AU779534B2 - Interleukin-1 receptor antagonist-related molecules and uses thereof - Google Patents

Interleukin-1 receptor antagonist-related molecules and uses thereof Download PDF

Info

Publication number
AU779534B2
AU779534B2 AU19450/01A AU1945001A AU779534B2 AU 779534 B2 AU779534 B2 AU 779534B2 AU 19450/01 A AU19450/01 A AU 19450/01A AU 1945001 A AU1945001 A AU 1945001A AU 779534 B2 AU779534 B2 AU 779534B2
Authority
AU
Australia
Prior art keywords
seq
polypeptide
lra
residue
amino acid
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.)
Ceased
Application number
AU19450/01A
Other versions
AU1945001A (en
Inventor
Michael Brian Bass
Roger Craveiro
Jennifer Giles
Sharon X. Mu
Christiaan M. Saris
Min Xia
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.)
Amgen Inc
Original Assignee
Amgen Inc
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Amgen Inc filed Critical Amgen Inc
Priority claimed from PCT/US2000/032940 external-priority patent/WO2001042304A1/en
Publication of AU1945001A publication Critical patent/AU1945001A/en
Application granted granted Critical
Publication of AU779534B2 publication Critical patent/AU779534B2/en
Anticipated expiration legal-status Critical
Ceased legal-status Critical Current

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/52Cytokines; Lymphokines; Interferons
    • C07K14/54Interleukins [IL]
    • 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
    • C07K16/24Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against cytokines, lymphokines or interferons
    • C07K16/244Interleukins [IL]
    • 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
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • 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
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2799/00Uses of viruses
    • C12N2799/02Uses of viruses as vector
    • C12N2799/021Uses of viruses as vector for the expression of a heterologous nucleic acid

Landscapes

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

Description

WO 01/42304 PCT/US00/32940 INTERLEUKIN-1 RECEPTOR ANTAGONIST-RELATED MOLECULES AND USES THEREOF Field of the Invention The present invention relates to novel lnterleukin-1 Receptor Antagonist- Related (IL-lra-R) polypeptides and nucleic acid molecules encoding the same.
The invention also relates to selective binding agents, vectors, host cells, and methods for producing IL-lra-R polypeptides. The invention further relates to pharmaceutical compositions and methods for the diagnosis, treatment, amelioration, and/or prevention of diseases, disorders, and conditions associated with IL-lra-R polypeptides.
Background of the Invention Technical advances in the identification, cloning, expression, and manipulation of nucleic acid molecules and the deciphering of the human genome have greatly accelerated the discovery of novel therapeutics. Rapid nucleic acid sequencing techniques can now generate sequence information at unprecedented rates and, coupled with computational analyses, allow the assembly of overlapping sequences into partial and entire genomes and the identification of polypeptide-encoding-regions. A comparison of a predicted amino acid sequence against a database compilation of known amino acid sequences allows one to determine the extent of homology to previously identified sequences and/or structural landmarks. The cloning and expression of a polypeptide-encoding region of a nucleic acid molecule provides a polypeptide product for structural and functional analyses. The manipulation of nucleic acid molecules and encoded polypeptides may confer advantageous properties on a product for use as a therapeutic.
In spite of the significant technical advances in genome research over the past decade, the potential for the development of novel therapeutics based on the human genome is still largely unrealized. Many genes encoding potentially beneficial polypeptide therapeutics or those encoding polypeptides, which may act WO 01/42304 PCT/US00/32940 as "targets" for therapeutic molecules, have still not been identified.
Accordingly, it is an object of the invention to identify novel polypeptides, and nucleic acid molecules encoding the same, which have diagnostic or therapeutic benefit.
One of the most potent inflammatory cytokines yet discovered is interleukin-1 IL-1 is thought to be involved in many diseases and medical conditions. It is produced (though not exclusively) by cells of the macrophage/monocyte lineage, and may be produced in two forms: IL-lalpha (IL-la) and IL-lbeta (IL-13). Interleukin-l receptor antagonist (IL-lra) is a human protein that acts as a natural inhibitor of interleukin-1.
Summary of the Invention The present invention relates to novel IL-lra-R nucleic acid molecules and encoded polypeptides.
The invention provides for an isolated nucleic acid molecule comprising a nucleotide sequence selected from the group consisting of: the nucleotide sequence as set forth in any of SEQ ID NO: 1, SEQ ID NO: 3, SEQ ID NO: 5, or SEQ ID NO: the nucleotide sequence of the DNA insert in ATCC Deposit No.
