mol. Biol. 215:403-10. BLAST nucleotide searches can be performed with NBLAST. BLAST protein searches can be performed with XBLAST. To obtain gapped alignments, gapped BLAST can be utilized as described in Altschul et al. (1997) Nucl. Acids Res. 25(17):3389-3402. All of the foregoing are available through http:\\www.ncbi.nlm.nih.gov. Another suitable algorithm is that of Myers and Miller, CABIOS (1989), which is incorporated into the ALIGN program (version 2.0), which is a part of the Wisconsin Sequence Analysis package of the Genetics Computing Group (GCG). Thus, variants within the invention include any polypeptide or nucleic acid sequence which is identified through BLAST or CABIOS analysis as being related to a KIM nucleic acid or polypeptide sequence disclosed herein (e.g., rat KIM1, SEQ ID NO:3 or human KIM1, SEQ ID NO:7). “Related” sequences are those for which, in the judgement of an ordinarily skilled practitioner, the results of sequence analysis indicate a biologically significant relationship with the reference sequence (e.g., rat KIM1 or human KIM1).
  • Preferably, sequence variants of the KIM proteins disclosed herein are those having an amino acid sequence which is at least forty (40) percent identical overall (i.e., when aligned) to a disclosed KIM polypeptide sequence, and sharing at least 80% of the aligned KIM cysteine residues. Particularly preferred sequence variants are those which meet the foregoing analysis criteria when analyzed, relative to a KIM reference sequence disclosed herein (e.g., rat KIM1, SEQ ID NO:3 or human KIM1, SEQ ID NO:7), using the program GAP, which is a part of the Wisconsin analysis package of the GCG. To run a GAP analysis, the following program parameters should be used: the gap creation penalty (gap weight) should be set to 3.0, and the gap extension penalty (gap length) should be set to 0.1. GAP uses the algorithm of Needelman et al. (1970), 48 J. Mol. Biol. 443-453, to create an alignment with the largest number of matched (aligned identical) residues and the fewest number of sequence gaps.
  • Just as it is possible to replace substituents of the scaffold, it is also possible to substitute functional groups which are bound to the scaffold with groups characterized by similar features. These substitutions will initially be conservative, i.e., the replacement group will have approximately the same size, shape, hydrophobicity and charge as the original group. Non-sequence modifications may include, for example, in vivo or in vitro chemical derivatization of portions of naturally occurring KIM protein, as well as changes in acetylation, methylation, phosphorylation, carboxylation or glycosylation.
  • Also included within the invention are agents which specifically bind to the protein, or a fragment of the protein (SEQ ID NO:3, 5 or 7). These agents include ligands and antibodies (including monoclonal, single chain, double chain, Fab fragments, and others, whether native, human, humanized, primatized, or chimeric). Additional descriptions of these categories of agents are in PCT application 95/16709, the specification of which is herein incorporated by reference.
  • Experimental Procedures
  • 1. Generation of RNA from Ischemic and Normal Rat Adult Kidneys
  • Ischemic injured rat kidneys are generated as described by Witzgall et al. (J. Clin Invest. 93: 2175-2188, 1994). Briefly, the renal artery and vein from one kidney of an adult Sprague-Dawley rat are clamped for 40 minutes and then reperfused. Injured kidneys are harvested from the rats at 24 hours and at 48 hours after reperfusion. Kidneys from sham-operated, normal adult Sprague-Dawley rats are also harvested.
  • Total RNA is prepared from the organs based on the protocol by Glisin et al. (Biochemistry 13: 2633, 1974). Briefly, the harvested organs are placed immediately into GNC buffer (4M guanidine thiocyanate, 0.5% SDS, 25 mM sodium citrate, 0.1% Sigma anti foam) and disrupted. on ice with a polytron. Cell debris is removed with a low speed spin in a clinical centrifuge and the supernatant fluid is placed on a 5.7 M CsCl, 25 mM sodium acetate, 1 mM EDTA cushion. RNA is pelleted through the cushion in a SW40Ti rotor at 22K for 15 hrs. RNA is resuspended in sterile DEPC-treated water, precipitated twice with 1/10 volume 3M sodium acetate and 2.5 volumes of EtOH. Poly A+ RNA is isolated using an mRNA purification kit (Pharmacia, catalog No.27-9258-02).
  • 2. Representational Difference Analysis (RDA) Method to Isolate 1-7, 3-2 and 4-7 RDA Fragments
  • Double stranded cDNA is synthesized from sham-operated and from 48 hr post-ischemic kidney poly A+ RNA using Gibco BRL “Superscript Choice™ System cDNA Synthesis Kit”, catalog No. 18090. First strand is synthesized by priming with oligo dT and using Superscript II™ reverse transcriptase. Second strand is generated using E. coli DNA polymerase I and RNase H followed by T4 DNA polymerase using BRL recommended conditions.
  • RDA analysis is performed essentially as described by Hubank and Schatz (Nucleic Acid Research 22: 5640-48, 1994). Briefly, 48 hr post-ischemic kidney cDNA is digested with the restriction enzyme Dpn II, and ligated to R-Bgl-12/24 oligonucleotides (see reference for exact sequence). PCR amplification (performed with Perkin-Elmer Taq polymerase and their corresponding PCR buffer) of the linker ligated cDNA is used to generate the initial representation. This PCR product is designated “tester amplicon.” The same procedure is used to generate “driver amplicon” from sham-operated rat kidney cDNA.
  • Hybridization of tester and driver amplicons followed by selective amplification are performed three times to generate Differential Product One (DP1), Two (DP2) and Three (DP3). Generation of the DP1 product is performed as described by Hubank and Schatz (Nucleic Acid Research 22: 5640-48, 1994). The DP2 and DP3 products are also generated as described by Hubank and Schatz (id.), except that the driver:tester ratios are changed to 5,333:1 for DP2 and to 40,000:1 or 4,000:1 for DP3.
  • Three RDA products are cloned from DP3 into the cloning vector pUC 18: RDA product 1-7 (252 bp) when the DP3 was generated using a ratio of 40,000:1, and product RDA 3-2 (445 bp) and 4-7 (483 bp) when the DP3 was generated using a ratio of 4,000:1. The DNA fragments are subcloned using the Pharmacia Sureclone™ kit (catalog No. 27-9300-01) to repair the ends of the PCR fragments with Klenow enzyme and to facilitate blunt end ligation of the fragments into the pUC18 vector.
  • 3. Northern Analysis
  • Poly A+ RNA (2.5 μg) from rat normal adult kidney (sham operated), from 48 hr post-ischemic injured adult kidney, and from day 18 embryonic kidney is electrophoresed and Northern blotted (Cate, Cell 45:685, 1986) to a GeneScreen™ membrane (Dupont). Hybridization in PSB buffer (50 mM Tris 7.5, 1M NaCl, 0.1% Na pyrophosphate, 0.2% PVP, 0.2% Ficoll, 0.2% BSA, 1% SDS), containing 10% dextran sulphate and 100 μg/ml tRNA, is performed at 65 C using three different probes: 1-7 RDA product, 3-2 RDA product and 4-7 RDA product. All are radiolabeled using Pharmacia's “Ready to Go™” random priming labeling kit (catalog No. 27-9251-01). RDA products 1-7, 3-2 and 4-7 hybridize to mRNAs present in all three samples, but most intensely to mRNAs in the 48 hr post-ischemic kidney RNA samples.
  • A Northern blot analysis of adult rat tissues indicates that the 1-7 gene is expressed at very low levels in normal adult kidney, testis, spleen and lung. The 3-2 gene is expressed in liver, kidney, spleen, and brain. The 4-7 gene is expressed in spleen, kidney, lung, testis, heart, brain, liver, and skeletal muscle. The presence of different sized mRNAs in some tissues in the 1-7 and 3-2 blot indicates that the primary transcription product of the 1-7 gene and of the 3-2 gene may undergo alternate splicing and/or polyadenylation.
  • 4. Isolation of 3-2 and 4-7 cDNA Clones
  • A cDNA library is generated from 4 μg of polyA+ RNA from 48 hr post-ischemic injured kidney using reagents from BRL Superscript Choice™ System for cDNA synthesis, and Stratagene™ Lambda ZapII cloning-kit (catalog No. 236201), according to protocols recommended by the manufacturers.
  • 105 clones are screened with the 3-2 RDA product as a probe (random primed labeled as described above). Eight positive clones are selected and four are randomly chosen for secondary analysis to obtain pure phage plaques. After tertiary screening, four pure phage clones are isolated. Cloned inserts from the phage are isolated by in vivo excision procedure according to Stratagene™ Lambda Zap II kit. The largest insert, of approximately 2.6 kb (referred to as cDNA clone 3-2), is subjected to DNA sequencing. The sequence of the insert (SEQ ID NO:1) is shown in FIG. 1. cDNA clone 3-2 (E. coli K-12, SOLR/p3-2#5-1) has been deposited as ATCC No. 98061. The sequence of cDNA clone 3-2 is identical to that of clone 1-7 cDNA (SEQ ID NO: 2), except that nucleotides 136-605 of SEQ ID NO:1 represent an insertion. Thus, SEQ ID NO:2 represents a splice variant form of SEQ ID NO: 1. The clone for 1-7 (E. coli K-12, SOLR/p1-7#3-1) has been deposited as ATCC No. 98060.
  • 105 clones are screened with the 1-7 RDA product as a probe (random primed radiolabeled as described above). Eight positive clones are selected and four are randomly chosen for secondary analysis to obtain pure phage plaques. After tertiary screening, four pure phage clones are isolated. Cloned inserts from the phage are isolated by in vivo excision procedure according to Stratagene™ Lambda Zap II kit. The largest insert of approximately 2.0 kb (referred to as cDNA clone 1-7) is subjected to DNA sequencing; the sequence of the insert (SEQ ID NO: 2) is shown in FIG. 2.
  • 105 clones are screened with the 4-7 RDA product as a probe (random primed labeled as described above and hybridized in PSB at 65 C). Eight positive clones are selected and four are randomly chosen for secondary analysis to obtain pure phage plaques. After secondary screening, two pure phage clones are isolated. Cloned inserts from the phage are isolated by in vivo excision procedure according to Stratagene™ Lambda Zap II kit. The largest insert, approximately 2.4 kb (referred to as cDNA clone 4-7), is subjected to DNA sequencing. The sequence of the insert, SEQ ID NO: 4, is shown in FIG. 3. The cDNA clone 4-7 (E. coli K-12, SOLR/p4-7#1-1) has been deposited as ATCC No. 98062.
  • 5. Characterization of the 1-7, 3-2 and 4-7 cDNA Clones
  • A.) DNA and Protein Sequences:
  • The sequence of 3-2 cDNA (FIG. 1; SEQ ID NO:1) contains an open reading frame of 307 amino acids (FIG. 1; SEQ ID NO:3). A signal sequence of 21 amino acids is inferred from Von Heijne analysis (Von Heijne et al., Nucl. Acid Res. 14:14683 (1986)), and a transmembrane region spanning approximately aa 235-257 indicates that the 3-2 product is a cell surface protein. The sequence of 1-7 cDNA (FIG. 2; SEQ ID NO:2) contains an open reading frame of 307 amino acids, which is identical to the open reading frame contained in the 3-2 cDNA (SEQ ID NO: 3). This 307 amino acid polypeptide is herein designated rat KIM1. The sequence of 4-7 cDNA (FIG. 3; SEQ ID NO:4) contains an open reading frame of 572 amino acids (SEQ ID NO:5). A transmembrane region is located at approximately amino acids 501-521. This 572 amino acid polypeptide is related to the polypeptide known as nmb. Weterman et al. (1995) 60 Int. J. Cancer 73-81.
  • B.) In situ Analysis of 1-7, 3-2 and 4-7 mRNAs in Contralateral and in Post-Ischemic Adult Rat Kidneys:
  • In situ hybridization is carried out according to the method described by Finch et al., Dev. Dynamics 203: 223-240, 1995. Briefly, both ischemic and contralateral kidneys are perfusion fixed with 4% paraformaldehyde in PBS. Kidneys are further fixed overnight at 4 C and processed. Paraffin sections are deparaffinized and rehydrated, fixed with 4% paraformaldehyde in PBS, digested with proteinase K, refixed, then acetylated with acetic anhydride in triethanolamine buffer. Sections are then dehydrated and hybridized with 32P-labeled riboprobes at 55° C. overnight, with 33P-labeled riboprobes generated from 3-2 RDA or 1-7 RDA products subcloned into BamH1 site of pGEM-11Z. After hybridization, sections were washed under high stringency conditions (2×SSC, 50% formamide at 65° C.). Sections are finally dehydrated, emulsion (NBT-2) coated for autoradiography, and exposed for at least a week. Silver grains are developed and sections are counterstained with toluidine blue and microphotographed.
  • Analysis of 1-7 and 3-2 mRNA expression by in situ hybridization indicates that these genes are greatly upregulated in damaged kidney cells compared to their expression in normal kidney sections. The expression seen is in regenerative cells of the cortex and outer medulla, most of which appear to be proximal tubule cells.