PTA-1423; a nucleotide sequence encoding the polypeptide as set forth in any of SEQ ID NO: 2, SEQ ID NO: 4, SEQ ID NO: 6, or SEQ ID NO: 36; a nucleotide sequence which hybridizes under moderately or highly stringent conditions to the complement of any of and a nucleotide sequence complementary to any of The invention also provides for an isolated nucleic acid molecule comprising a nucleotide sequence selected from the group consisting of: a nucleotide sequence encoding a polypeptide which is at least about 70 percent identical to the polypeptide as set forth in any of SEQ ID NO: 2, 2 WO 01/42304 PCT/US00/32940 SEQ ID NO: 4, SEQ ID NO: 6, or SEQ ID NO: 36, wherein the encoded polypeptide has an activity of the polypeptide set forth in any of SEQ ID NO: 2, SEQ ID NO: 4, SEQ ID NO: 6, or SEQ ID NO: 36; a nucleotide sequence encoding an allelic variant or splice variant of the nucleotide sequence as set forth in any of SEQ ID NO: 1, SEQ ID NO: 3, SEQ ID NO: 5, or SEQ ID NO: 35, the nucleotide sequence of the DNA insert in ATCC Deposit No. PTA-1423, or a region of the nucleotide sequence of any of SEQ ID NO: 1, SEQ ID NO: 3, SEQ ID NO: 5, or SEQ ID NO: 35, the DNA insert in ATCC Deposit No. PTA-1423, or encoding a polypeptide fragment of at least about amino acid residues, wherein the polypeptide fragment has an activity of the encoded polypeptide as set forth in any of SEQ ID NO: 2, SEQ ID NO: 4, SEQ ID NO: 6, or SEQ ID NO: 36, or is antigenic; a region of the nucleotide sequence of any of SEQ ID NO: 1, SEQ ID NO: 3, SEQ ID NO: 5, or SEQ ID NO: 35, the DNA insert in ATCC Deposit No. PTA-1423, or any of comprising a fragment of at least about 16 nucleotides; a nucleotide sequence which hybridizes under moderately or highly stringent conditions to the complement of any of(a) and a nucleotide sequence complementary to any of The invention further provides for an isolated nucleic acid molecule comprising a nucleotide sequence selected from the group consisting of: a nucleotide sequence encoding a polypeptide as set forth in any of SEQ ID NO: 2, SEQ ID NO: 4, SEQ ID NO: 6, or SEQ ID NO: 36 with at least one conservative amino acid substitution, wherein the encoded polypeptide has an activity of the polypeptide set forth in any of SEQ ID NO: 2, SEQ ID NO: 4, SEQ ID NO: 6, or SEQ ID NO: 36; a nucleotide sequence encoding a polypeptide as set forth in any of SEQ ID NO: 2, SEQ ID NO: 4, SEQ ID NO: 6, or SEQ ID NO: 36 with at least one amino acid insertion, wherein the encoded polypeptide has an activity of the WO 01/42304 PCT/US00/32940 polypeptide set forth in any of SEQ ID NO: 2, SEQ ID NO: 4, SEQ ID NO: 6, or SEQ ID NO: 36; a nucleotide sequence encoding a polypeptide as set forth in any of SEQ ID NO: 2, SEQ ID NO: 4, SEQ ID NO: 6, or SEQ ID NO: 36 with at least one amino acid deletion, wherein the encoded polypeptide has an activity of the polypeptide set forth in any of SEQ ID NO: 2, SEQ ID NO: 4, SEQ ID NO: 6, or SEQ ID NO: 36; a nucleotide sequence encoding a polypeptide as set forth in any of SEQ ID NO: 2, SEQ ID NO: 4, SEQ ID NO: 6, or SEQ ID NO: 36 which has a Cand/or N- terminal truncation, wherein the encoded polypeptide has an activity of the polypeptide set forth in any of SEQ ID NO: 2, SEQ ID NO: 4, SEQ ID NO: 6, or SEQ ID NO: 36; a nucleotide sequence encoding a polypeptide as set forth in any of SEQ ID NO: 2, SEQ ID NO: 4, SEQ ID NO: 6, or SEQ ID NO: 36 with at least one modification selected from the group consisting of amino acid substitutions, amino acid insertions, amino acid deletions, C-terminal truncation, and N-terminal truncation, wherein the encoded polypeptide has an activity of the polypeptide set forth in any of SEQ ID NO: 2, SEQ ID NO: 4, SEQ ID NO: 6, or SEQ ID NO: 36; a nucleotide sequence of any of comprising a fragment of at least about 16 nucleotides; a nuclcotide sequence which hybridizes under moderately or highly stringent conditions to the complement of any of(a) and a nucleotide sequence complementary to any of(a) The present invention provides for an isolated polypeptide comprising an amino acid sequence selected from the group consisting of: the amino acid sequence as set forth in any of SEQ ID NO: 2, SEQ ID NO: 4, SEQ ID NO: 6, or SEQ ID NO: 36; and the amino acid sequence encoded by the DNA insert in ATCC Deposit No. PTA-1423.