  • Analysis of the 4-7 in situ RNA expression pattern also reveals abundant expression of this gene in the injured ischemic kidney compared to the normal adult kidney. The site of expression appears to be infiltrating cells.
  • 6.) Isolation of a Human cDNA Clone Which Cross Hybridizes to the Rat 3-2 cDNA
  • A 32P-labeled DNA probe comprising nucleotides 546-969 of the insert of clone 3-2 shown in FIG. 1 is generated and used to screen a human embryonic liver lambda gt10 cDNA library (Clontech Catalog #HL5003a). 1×106 plaques are screened in duplicate using standard conditions as described above but temperature for screening was 55 C. For the high stringency wash, the filters are washed in 2×SSC at 55 C. Fifty positive phage are identified and plaque purified, and DNA is prepared. The phage DNAs are subjected to Southern analysis using the same probe as above. The Southern blot filter is subjected to a final wash with 0.5×SSC at 55 C. Two clones are identified as positive. The insert of clone H13-10-85 is sequenced and a region is found that encodes a protein with a high level of identity to the 3-2 protein shown in FIG. 3.
  • The nucleotide sequence (SEQ ID NO:6) and predicted amino acid sequence (SEQ ID NO:7) of the human 3-2 related protein are shown in FIG. 4. As shown by the bestfit analysis depicted in FIG. 5, the human 3-2 related protein is 43.8% identical and 59.1% similar to the rat 3-2 protein. Both contain IgG, mucin, transmembrane, and cytoplasmic domains. The six cysteines within the IgG domains of both proteins are conserved. Accordingly, the polypeptide of SEQ D NO:7 is designated human KIM1 (or KIM-1) herein. It is believed that the mammalian genome (e.g., the human genome) includes genes encoding a family of KIM-1 related proteins. Thus, the human KIM-1 disclosed herein is more precisely designated human KIM-1a.
  • 7) Production of KIM-1 Ig Fusion Protein
  • A fusion protein of the extracellular, domain of KIM and the Fc region of immunoglobulin (Ig) is a useful tool for the study of the molecular and cellular biology of the injured/regenerating kidney and as a therapeutic molecule. To produce Kim Ig fusion protein with the extracellular domain of human and rat KIM-1 protein, a fragment of the extracellular domain of KIM-1 cDNA was amplified by PCR and cloned in the Biogen expression vector, pCA125, for transient expression in COS cells. The expression vector pCA125 produces a fusion protein which has a structure from gene cloned at N-terminus and a human Ig Fc region at the C-terminus. COS cells were transfected with the plasmids SJR 103 or 104; these plasmids express a fusion protein which contains the human KIM sequences 263-1147 (SEQ ID NO:6; SJR 103) or rat KIM sequences 599-1319 (SEQ ID NO:1; SJR 104) of the extracellular domain fused to human Ig Fc region. The cells were grown in 10% FBS in DMEM in the cell factory (Nunc, Naperville, Ill.). Two to three days post-transfection, medium was harvested, concentrated using Amicon concentrator, and fusion protein was purified using Protein-A Sepharose column. After purification, purity of fusion protein was evaluated by SDS-PAGE.
  • 8) Production of Antibodies, Including Monoclonal Antibody (MAb) AKG7, with Binding Specificity for Human KIM Protein
  • Antibody production involves administration of one or more immunogenic doses of a KIM polypeptide preparation (whether isolated, or as part of a cell suspension, fraction or extract) to an appropriate non-human animal, such as a mouse, rat, rabbit, gunea pig, turkey, goat, sheep, pig or horse. To enhance immunogenicity, the preparation is emulsified with a conventional adjuvant (e.g., Complete Freund's Adjuvant, CFA). Serum immunoglobulins (Ig), using peripheral blood samples withdrawn at intervals (e.g., weekly) after initial or a subsequent immunization, are monitored to detect the onset and/or maturation of a humoral immune response to KIM. Any conventional technique, e.g., ELISA, RIA, Western blotting, or the like, can be used to detect and/or quantitate (titer) the KIM-specific Ig. High titer immune sera are preferred for use as anti-KIM polyclonal antibodies herein. High titer sera (e.g., having a titer of at least 1:1000) can be harvested in bulk for purification, of anti-KIM Ig, using-conventional immunoaffinity chromatography (e.g., using a Protein A resin).
  • Animals (e.g., mice, rats, guinea pigs, hamsters) having a high serum titer of anti-KIM Ig also can be used for the conventional production of monoclonal antibodies (MAb) with binding specificity for KIM. Splenocytes are isolated from such animals and are fused with a myeloma cell line according to standard techniques to produce hybridomas. Such hybridomas are cloned by limiting dilution, fluorescence-activated cell sorting, or any other suitable cell isolation technique, to produce monoclonal hybridoma cell lines of the present invention. Hybridoma lines of the present invention are useful for the production of anti-KIM MAbs, and also as sources from which nucleic acids encoding the present anti-KIM MAbs can be routinely isolated. Such nucleic acids, encoding heavy and/or light chain polypeptides of the present MAbs, can be used to produce recombinant, engineered versions of the present MAbs, including truncated MAb polypeptides, chimeric or humanized MAbs, and MAb fusion proteins.
  • By way of illustration, mice have been immunized with a human KIM1-Ig fusion protein as disclosed herein, which was immobilized on Protein A Sepharose beads and emulsified in CFA. The fusion protein included the extracellular domain of human KIM1 (amino acids 1 to 291) fused to a truncated human IgG1 polypeptide comprising hinge-C2-C3 domains of the IgG1 heavy chain polypeptide. The onset and maturation of a humoral immune response was monitored using a conventional ELISA, in which another Ig fusion protein was used as a blocking agent to subtract serum Ig's having undesired specificity for the IgG1 portion of the KIM1 fusion protein. Following conventional isolation of immune splenocytes, fusion, ELISA screening, and cloning by limiting dilution, ten MAbs were obtained with desirable binding specificity for human KIM1. Among these, one, designated AKG7, is preferred herein. The AKG7 antibody is useful in ELISA, Western blot and immunohistochemistry analysis of human KIM1 polypeptides. The present technique can be repeated and/or adapted routinely to produce any number of MAbs to domains (e.g., the extracellular domain) of KIM proteins, that have the KIM-specific binding properties of MAb AKG7 and are useful in the analytical, diagnostic and therapeutic methods described herein.
  • MAbs of the present invention, such as AKG7, are obtained from cell culture media conditioned by the present hybridoma lines, or from ascites fluid, or from media conditioned by an engineered host cell expressing a heavy and/or light chain nucleic acid construct encoding an anti-KIM MAb of the present invention. Such MAbs are purified using conventional immunoaffinity chromatography, and used in the various diagnostic, analytical and therapeutic methods described herein.
  • DIAGNOSTIC USES OF THE COMPOUNDS OF THE INVENTION
  • Anti-KIM antibodies of the invention, which specifically bind to the protein of SEQ ID NO:3, SEQ ID NO:5 or SEQ D NO:7 or a fragment thereof, are useful in several diagnostic methods. These agents may be labeled with detectable markers, such as fluoroscopically or radiographically opaque substances, and administered to a subject to allow imaging of tissues which express KIM protein. The agents may also be bound to substances, such as horseradish peroxidase, which can be used as immunocytochemical stains to allow visualization of areas of KIM protein-positive cells on histological sections. A specific antibody could be used alone in this manner, and sites where it is bound can be visualized in a sandwich assay using an anti-immunoglobulin antibody which is itself bound to a detectable marker.
  • For example and not by way of limitation, the AKG7 MAb can be used to visualize cells producing KIM-1 protein in immunohistochemistry analysis of human renal tissue samples. Immunohistochemistry is carried out on paraformaldehyde-fixed paraffin sections by deparaffinizing the sections, then ablating endogenous peroxidase activity by incubation in 2% hydrogen peroxide in methanol for 20 min. Sections are then heated in a microwave oven in 0.1 M citrate buffer, pH 6.0, for 10 min., then blocked with diluted goat serum (1:67) overnight at 4° C. Thereafter, sections are incubated with affinity purified AKG7 or control IgG at a concentration of 5 μg/mL. After 1 hr, the sections are washed in PBS and incubated with biotinylated goat anti-mouse IgG for 30 min. After further washes with PBS, the sections are incubated with avidin-biotinylated horseradish peroxidase complex (Vectastain Elite ABC kit, Vector Laboratories) for 1 hr. Sections are washed in PBS and developed by the addition of 50 mM sodium phosphate buffer, pH 7.6, containing 0.12% 3,3-diaminobenzidine tetrahydrochloride (Sigma), 0.0075% nickel chloride, 0.0075% cobalt chloride, and 0.0075% hydrogen peroxide for 2-5 min., producing a dark brown stain indicating immunoreactivity. Sections are then counterstained with 0.01% toluidine blue. This technique has been used to visualize KIM-1 immunoreactivity in paraffin sections from two different human patients with renal cell carcinoma. Sections comprising tumor tissue and adjacent normal renal tissue were assessed. In patients, strong KIM-1 immunoreactivity was noted in tumor tissue. By contract, immunoreactivity was virtually absent in adjacent normal tissue (as judged by pathology). Immunoreactivity was also virtually absent in normal control renal tissue sections. These data are consistent with the utility of antibodies to KIM-1 as reagents for the diagnosis, prognostication and/or staging of renal cancers.
  • Specific antibodies to KIM protein are also useful in immunoassays to measure KIM presence or concentration in samples of body tissues and fluids. Such concentrations may be correlated with different disease states. As an embodiment of particular interest, the invention includes a method of diagnosing renal injury, or of monitoring a process of renal repair, by measuring the concentration of KIM or of KIM fragments in the urine, plasma or serum of a patient. Similarly, KIM can be measured in urine sediment, in particular in cellular debris in the urine sediment. Casts of renal tubule cells, which may be present in urine sediment from patients with ongoing renal disease, may contain elevated levels of KIM protein and mRNA.
  • Specific antibodies to KIM protein may also be bound to solid supports, such as beads or dishes, and used to remove the ligand from a solution, either for measurement, or for purification and characterization of the protein or its attributes (such as posttranslational modifications). Such characterization of a patient's KIM protein might be useful in identifying deleterious mutants or processing defects which interfere with KIM function and are associated with abnormal patient phenotypes. Each of these techniques is routine to those of skill in the immunological arts.
  • Additional imaging methods utilize KIM or KIM fragments, fused to imageable moieties, for diagnostic imaging of tissues that express KIM ligands, particularly tumors.
  • Further diagnostic techniques are based on demonstration of upregulated KIM mRNA in tissues, as an indication of injury-related processes. This technique has been tested and found workable in a model of ischemic injury in rats, as follows.
  • To determine if the amount of KIM-1 protein is increased after injury, kidney homogenates of contralateral and postischemic kidneys were examined 24 and 48 hours following a 40 minute clamping of the renal artery and vein of a single kidney for each rat. The kidney homogenate was assessed for the presence of KIM-1 protein. Western blot analysis identifies three proteins detected by two different antibodies after ischemic injury, which are not detectable in homogenates from contralateral kidneys which were not exposed to ischemic injury. The apparent molecular weights of the bands are approximately 40 kDa, 50 kDa and 70-80 kDa. The three protein species detected by western blotting could represent glycosylated forms of the same protein given the presence of potential N and O linked glycosylation sites. The fact that each of these proteins react with two different sets of polyclonal antibodies supports the idea that they are related to KIM-1 and are not cross-reacting bands. Confirmation of this prediction came from the results of partial CNBr cleavage of the three proteins which revealed they shared common CNBr cleavage fragments. Since the cytoplasmic domain of the KIM-1 protein is not predicted to contain any major post-translational modifications, the two smallest products of the digest (4.7 kDa and 7.4 kDa) detected with antibodies directed against the cytoplasmic domain of KIM-1 should be the same size for the three different KIM-1 protein bands if they originate from the same protein. The KIM1 40 kDa and 70-80 kDa proteins indeed produced fragments migrating at the predicted size. Digest of the 50 kDa protein band gave also the same C-terminal signature band peptide.