WO 01/42304 PCT/US00/32940 The invention also provides for an isolated polypeptide comprising the amino acid sequence selected from the group consisting of: an amino acid sequence for an ortholog of any of SEQ ID NO: 2, SEQ ID NO: 4, SEQ ID NO: 6, or SEQ ID NO: 36; an amino acid sequence which is at least about 70 percent identical to the amino acid sequence of any of SEQ ID NO: 2, SEQ ID NO: 4, SEQ ID NO: 6, or SEQ ID NO: 36, wherein the polypeptide has an activity of the polypeptide set forth in any of SEQ ID NO: 2, SEQ ID NO: 4, SEQ ID NO: 6, or SEQ ID NO: 36; a fragment of the amino acid sequence set forth in any of SEQ ID NO: 2, SEQ ID NO: 4, SEQ ID NO: 6, or SEQ ID NO: 36 comprising at least about 25 amino acid residues, wherein the fragment has an activity of the polypeptide set forth in any of SEQ ID NO: 2, SEQ ID NO: 4, SEQ ID NO: 6, or SEQ ID NO: 36, or is antigenic; and an amino acid sequence for an allelic variant or splice variant of the amino acid sequence as set forth in any of SEQ ID NO: 2, SEQ ID NO: 4, SEQ ID NO: 6, or SEQ ID NO: 36, the amino acid sequence encoded by the DNA insert in ATCC Deposit No. PTA-1423, or The invention further provides for an isolated polypeptide comprising the amino acid sequence selected from the group consisting of: the amino acid sequence as set forth in any of SEQ ID NO: 2, SEQ ID NO: 4, SEQ ID NO: 6, or SEQ ID NO: 36 with at least one conservative amino acid substitution, wherein the polypeptide has an activity of the polypeptide set forth in any of SEQ ID NO: 2, SEQ ID NO: 4, SEQ ID NO: 6, or SEQ ID NO: 36; the amino acid sequence as set forth in any of SEQ ID NO: 2, SEQ ID NO: 4, SEQ ID NO: 6, or SEQ ID NO: 36 with at least one amino acid insertion, wherein the polypeptide has an activity of the polypeptide set forth in any of SEQ ID NO: 2, SEQ ID NO: 4, SEQ ID NO: 6, or SEQ ID NO: 36; the amino acid sequence as set forth in any of SEQ ID NO: 2, SEQ ID NO: 4, SEQ ID NO: 6, or SEQ ID NO: 36 with at least one amino acid WO 01/42304 PCT/US0O/32940 deletion, wherein the polypeptide has an activity of the polypeptide set forth in any of SEQ ID NO: 2, SEQ ID NO: 4, SEQ ID NO: 6, or SEQ ID NO: 36; the amino acid sequence as set forth in any of SEQ ID NO: 2, SEQ ID NO: 4, SEQ ID NO: 6, or SEQ ID NO: 36 which has a C- and/or N- terminal truncation, wherein the polypeptide has an activity of the polypeptide set forth in any of SEQ ID NO: 2, SEQ ID NO: 4, SEQ ID NO: 6, or SEQ ID NO: 36; and the amino acid sequence as set forth in any of SEQ ID NO: 2, SEQ ID NO: 4, SEQ ID NO: 6, or SEQ ID NO: 36 with at least one modification selected from the group consisting of amino acid substitutions, amino acid insertions, amino acid deletions, C-terminal truncation, and N-terminal truncation, wherein the polypeptide has an activity of the polypeptide set forth in any of SEQ ID NO: 2, SEQ ID NO: 4, SEQ ID NO: 6, or SEQ ID NO: 36.