  • The KIM-1 sequence presents two putative sites for N-glycosylation and a mucin domain where O-glycosylation could cover the,polypeptide chain. The three KIM-1 bands detected in postischemic kidney could correspond to glycosylation variants of the same core protein. De-N-glycosylation with PNGase F resulted in a shift of all three bands to a lower molecular weight, corresponding to a loss of about 3 kDa, indicating that all three proteins are N-glycosylated. Differences in O-glycosylation might explain the differences in sizes of these three bands.
  • Further studies are expected to corroborate the correlation between KIM-1 expression and disease status of renal tissue. For example and not by way of limitation, Northern blot analysis can be carried out to establish levels of KIM-1 expression in normal and cancerous renal tissue samples. Poly(A)+ RNA is obtained from tissue samples of interest (e.g., kidney biopsy or necropsy samples, or control samples), size fractionated by electrophoresis, and transferred to a GeneScreen membrane (NEN Life Science Products). Hybridization with a KIM-1 probe corresponding to part or all of the open reading frame of the KIM-1 gene of interest (e.g., human KIM-1, SEQ ID NO:6) is carried out in plaque screening buffer (PSB, 50 mM Tris, pH 7.5, 1 M NaCl, 0.1% sodium pyrophosphate, 0.2% polyvinylpyrrolidone, 0.2% Ficoll, 0.2% bovine serum albumin, 1% SDS) containing 10% dextran sulfate and 100 μg/mL tRNA at 65° C. The KIM-1 probe is preferably detectably labeled, e.g., radiolabelled. The membrane is then washed to remove unbound probe, and KIM-1 mRNA is visualized using standard membrane development techniques. Thereafter, the KIM-1 probe is stripped from the blot, and the blot is rehybridized with a control probe, such as a β-actin probe.
  • The results of an exemplary Northern blot analysis are set forth in FIG. 6: biopsy samples containing histologically normal renal tissue were compared to renal tumor tissues (renal cell carcinoma) obtained from four different patients afflicted with renal cancers. KIM-1 expression was virtually undetectable in three of the four normal samples and was weakly detectable in the fourth. In contract, two of the four renal cancer samples displayed strong KIM-1 expression. These results are consistent with the utility of KIM-1 probes as reagents for the diagnosis, prognostication or staging of renal cancers. Without being limited hereby, it is believed that aberrant KIM-1 gene expression may correlate with the degree of neoplastic transformation, agressiveness or metastatic propensity of a given renal cell carcinoma. Thus, assessment of KIM-1 gene expression is believed to provide information useful in conjunction with other, conventional diagnostic and/or clinical criteria for characterizing renal cancers.
  • THERAPEUTIC USES OF THE COMPOUNDS OF THE INVENTION
  • The therapeutic methods of the invention involve selectively promoting or inhibiting cellular responses that are dependent on KIM ligation. Where the KIM and the KIM ligand are both membrane bound, and expressed by different cells, the signal transduction may occur in the KIM-expressing cell, in the KIM ligand-expressing cell, or in both.
  • KIM ligation-triggered response in a KIM ligand-expressing cell may be generated by contacting the cell with exogenous KIM, KIM fusion proteins or activating antibodies against KIM ligand, either in vitro or in vivo. Further, responses of the KIM ligand-expressing cell that would otherwise be triggered by endogenous KIM could be blocked by contacting the KIM ligand-expressing cell with a KIM ligand antagonist (e.g., an antagonist antibody that binds to KIM ligand), or by contacting the endogenous KIM with an anti-KIM antibody or other KIM-binding molecule which prevents the effective ligation of KIM with a KIM ligand.
  • Similarly, the responses triggered by KIM ligation in the KIM-expressing cell may be promoted or inhibited with exogenous compounds. For example, KIM ligation-triggered response in a KIM-expressing cell may be generated by contacting the cell with a soluble KIM ligand, or certain anti-KIM activating antibodies. Further, responses of the KIM-expressing cell that would otherwise be triggered by interaction with endogenous KIM ligand could be blocked by contacting the KIM-expressing cell with an antagonist to KIM (e.g., a blocking antibody that binds to KIM in a manner that prevents effective, signal-generating KIM ligation), or by contacting the endogenous KIM ligand with an anti-KIM ligand antibody or other KIM ligand-binding molecule which prevents the effective ligation of KIM with the KIM ligand.
  • Which of the interventions described above are useful for particular therapeutic uses depend on the relevant etiologic mechanism of either the pathologic process to be inhibited, or of the medically desirable process to be promoted, as is apparent to those of skill in the medical arts. For example, where KIM ligation results in desirable cellular growth, maintenance of differentiated phenotype, resistance to apoptosis induced by various insults, or other medically advantageous responses, one of the above-described interventions that promote ligation-triggered response may be employed. In the alternative, one of the inhibitory interventions may be useful where KIM ligation invokes undesirable consequences, such as neoplastic growth, deleterious loss of cellular function, susceptibility to apoptosis, or promotion of inflammation events.
  • Following are examples of the previously described therapeutic methods of the invention. One therapeutic use of the KIM-related compounds of the invention is for treating a subject with renal disease, promoting growth of new tissue in a subject, or promoting survival of damaged tissue in a subject, and includes the step of administering to the subject a therapeutically effective amount of a KIM protein of the invention, or of a pharmaceutical composition which includes a protein of the invention. The protein used in these methods may be a fragment of a full-length KIM protein, a soluble KIM ligand protein or fusion fragment, or a KIM agonist. These methods may also be practiced by administering to the subject a therapeutically effective amount of an agonist antibody of the invention, or a pharmaceutical composition which includes an agonist antibody of the invention. A KIM protein may be administered concurrently with a therapeutically effective amount of a second compound which exerts a medically desirable adjunct effect. While tissues of interest for these methods may include any tissue, preferred tissues include renal tissue, liver, neural tissue, heart, stomach, small intestine, spinal cord, or lung. Particular renal conditions which may be beneficially treated with the compounds of the invention include acute renal failure, acute nephritis, chronic renal failure, nephrotic syndrome, renal tubule defects, kidney transplants, toxic injury, hypoxic injury, and trauma. Renal tubule defects include those of either hereditary or acquired nature, such as polycystic renal disease, medullary cystic disease, and medullary sponge kidney. This list is not limited, and may include many other renal disorders (see, e.g., Harrison's Principles of Internal Medicine, 13th ed., 1994, which is herein incorporated by reference.) The subject of the methods may be human.
  • A therapeutic intervention for inhibiting growth of undesirable, KIM ligand-expressing tissue in a subject includes the step of administering to the subject a therapeutically effective amount of a KIM antagonist (e.g., an antagonist antibody that binds to KIM ligand), or by administering a therapeutically effective amount of an anti-KIM antibody or other KIM-binding molecule which blocks KIM binding to the KIM ligand-expressing tissue. In an embodiment of interest, the KIM antagonist or anti-KIM antibody may be used therapeutically to inhibit or block growth of tumors which depend on KIM protein for growth.
  • Other methods of the invention include killing KIM ligand-expressing tumor cells, or inhibiting their growth, by contacting the cells with a fusion protein of a KIM and a toxin or radionuclide, or an anti-KIM ligand antibody conjugated to a toxin or radionuclide. The cell may be within a subject, and the protein or the conjugated antibody is administered to the subject.
  • Also encompassed within the invention is a method for targeting a toxin or radionuclide to a cell expressing a KIM, comprising contacting the cell with a fusion protein comprising a KIM ligand and a toxin or radionuclide, or an anti-KIM antibody conjugated to a toxin or radionuclide. Another embodiment includes the method of suppressing growth of a tumor cell which expresses KIM, comprising contacting the cell with a fusion protein of KIM ligand and a toxin or radionuclide or with an anti-KIM antibody conjugated to a toxin or radionuclide; the cell may be within a subject, and the protein administered to the subject.
  • The term “subject” used herein is taken to mean any mammal to which KIM may be administered. Subjects specifically intended for treatment with the method of the invention include humans, as well as nonhuman primates, sheep, horses, cattle, goats, pigs, dogs, cats, rabbits, guinea pigs, hamsters, gerbils, rats and mice, as well as the organs, tumors, and cells derived or originating from these hosts.
  • USE OF COMPOUNDS OF THE INVENTION IN GENE THERAPY
  • The KIM genes of the invention are introduced into damaged tissue, or into tissue where stimulated growth is desirable. Such gene therapy stimulates production of KIM protein by the transfected cells, promoting cell growth and/or survival of cells that express the KIM protein.
  • In a specific embodiment of a gene therapy method, a gene coding for a KIM protein may be introduced into a renal target tissue. The KIM protein would be stably expressed and stimulate tissue growth, division, or differentiation, or could potentiate cell survival. Furthermore, a KIM gene may be introduced into a target cell using a variety of well-known methods that use either viral or non-viral based strategies.
  • Non-viral methods include electroporation, membrane fusion with liposomes, high velocity bombardment with DNA-coated microprojectiles, incubation with calcium-phosphate-DNA precipitate, DEAE-dextran mediated transfection, and direct micro-injection into single cells. For instance, a KIM gene may be introduced into a cell by calcium phosphate coprecipitation (Pillicer et al., Science, 209: 1414-1422 (1980); mechanical microinjection and/or particle acceleration (Anderson et al., Proc. Nat. Acad. Sci. USA, 77: 5399-5403 (1980); liposome based DNA transfer (e.g., LIPOFECTIN-mediated transfection—Fefgner et al., Proc. Nat. Acad. Sci., USA, 84: 471-477,1987; Gao and Huang, Biochim. Biophys. Res. Comm., 179: 280-285, 1991; DEAE Dextran-mediated transfection; electroporation (U.S. Pat. No. 4,956,288); or polylysine-based methods in which DNA is conjugated to deliver DNA preferentially to liver hepatocytes (Wolff et al., Science, 247: 465-468, 1990; Curiel et al., Human Gene Therapy 3: 147-154, 1992).
  • Target cells may be transfected with the genes of the invention by direct gene transfer. See, e.g., Wolff et al., “Direct Gene Transfer Into Moose Muscle In Vivo”, Science 247:1465-68, 1990. In many cases, vector-mediated transfection will be desirable. Any of the methods known in the art for the insertion of polynucleotide sequences into a vector may be used. ( See, for example, Sambrook et al., Molecular Cloning: A Laboratory Manual, Cold Spring Harbor Laboratory, Cold Spring Harbor, N.Y., 1989; and Ausubel et al., Current Protocols in Molecular Biology, J. Wiley & Sons, NY, 1992, both of which are incorporated herein by reference.) Promoter activation may be tissue specific or inducible by a metabolic product or administered substance. Such promoters/enhancers include, but are not limited to, the native c-ret ligand protein promoter, the cytomegalovirus immediate-early promoter/enhancer (Karasuyama et al., J. Exp. Med., 169: 13, 1989); the human beta-actin promoter (Gunning et al., Proc. Nat. Acad. Sci. USA, 84: 4831, 1987; the glucocorticoid-inducible promoter present in the mouse mammary tumor virus long terminal repeat (MMTV LTR) (Klessig et al., Mol. Cell. Biol., 4: 1354, 1984); the long terminal repeat sequences of Moloney murine leukemia virus (MuLV LTR) (Weiss et al., RNA Tumor Viruses, Cold Spring Harbor Laboratory, Cold Spring Harbor, N.Y., 1985); the SV40 early region promoter (Bernoist and Chambon, Nature, 290:304, 1981); the promoter of the Rous sarcoma virus (RSV) (Yamamoto et al., Cell, 22:787, 1980); the herpes simplex virus (HSV) thymidine kinase promoter (Wagner et al., Proc. Nat. Acad. Sci. USA, 78: 1441, 1981); the adenovirus promoter (Yamada et al., Proc. Nat. Acad. Sci. USA, 82: 3567, 1985).
  • The KIM genes may also be introduced by specific viral vectors for use in gene transfer systems which are now well established. See for example: Madzak et al., J. Gen. Virol., 73: 1533-36, 1992 (papovavirus SV40); Berkner et al., Curr. Top. Microbiol. Immunol., 158: 39-61, 1992 (adenovirus); Hofmann et al., Proc. Natl. Acad. Sci. 92: 10099-10103, 1995 (baculovirus); Moss et al., Curr. Top. Microbiol. Immunol., 158: 25-38, 1992 (vaccinia virus); Muzyczka, Curr. Top. Microbiol. Immunol., 158: 97-123, 1992 (adeno-associated virus); Margulskee, Curr. Top. Microbiol. Immunol., 158: 67-93, 1992 (herpes simplex virus (HSV) and Epstein-Barr virus (HBV)); Miller, Curr. Top. Microbiol. Immunol., 158: 1-24, 1992 (retrovirus); Brandyopadhyay et al., Mol. Cell. Biol., 4: 749-754, 1984 (retrovirus); Miller et al., Nature, 357: 455-450, 1992 (retrovirus); Anderson, Science, 256: 808-813, 1992 (retrovirus), Current Protocols in Molecular Biology: Sections 9.10-9.14 (Ausubel et al., Eds.), Greene Publishing Associcates, 1989, all of which are incorporated herein by reference.
  • Preferred vectors are DNA viruses that include adenoviruses (preferably Ad-2 or Ad-5 based vectors), baculovirus, herpes viruses (preferably herpes simplex virus based vectors), and parvoviruses (preferably “defective” or non-autonomous parvovirus based vectors, more preferably adeno-associated virus based vectors, most preferably AAV-2 based vectors). See, e.g., Ali et al., Gene Therapy 1: 367-384, 1994; U.S. Pat. Nos. 4,797,368 and 5,399,346 and discussion below.
  • The choice of a particular vector system for transferring, for instance, a KIM sequence will depend on a variety of factors. One important factor is the nature of the target cell population. Although retroviral vectors have been extensively studied and used in a number of gene therapy applications, they are generally unsuited for infecting cells that are not dividing but may be useful in cancer therapy since they only integrate and express their genes in replicating cells. They are useful for ex vivo approaches and are attractive in this regard due to their stable integration into the target cell genome.
  • Adenoviruses are eukaryotic DNA viruses that can be modified to efficiently deliver a therapeutic or reporter transgene to a variety of cell types. The general adenoviruses types 2 and 5 (Ad2 and Ad5, respectively), which cause respiratory disease in humans, are currently being developed for gene therapy of Duchenne Muscular Dystrophy (DMD) and Cystic Fibrosis (CF). Both Ad2 and Ad5 belong to a subclass of adenovirus that are not associated with human malignancies. Adenovirus vectors are capable of providing extremely high levels of transgene delivery to virtually all cell types, regardless of the mitotic state. High titers (1013 plaque forming units/ml) of recombinant virus can be easily generated in 293 cells (an adenovirus-transformed, complementation human embryonic kidney cell line: ATCC CRL1573) and cryo-stored for extended periods without appreciable losses. The efficacy of this system in delivering a therapeutic transgene in vivo that complements a genetic imbalance has been demonstrated in animal models of various disorders. See Watanabe, Atherosclerosis, 36: 261-268, 1986; Tanzawa et al., FEBS Letters 118(1):81-84, 1980; Golasten et al., New Engl. J. Med. 309:288-296, 1983; Ishibashi et al., J. Clin. Invest. 92: 883-893, 1993; and Ishibashi et al., J. Clin. Invest. 93: 1889-1893, 1994, all of which are incorporated herein by reference. Indeed, recombinant replication defective adenovirus encoding a cDNA for the cystic fibrosis transmembrane regulator (CFTR) has been approved for use in at least two human CF clinical trials. See, e.g., Wilson, Nature 365:691-692, 1993. Further support of the safety of recombinant adenoviruses for gene therapy is the extensive experience of live adenovirus vaccines in human populations.
  • The first-generation recombinant, replication-deficient adenoviruses which have been developed for gene therapy of DMD and other inherited disorders contain deletions of the entire E1a and part of the E1b regions. This replication-defective virus is grown in 293 cells containing a functional adenovirus E1a gene which provides a transacting E1a protein. E1-deleted viruses are capable of replicating and producing infectious virus in the 293 cells, which provide E1a and E1b region gene products in trans. The resulting virus is capable of infecting many cell types and can express the introduced gene (providing it carries its own promoter), but cannot replicate in a cell that does not carry the E1 region DNA unless the cell is infected at a very high multiplicity of infection. Adenoviruses have the advantage that they have a broad host range, can infect quiescent or terminally differentiated cells such as neurons, and appear essentially non-oncogenic. Adenoviruses do not appear to integrate into the host genome. Because they exist extrachromasomally, the risk of insertional mutagenesis is greatly reduced. Ali et al., supra, at 373. Recombinant adenoviruses (rAdV) produce very high titers, the viral particles are moderately stable, expression levels are high, and a wide range of cells can be infected. Their natural host cells are airway epithelium, so they are useful for therapy of lung cancers.
  • Baculovirus-mediated transfer has several advantages. Baculoviral gene transfer can occur in replicating and nonreplicating cells, and can occur in renal cells, as well as in hepatocytes, neural cells, spleen, skin, and muscle. Baculovirus is non-replicating and nonpathogenic in mammalian cells. Humans lack pre-existing antibodies to recombinant baculovirus which could block infection. In addition, baculovirus is capable of incorporating and transducing very large DNA inserts.
  • Adeno-associated viruses (AAV) have also been employed as vectors for somatic gene therapy. AAV is a small, single-stranded (ss) DNA virus with a simple genomic organization (4-7 kb) that makes it an ideal substrate for genetic engineering. Two open reading frames encode a series of rep and cap polypeptides. Rep polypeptides (rep78, rep68, rep 62 and rep 40) are involved in replication, rescue and integration of the AAV genome. The cap proteins (VP1, VP2 and VP3) form the virion capsid. Flanking the rep and cap open reading frames at the 5′ and 3′ ends are 145 bp inverted terminal repeats (ITRs), the first 125 bp of which are capable of forming Y- or T-shaped duplex structures. Of importance for the development of AAV vectors, the entire rep and cap domains can be excised and replaced with a therapeutic or reporter transgene. See B. J. Carter, in Handbook of Parvoviruses, ed., P. Tijsser, CRC Press, pp. 155-168 (1990). It has been shown that the ITRs represent the minimal sequence required for replication, rescue, packaging, and integration of the AAV genome.
  • Adeno-associated viruses (AAV) have significant potential in gene therapy. The viral particles are very stable and recombinant AAVs (rAAV) have “drug-like” characteristics in that rAAV can be purified by pelleting or by CsCl gradient banding. They are heat stable and can be lyophilized to a powder and rehydrated to full activity. Their DNA stably integrates into host chromosomes so expression is long-term. Their host range is broad and AAV causes no known disease so that the recombinant vectors are non-toxic.
  • Once introduced into a target cell, sequences of interest can be identified by conventional methods such as nucleic acid hybridization using probes comprising sequences that are homologous/complementary to the inserted gene sequences of the vector. In another approach, the sequence(s) may be identified by the presence or absence of a “marker” gene function (e.g, thymidine kinase activity, antibiotic resistance, and the like) caused by introduction of the expression vector into the target cell.
  • Formulations and Administration
  • The compounds of the invention are formulated according to standard practice, such as prepared in a carrier vehicle. The term “pharmacologically acceptable carrier” means one or more organic or inorganic ingredients, natural or synthetic, with which the mutant proto-oncogene or mutant oncoprotein is combined to facilitate its application. A suitable carrier includes sterile saline although other aqueous and non-aqueous isotonic sterile solutions and sterile suspensions known to be pharmaceutically acceptable are known to those of ordinary skill in the art. In this regard, the term “carrier” encompasses liposomes and the HIV-1 tat protein (See Chen et al., Anal. Biochem. 227: 168-175, 1995) as well as any plasmid and viral expression vectors.
  • Any of the novel polypeptides of this invention may be used in the form of a pharmaceutically acceptable salt. Suitable acids and bases which are capable of forming salts with the polypeptides of the present invention are well known to those of skill in the art, and include inorganic and organic acids and bases.
  • A compound of the invention is administered to a subject in a therapeutically-effective amount, which means an amount of the compound which produces a medically desirable result or exerts an influence on the particular condition being treated. An effective amount of a compound of the invention is capable of ameliorating or delaying progression of the diseased, degenerative or damaged condition. The effective amount can be determined on an individual basis and will be based, in part, on consideration of the physical attributes of the subject, symptoms to be treated and results sought. An effective amount can be determined by one of ordinary skill in the art employing such factors and using no more than routine experimentation.
  • A liposome delivery system for a compound of the invention may be any of a variety of unilamellar vesicles, multilamellar vesicles, or stable plurilamellar vesicles, and may be prepared and administered according to methods well known to those of skill in the art, for example in accordance with the teachings of U.S. Pat. Nos. 5,169,637, 4,762,915, 5,000,958 or 5,185,154. In addition, it may be desirable to express the novel polypeptides of this invention, as well as other selected polypeptides, as lipoproteins, in order to enhance their binding to liposomes. As an example, treatment of human acute renal failure with liposome-encapsulated KIM protein may be performed in vivo by introducing a KIM protein into cells in need of such treatment using liposomes. The liposomes can be delivered via catheter to the renal artery. The recombinant KIM protein is purified, for example, from CHO cells by immunoaffinity chromatography or any other convenient method, then mixed with liposomes and incorporated into them at high efficiency. The encapsulated protein may be tested in vitro for any effect on stimulating cell growth.
  • The compounds of the invention may be administered in any manner which is medically acceptable. This may include injections, by parenteral routes such as intravenous, intravascular, intraarterial, subcutaneous, intramuscular, intratumor, intraperitoneal, intraventricular, intraepidural, or others as well as oral, nasal, ophthalmic, rectal, or topical. Sustained release administration is also specifically included in the invention, by such means as depot injections or erodible implants. Localized delivery is particularly contemplated, by such means as delivery via a catheter to one or more arteries, such as the renal artery or a vessel supplying a localized tumor.
  • Equivalents
  • Those skilled in the arts will recognize, or be able to ascertain using no more than routine experimentation, many equivalents to the specific embodiments of the invention described herein. All such equivalents are embraced by the following claims.