The invention still further provides for an isolated polypeptide comprising the amino acid sequence as set forth in either SEQ ID NO: 1 or SEQ ID NO: 3 with at least one amino acid substitution selected from the group consisting of: arginine at position 2; alanine, lysine, or arginine at position 3; serine at position 7; lysine at position 8; alanine, cysteine, lysine, threonine, or serine at position 9; cysteine or phenylalanine at position 10; arginine or trptophan at position 13; serine at position 15; arginine at position 18; serine or threonine at position 19; threonine at position 21; serine at position 23; arginine at position 34; tyrosine, serine, or arginine at position 37; lysine, arginine, threonine, or serine at position 38; threonine at position 41; serine, phenylalanine, or alanine at position 43; alanine at position 44; serine or lysine at position 48; alanine, threonine, or phenylalanine at position 52; serine at position 53; serine at position 54; alanine or tyrosine at position 58; lysine at position 65; phenylalanine at position 66; tyrosine at position 67; serine, tyrosine, or phenylalanine at position 69; lysine or serine at position 73; threonine or arginine at position 78; serine or alanine at position 90; alanine at position 91; serine at position 96; lysine or arginine at position 97; lysine or serine at position 98; alanine at position 100; tyrosine at position 102; arginine or alanine at position 104; lysine at position 105; threonine 6 WO 01/42304 PCT/US00/32940 at position 106; arginine at position 108; lysine, threonine, or trptophan at position 109; threonine or serine at position 110; serine at position 111; serine at position 114; serine at position 116; phenylalanine, cysteine, or tyrosine at position 117; tyrosine at position 121; serine or alaninc at position 123; cysteine, serine, or threonine at position 126; serine at position 136; phenylalanine or arginine at position 138; threonine, arginine, or alanine at position 141; lysine or tyrosine at position 142; trptophan or threonine at position 143; alanine at position 145; threonine or serine at position 147; cysteine at position 151; and serine, cysteine, or phenylalanine at position 152; wherein the polypeptide has an activity of the polypeptide set forth in any of SEQ ID NO: 2, SEQ ID NO: 4, or SEQ ID NO: 6.
The invention still further provides for an isolated polypeptide comprising the amino acid sequence as set forth in SEQ ID NO: 5 with at least one amino acid substitution selected from the group consisting of: arginine at position 21; alanine, lysine, or arginine at position 22; serine at position 26; lysine at position 27; alanine, cysteine, lysine, threonine, or serine at position 28; cysteine or phenylalanine at position 29; arginine or trptophan at position 32; serine at position 34; arginine at position 37; serine or threonine at position 38; threonine at position 40; serine at position 42; arginine at position 53; tyrosine, serine, or arginine at position 56; lysine, arginine, threonine, or serine at position 57; threonine at position 60; serine, phenylalanine, or alanine at position 62; alanine at position 63; serine or lysine at position 67; alanine, threonine, or phenylalanine at position 71; serine at position 72; serine at position 73; alanine or tyrosine at position 77; lysine at position 84; phenylalanine at position 85; tyrosine at position 86; serine, tyrosine, or phenylalanine at position 88; lysine or serine at position 92; threonine or arginine at position 97; serine or alanine at position 109; alanine at position 110; serine at position 115; lysine or arginine at position 116; lysine or serine at position 117; alanine at position 119; tyrosine at position 121; arginine or alanine at position 123; lysine at position 124; thrconine at position 125; arginine at position 127; lysine, threonine, or trptophan at position 128; threonine or serine at position 129; serine at position 130; serine at position 133; 7 WO 01/42304 PCT/US00/32940 serine at position 135; phenylalanine, cysteine, or tyrosine at position 136; tyrosine at position 140; serine or alanine at position 142; cysteine, serine, or threonine at position 145; serine at position 155; phenylalanine or arginine at position 157; threonine, arginine, or alanine at position 160; lysine or tyrosine at position 161; trptophan or threonine at position 162; alanine at position 164; threonine or serine at position 166; cysteine at position 170; and serine, cysteine, or phenylalanine at position 171; wherein the polypeptide has an activity of the polypeptide set forth in any of SEQ ID NO: 2, SEQ ID NO: 4, or SEQ ID NO: 6.
Also provided are fusion polypeptides comprising IL-Ira-R amino acid sequences.