Claims (31)

1-3. (canceled)
4. An isolated nucleic acid comprising a nucleotide sequence selected from the group consisting of
SEQ ID NO:1, SEQ ID NO:2, SEQ ID NO:4, and SEQ ID NO:6.
5-7. (canceled)
8. An isolated nucleic acid comprising a nucleotide sequence encoding a polypeptide comprising an amino acid sequence selected from the group consisting of SEQ ID NO:3, SEQ ID NO:5, and SEQ ID NO:7.
9-13. (canceled)
14. A vector comprising the nucleic acid of claim 4.
15. A host cell comprising the vector of claim 14.
16. A polypeptide production method, comprising the steps of: culturing a host cell of claim 15 in a cell culture medium; and, recovering a KIM polypeptide expressed from the vector insert within said host cell.
17. An isolated polypeptide comprising an amino acid sequence
a) selected from the group consisting of SEQ ID NO:3, SEQ ID NO:5, and SEQ ID NO:7.
18. A soluble variant of the polypeptide of claim 17.
19. A fusion protein comprising the polypeptide of claim 17; and, an immunoglobulin, toxin, or imageable compound.
20. The polypeptide of claim 17 conjugated to an immunoglobulin, toxin, imageable compound or radionuclide.
21. An antibody that binds specifically to the polypeptide claim 17.
22. An antibody of claim 21 conjugated to a toxin, imageable compound or radionuclide.
23. A hybridoma that produces an antibody of claim 21.
24. A pharmaceutical composition comprising a physiologically acceptable carrier having dispersed therein, to a therapeutically effective concentration, a vector of claim 14.
25. A pharmaceutical composition comprising a physiologically acceptable carrier having dispersed therein, to a therapeutically effective concentration, the polypeptide of claim 17.
26. A pharmaceutical composition comprising a physiologically acceptable carrier having dispersed therein, to a therapeutically effective concentration, the antibody of claim 21.
27. A method of assessing presence or course of resolution of renal injury in a subject, comprising the step of measuring the concentration of a KIM nucleic acid or a KIM polypeptide in urine, serum, or urine sediment of a subject afflicted with, or at risk of developing, renal injury or disease.
28. A method of imaging cells or tissue producing a KIM polypeptide, comprising the step of contacting the cells or tissue with an imageable KIM antibody conjugate of claim 22.
29. (canceled)
30. A method of targeting a toxin, radionuclide or imageable compound to cells producing a KIM polypeptide, comprising the step of contacting the cells with a KIM antibody conjugate of claim 22.
31. (canceled)
32. A method of treating a subject afflicted with, or at risk of developing, renal disease, comprising the step of administering the pharmaceutical composition of claim 25 to the subject.
33. A method of promoting tissue growth in a subject, comprising the step of administering the pharmaceutical composition of claim 25 to the subject.
34. A method of promoting survival of damaged tissue in a subject, comprising the step of administering the pharmaceutical composition of claim 25 to the subject.
35-36. (canceled)
37. A method of treating a subject afflicted with, or at risk of developing, renal disease, comprising the step of administering the pharmaceutical composition of claim 26 to the subject.
38. A method of promoting tissue growth in a subject, comprising the step of administering the pharmaceutical composition of claim 26 to the subject.
39. A method of promoting survival of damaged tissue in a subject, comprising the step of administering the pharmaceutical composition of claim 26 to the subject.
40. The method of claim 37, wherein the renal disease is renal cancer.
US11/463,239 1996-05-24 2006-08-08 Modulators of tissue regeneration Abandoned US20070141590A1 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
US11/463,239 US20070141590A1 (en) 1996-05-24 2006-08-08 Modulators of tissue regeneration

Applications Claiming Priority (6)

Application Number Priority Date Filing Date Title
US1822896P 1996-05-24 1996-05-24
US2344296P 1996-08-23 1996-08-23
PCT/US1997/009303 WO1997044460A1 (en) 1996-05-24 1997-05-23 Modulators of tissue regeneration
US09/197,970 US6664385B1 (en) 1996-05-24 1998-11-23 Kidney injury-related molecules
US10/655,506 US20050089868A1 (en) 1996-05-24 2003-09-04 Modulators of tissue regeneration
US11/463,239 US20070141590A1 (en) 1996-05-24 2006-08-08 Modulators of tissue regeneration

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
US10/655,506 Division US20050089868A1 (en) 1996-05-24 2003-09-04 Modulators of tissue regeneration

Publications (1)

Publication Number Publication Date
US20070141590A1 true US20070141590A1 (en) 2007-06-21

Family

ID=26690881

Family Applications (4)

Application Number Title Priority Date Filing Date
US09/197,970 Expired - Lifetime US6664385B1 (en) 1996-05-24 1998-11-23 Kidney injury-related molecules
US10/655,506 Abandoned US20050089868A1 (en) 1996-05-24 2003-09-04 Modulators of tissue regeneration
US11/463,239 Abandoned US20070141590A1 (en) 1996-05-24 2006-08-08 Modulators of tissue regeneration
US11/463,227 Abandoned US20060286031A1 (en) 1996-05-24 2006-08-08 Modulators of tissue regeneration

Family Applications Before (2)

Application Number Title Priority Date Filing Date
US09/197,970 Expired - Lifetime US6664385B1 (en) 1996-05-24 1998-11-23 Kidney injury-related molecules
US10/655,506 Abandoned US20050089868A1 (en) 1996-05-24 2003-09-04 Modulators of tissue regeneration

Family Applications After (1)

Application Number Title Priority Date Filing Date
US11/463,227 Abandoned US20060286031A1 (en) 1996-05-24 2006-08-08 Modulators of tissue regeneration

Country Status (27)

Country Link
US (4) US6664385B1 (en)
EP (2) EP0907735B9 (en)
JP (3) JP4602482B2 (en)
CN (1) CN1147584C (en)
AT (1) ATE318903T1 (en)
AU (1) AU712289B2 (en)
BG (1) BG64678B1 (en)
BR (1) BR9709115A (en)
CA (1) CA2257851C (en)
CZ (1) CZ295936B6 (en)
DE (1) DE69735364T3 (en)
DK (1) DK0907735T5 (en)
EA (1) EA004402B1 (en)
EE (1) EE04817B1 (en)
ES (1) ES2258793T5 (en)
HK (1) HK1021746A1 (en)
HU (1) HU226205B1 (en)
IL (1) IL127162A (en)
IS (1) IS2636B (en)
NO (2) NO327597B1 (en)
NZ (1) NZ336467A (en)
PL (1) PL188826B1 (en)
PT (1) PT907735E (en)
SI (1) SI0907735T2 (en)
SK (1) SK285461B6 (en)
TR (1) TR199802421T2 (en)
WO (1) WO1997044460A1 (en)

Cited By (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20050089868A1 (en) * 1996-05-24 2005-04-28 Michele Sanicola-Nadel Modulators of tissue regeneration
US20100080798A1 (en) * 2002-12-30 2010-04-01 Biogen Idec Ma Inc. Kim-1 antagonists and use to modulate immune system
US20100150905A1 (en) * 2005-03-02 2010-06-17 Biogen Idec Ma Inc. Kim-1 antibodies for treatment of th2-mediated conditions

Families Citing this family (50)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
AU7592998A (en) * 1997-05-23 1998-12-11 Biogen, Inc. Modulators of tissue regeneration
WO1999049029A1 (en) 1998-03-20 1999-09-30 Benitec Australia Ltd Control of gene expression
AUPP249298A0 (en) 1998-03-20 1998-04-23 Ag-Gene Australia Limited Synthetic genes and genetic constructs comprising same I
US6423885B1 (en) 1999-08-13 2002-07-23 Commonwealth Scientific And Industrial Research Organization (Csiro) Methods for obtaining modified phenotypes in plant cells
EP1160321A1 (en) * 2000-05-31 2001-12-05 Sanofi-Synthelabo Kidney Injury Novel Gene-1: Isolation and therapeutic applications
US7179901B2 (en) 2000-06-16 2007-02-20 Biogen Idec Ma Inc. Renal regulatory elements and methods of use thereof
US6812002B2 (en) 2000-08-30 2004-11-02 Pfizer Inc. Osteoactivin protein and nucleic acids encoding the same, compositions and methods of stimulating bone differentiation
AU2002305785B2 (en) * 2001-06-01 2008-01-17 Biogen Ma Inc. Molecules and methods for inhibiting shedding of KIM-1
CA2452196A1 (en) * 2001-06-29 2003-01-09 The Board Of Trustees Of The Leland Stanford Junior University T cell regulatory genes and methods of use thereof
US8709412B2 (en) 2001-06-29 2014-04-29 The Board Of Trustees Of The Leland Stanford Junior University Modulation of TIM receptor activity in combination with cytoreductive therapy
CA2479732A1 (en) * 2002-03-19 2003-10-02 Curagen Corporation Therapeutic polypeptides, nucleic acids encoding same, and methods of use
ES2557769T3 (en) * 2003-03-19 2016-01-28 Amgen Fremont Inc. Antibodies against the T cell immunoglobulin domain antigen and mucin domain 1 (TIM-1) and uses thereof
MXPA05010385A (en) * 2003-03-27 2006-03-08 Childrens Hosp Medical Center A method and kit for detecting the early onset of renal tubular cell injury.
US20050272101A1 (en) * 2004-06-07 2005-12-08 Prasad Devarajan Method for the early detection of renal injury
AU2005240190C1 (en) 2004-05-06 2012-01-19 Children's Hospital Medical Center NGAL for reduction and amelioration of ischemic and nephrotoxic injuries
RU2283666C9 (en) * 2004-05-14 2007-03-10 Бизяев Алексей Вячеславович Agent for accelerated repairing of structure and function of damaged tissues and organs
US20060003345A1 (en) * 2004-06-30 2006-01-05 Pfizer Inc RNA bioassay
EP3489689B1 (en) * 2004-12-20 2020-06-17 Antibodyshop A/S Determination of neutrophil gelatinase-associated lipocalin (ngal) as a diagnostic marker for renal disorders
US20070037232A1 (en) * 2005-03-31 2007-02-15 Barasch Jonathan M Detection of NGAL in chronic renal disease
US20080090304A1 (en) * 2006-10-13 2008-04-17 Barasch Jonathan Matthew Diagnosis and monitoring of chronic renal disease using ngal
CA2629453C (en) 2005-11-10 2018-03-06 Curagen Corporation Method of treating ovarian and renal cancer using antibodies against t cell immunoglobulin domain and mucin domain 1 (tim-1) antigen
US20090197280A1 (en) 2006-05-30 2009-08-06 Kristian Bangert Methods and Devices for Rapid Assessment of Severity of Injury
WO2008017306A1 (en) * 2006-08-07 2008-02-14 Antibodyshop A/S Diagnostic test to exclude significant renal injury
JP2010521694A (en) * 2007-03-21 2010-06-24 バイオポルト・ダイアグノスティクス・アクティーゼルスカブ Diagnostic tests for kidney injury
US8846036B2 (en) 2007-10-19 2014-09-30 Abbott Laboratories Antibodies that bind to mammalian NGAL and uses thereof
US20090297479A1 (en) * 2008-03-28 2009-12-03 Kiyoshi Ariizumi Dc-hil conjugates for treatment of t-cell disorders
EP2324355B1 (en) * 2008-08-28 2014-01-22 Astute Medical, Inc. Methods and compositions for diagnosis and prognosis of renal injury and renal failure
US9057735B2 (en) * 2008-08-29 2015-06-16 Astute Medical, Inc. Methods and compositions for diagnosis and prognosis of renal injury and renal failure
WO2010048346A1 (en) 2008-10-21 2010-04-29 Astute Medical, Inc. Methods and compositions for diagnosis and prognosis of renal injury and renal failure
CA2740923A1 (en) * 2008-10-21 2010-04-29 Astute Medical, Inc. Methods and compositions for diagnosis and prognosis of renal injury and renal failure
WO2010054389A1 (en) 2008-11-10 2010-05-14 Astute Medical, Inc. Methods and compositions for diagnosis and prognosis of renal injury and renal failure
WO2010059996A1 (en) * 2008-11-22 2010-05-27 Astute Medical, Inc. Methods and compositions for diagnosis and prognosis of renal injury and renal failure
CN102300875B (en) * 2009-01-28 2014-07-16 财团法人工业技术研究院 Biomarkers associated with nephropathy
AU2010279249B2 (en) 2009-02-06 2015-08-13 Astute Medical, Inc. Methods and compositions for diagnosis and prognosis of renal injury and renal failure
US9229010B2 (en) 2009-02-06 2016-01-05 Astute Medical, Inc. Methods and compositions for diagnosis and prognosis of renal injury and renal failure
EP2462446B1 (en) * 2009-08-07 2017-09-27 Astute Medical, Inc. Methods and compositions for diagnosis and prognosis of renal injury and renal failure
CN104793000A (en) 2009-11-07 2015-07-22 阿斯图特医药公司 Methods and compositions for diagnosis and prognosis of renal injury and renal failure
NZ600828A (en) 2009-12-20 2014-09-26 Astute Medical Inc Methods and compositions for diagnosis and prognosis of renal injury and renal failure
PT2666872T (en) 2010-02-05 2016-07-08 Astute Medical Inc Methods and compositions for diagnosis and prognosis of renal injury and renal failure
EP2539712A4 (en) 2010-02-26 2013-09-18 Astute Medical Inc Methods and compositions for diagnosis and prognosis of renal injury and renal failure
EP2576587B1 (en) 2010-05-24 2018-03-21 The Trustees of Columbia University in the City of New York Mutant ngal proteins and uses thereof
US20130203074A1 (en) 2010-06-23 2013-08-08 Astute Medical, Inc. Methods and compositions for diagnosis and prognosis of renal injury and renal failure
EP3339860A1 (en) 2010-06-23 2018-06-27 Astute Medical, Inc. Methods and compositions for diagnosis and prognosis of renal injury and renal failure
JP5905032B2 (en) * 2011-02-09 2016-04-20 ラヴィヴォ・アーベー Synbiotic composition for recovery and reconstitution of intestinal microbiota
ES2734494T3 (en) 2011-12-08 2019-12-10 Astute Medical Inc Methods and uses for the diagnosis of kidney injury and kidney failure
WO2014081980A2 (en) 2012-11-21 2014-05-30 The Trustees Of Columbia University In The City Of New York Mutant ngal proteins and uses thereof
WO2014113558A1 (en) 2013-01-17 2014-07-24 Astute Medical, Inc. Methods and compositions for diagnosis and prognosis of renal injury and renal failure
US10420337B2 (en) 2013-03-15 2019-09-24 Lifeline Scientific, Inc. Transporter with a glucose sensor for determining viability of an organ or tissue
WO2015134671A1 (en) 2014-03-04 2015-09-11 Targeson, Inc. Molecular imaging contrast agents and uses thereof
JP2019523889A (en) 2016-06-06 2019-08-29 アスチュート メディカル,インコーポレイテッド Management of acute kidney injury using tissue inhibitors of insulin-like growth factor binding protein 7 and metalloprotease 2

Citations (8)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US2003A (en) * 1841-03-12 Improvement in horizontal windivhlls
US5622861A (en) * 1994-08-05 1997-04-22 The United States Of America As Represented By The Department Of Health And Human Services Recombinant DNA encoding hepatitis A virus receptor
US5654403A (en) * 1991-10-28 1997-08-05 Burroughs Wellcome Co. Immunoglobulins stabilized with a chelator of copper ions
US5989554A (en) * 1994-11-10 1999-11-23 Promega Corporation High level expression and facile purification of proteins, peptides and conjugates for immunization, purification and detection applications
US6165745A (en) * 1992-04-24 2000-12-26 Board Of Regents, The University Of Texas System Recombinant production of immunoglobulin-like domains in prokaryotic cells
US20030021583A1 (en) * 2001-07-27 2003-01-30 Shu Lin Changing a playback speed for video presentation recorded in a field structure format
US6664385B1 (en) * 1996-05-24 2003-12-16 Biogen, Inc. Kidney injury-related molecules
US7041290B2 (en) * 2001-06-01 2006-05-09 Biogen Idec Ma Inc. Molecules and methods for inhibiting shedding of KIM-1

Family Cites Families (13)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5019368A (en) * 1989-02-23 1991-05-28 Cancer Biologics, Inc. Detection of necrotic malignant tissue and associated therapy
WO1995021922A2 (en) * 1994-02-14 1995-08-17 Abbott Laboratories Non-a, non-b, non-c, non-d, non-e hepatitis reagents and methods for their use
WO1996006159A1 (en) * 1994-08-18 1996-02-29 The Trustees Of Columbia University In The City Of New York Unique associated kaposi's sarcoma virus sequences and uses thereof
US6066322A (en) * 1995-03-03 2000-05-23 Millennium Pharmaceuticals, Inc. Methods for the treatment of immune disorders
US7179901B2 (en) * 2000-06-16 2007-02-20 Biogen Idec Ma Inc. Renal regulatory elements and methods of use thereof
US7838220B2 (en) * 2001-06-29 2010-11-23 The Board Of Trustees Of The Leland Stanford Junior University T cell regulatory genes associated with immune disease
CA2452196A1 (en) * 2001-06-29 2003-01-09 The Board Of Trustees Of The Leland Stanford Junior University T cell regulatory genes and methods of use thereof
US7687454B2 (en) * 2001-12-03 2010-03-30 The University Of British Columbia Effectors of innate immunity determination
EP4091631A1 (en) * 2002-01-30 2022-11-23 The Brigham and Women's Hospital, Inc. A tim-3 binding molecule for use in the treatment of a disease
CA2479732A1 (en) * 2002-03-19 2003-10-02 Curagen Corporation Therapeutic polypeptides, nucleic acids encoding same, and methods of use
NZ541404A (en) * 2002-12-30 2007-05-31 Biogen Idec Inc Antibody or antigen-binding fragment thereof that binds to KIM-1, or a polypeptide comprising a KIM-I immunoglobulin domain to treat autoimmune diseases
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
BRPI0509139A (en) * 2004-03-24 2007-08-28 Telos Fharmaceuticals Inc composition, and method of stimulating an immune response in an individual, prophylactically treating a disease, ameliorating a sign or symptom associated with a disease, tumor targeting, tumor growth inhibition, and detection of a tumor

Patent Citations (10)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US2003A (en) * 1841-03-12 Improvement in horizontal windivhlls
US5654403A (en) * 1991-10-28 1997-08-05 Burroughs Wellcome Co. Immunoglobulins stabilized with a chelator of copper ions
US6165745A (en) * 1992-04-24 2000-12-26 Board Of Regents, The University Of Texas System Recombinant production of immunoglobulin-like domains in prokaryotic cells
US5622861A (en) * 1994-08-05 1997-04-22 The United States Of America As Represented By The Department Of Health And Human Services Recombinant DNA encoding hepatitis A virus receptor
US5989554A (en) * 1994-11-10 1999-11-23 Promega Corporation High level expression and facile purification of proteins, peptides and conjugates for immunization, purification and detection applications
US6664385B1 (en) * 1996-05-24 2003-12-16 Biogen, Inc. Kidney injury-related molecules
US20050089868A1 (en) * 1996-05-24 2005-04-28 Michele Sanicola-Nadel Modulators of tissue regeneration
US7041290B2 (en) * 2001-06-01 2006-05-09 Biogen Idec Ma Inc. Molecules and methods for inhibiting shedding of KIM-1
US20060153836A1 (en) * 2001-06-01 2006-07-13 Biogen Idec MA Inc., Massachusetts corporation Molecules and methods for inhibiting shedding of KIM-1
US20030021583A1 (en) * 2001-07-27 2003-01-30 Shu Lin Changing a playback speed for video presentation recorded in a field structure format

Cited By (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20050089868A1 (en) * 1996-05-24 2005-04-28 Michele Sanicola-Nadel Modulators of tissue regeneration
US20100080798A1 (en) * 2002-12-30 2010-04-01 Biogen Idec Ma Inc. Kim-1 antagonists and use to modulate immune system
US20100150905A1 (en) * 2005-03-02 2010-06-17 Biogen Idec Ma Inc. Kim-1 antibodies for treatment of th2-mediated conditions
US8206705B2 (en) 2005-03-02 2012-06-26 Biogen Idec Ma Inc. KIM-1 antibodies for treatment of TH2-mediated conditions

Also Published As

Publication number Publication date
DK0907735T3 (en) 2006-06-26
US20060286031A1 (en) 2006-12-21
DK0907735T5 (en) 2010-06-14
WO1997044460A1 (en) 1997-11-27
EP0907735B1 (en) 2006-03-01
EP0907735B9 (en) 2010-05-19
HK1021746A1 (en) 2000-06-30
JP4316640B2 (en) 2009-08-19
AU3567697A (en) 1997-12-09
BG102967A (en) 2000-05-31
BR9709115A (en) 1999-08-03
NO985427L (en) 1999-01-25
TR199802421T2 (en) 1999-02-22
CA2257851C (en) 2011-11-15
EE04817B1 (en) 2007-04-16
EP0907735A1 (en) 1999-04-14
NO985427D0 (en) 1998-11-20
EP0907735B2 (en) 2009-10-07
SI0907735T1 (en) 2006-08-31
IS2636B (en) 2010-06-15
IL127162A0 (en) 1999-09-22
IL127162A (en) 2007-07-24
JP2008067701A (en) 2008-03-27
AU712289B2 (en) 1999-11-04
ATE318903T1 (en) 2006-03-15
US20050089868A1 (en) 2005-04-28
CN1223685A (en) 1999-07-21
ES2258793T3 (en) 2006-09-01
DK0907735T4 (en) 2009-12-21
DE69735364T2 (en) 2006-11-30
HU226205B1 (en) 2008-06-30
DE69735364D1 (en) 2006-04-27
ES2258793T5 (en) 2010-01-25
JP2009039111A (en) 2009-02-26
PL188826B1 (en) 2005-04-29
EA004402B1 (en) 2004-04-29
IS4902A (en) 1998-11-20
CA2257851A1 (en) 1997-11-27
US6664385B1 (en) 2003-12-16
CN1147584C (en) 2004-04-28
EA199801044A1 (en) 1999-04-29
JP4602482B2 (en) 2010-12-22
SI0907735T2 (en) 2010-01-29
NZ336467A (en) 2000-10-27
CZ381398A3 (en) 1999-05-12
EP1655367A1 (en) 2006-05-10
CZ295936B6 (en) 2005-12-14
SK160998A3 (en) 1999-07-12
PT907735E (en) 2006-06-30
DE69735364T3 (en) 2010-05-06
HUP9902770A3 (en) 2001-09-28
JP2001505761A (en) 2001-05-08
HUP9902770A2 (en) 1999-12-28
SK285461B6 (en) 2007-02-01
NO20090945L (en) 1999-01-25
BG64678B1 (en) 2005-11-30
EE9800409A (en) 1999-06-15
NO327597B1 (en) 2009-08-31
PL330313A1 (en) 1999-05-10

Similar Documents

Publication Publication Date Title
US6664385B1 (en) Kidney injury-related molecules
EP0914339B1 (en) RET LIGAND 3 (RetL3) FOR STIMULATING NEURAL AND RENAL GROWTH
US6861509B1 (en) Antibodies to Ret and RetL3
WO1995016709A2 (en) Anti-mullerian hormone receptor polypeptides and antibodies thereto
KR100498530B1 (en) Tissue regeneration regulator
US20040101528A1 (en) Type I IL-1 receptors
MXPA98009812A (en) Modulators of the regeneration of the tej
KR100554901B1 (en) RET Ligand to stimulate nerve and kidney proliferation
CA2496906A1 (en) Ret ligand (retl) for stimulating neural and renal growth

Legal Events

Date Code Title Description
AS Assignment

Owner name: THE GENERAL HOSPITAL CORPORATION, MASSACHUSETTS

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:BONVENTRE, JOSEPH V.;ICHIMURA, TAKAHARU;REEL/FRAME:018354/0528;SIGNING DATES FROM 19990413 TO 19990429

Owner name: BIOGEN, INC., MASSACHUSETTS

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:SANICOLA-NADEL, MICHELE;HESSION, CATHERINE;WEI, HENRY;AND OTHERS;REEL/FRAME:018354/0349;SIGNING DATES FROM 19990325 TO 19990330

AS Assignment

Owner name: BIOGEN IDEC MA INC., MASSACHUSETTS

Free format text: CHANGE OF NAME;ASSIGNOR:BIOGEN, INC.;REEL/FRAME:018376/0314

Effective date: 20031113

STCB Information on status: application discontinuation

Free format text: ABANDONED -- FAILURE TO RESPOND TO AN OFFICE ACTION

AS Assignment

Owner name: NATIONAL INSTITUTES OF HEALTH - DIRECTOR DEITR, MARYLAND

Free format text: CONFIRMATORY LICENSE;ASSIGNOR:MASSACHUSETTS GENERAL HOSPITAL;REEL/FRAME:062737/0711

Effective date: 20230216