The present invention also provides for an expression vector comprising the isolated nucleic acid molecules as set forth herein, recombinant host cells comprising the recombinant nucleic acid molecules as set forth herein, and a method of producing an IL-lra-R polypeptide comprising culturing the host cells and optionally isolating the polypeptide so produced.
A transgenic non-human animal comprising a nucleic acid molecule encoding an IL-I ra-R polypeptide is also encompassed by the invention. The IL- Ira-R nucleic acid molecules are introduced into the animal in a manner that allows expression and increased levels of an IL-lra-R polypeptide, which may include increased circulating levels. Alternatively, the IL-lra-R nucleic acid molecules are introduced into the animal in a manner that prevents expression of endogenous IL-lra-R polypeptide generates a transgenic animal possessing an IL-lra-R polypeptide gene knockout). The transgenic non-human animal is preferably a mammal, and more preferably a rodent, such as a rat or a mouse.
Also provided are derivatives of the IL-lra-R polypeptides of the present invention.
Additionally provided are selective binding agents such as antibodies and peptides capable of specifically binding the IL-lra-R polypeptides of the invention. Such antibodies and peptides may be agonistic or antagonistic.
Pharmaceutical compositions comprising the nucleotides, polypeptides, or 8 WO 01/42304 PCT/US00/32940 selective binding agents of the invention and one or more pharmaceutically acceptable formulation agents are also encompassed by the invention. The pharmaceutical compositions are used to provide therapeutically effective amounts of the nucleotides or polypeptides of the present invention. The invention is also directed to methods of using the polypeptides, nucleic acid molecules, and selective binding agents.
The IL-lra-R polypeptides and nucleic acid molecules of the present invention may be used to treat, prevent, ameliorate, and/or detect diseases and disorders, including those recited herein.
The present invention also provides a method of assaying test molecules to identify a test molecule that binds to an IL-lra-R polypeptide. The method comprises contacting an IL-lra-R polypeptide with a test molecule to determine the extent of binding of the test molecule to the polypeptide. The method further comprises determining whether such test molecules are agonists or antagonists of an IL-lra-R polypeptide. The present invention further provides a method of testing the impact of molecules on the expression of IL-lra-R polypeptide or on the activity of IL-Ira-R polypeptide.
Methods of regulating expression and modulating increasing or decreasing) levels of an IL-lra-R polypeptide are also encompassed by the invention. One method comprises administering to an animal a nucleic acid molecule encoding an IL-lra-R polypeptide. In another method, a nucleic acid molecule comprising elements that regulate or modulate the expression of an ILlra-R polypeptide may be administered. Examples of these methods include gene therapy, cell therapy, and anti-sense therapy as further described herein.
In another aspect of the present invention, the IL-lra-R polypeptides may be used for identifying receptors thereof ("IL-lra-R polypeptide receptors").
Various forms of "expression cloning" have been extensively used to clone receptors for protein ligands. See, Simonsen and Lodish, 1994, Trends Pharmacol. Sci. 15:437-41 and Tartaglia et al., 1995, Cell 83:1263-71. The isolation of an IL-lra-R polypeptide receptor is useful for identifying or developing novel agonists and antagonists of the IL-lra-R polypeptide signaling 9 WO 01/42304 PCT/US00/32940 pathway. Such agonists and antagonists include soluble IL-lra-R polypeptide receptors, anti-IL-lra-R polypeptide receptor-selective binding agents (such as antibodies and derivatives thereof), small molecules, and antisense oligonucleotides, any of which can be used for treating one or more disease or disorder, including those disclosed herein.
Brief Description of the Figures Figures IA-1B illustrate the nucleotide sequence of the human IL-lra-R gene (SEQ ID NO: 1) and the deduced amino acid sequence of human IL-lra-R polypeptide (SEQ ID NO: 2); Figures 2A-2B illustrate the nucleotide sequence of a human IL-lra-R gene variant (SEQ ID NO: 3) and the deduced amino acid sequence of the human ILlra-R polypeptide (SEQ ID NO: 4) encoded by this vaiant; Figure 3 illustrates the nucleotide sequence of a human IL-Ira-R splice variant (SEQ ID NO: 5) and the deduced amino acid sequence of the human IL-lra-R polypeptide (SEQ ID NO: 6) encoded by this splice variant; Figures 4A-4B illustrate the amino acid sequence alignment of human IL- la (ILlalpha; SEQ ID NO: human IL-1p (IL-l_beta; SEQ ID NO: human IL-I receptor antagonist (IL-1RA; SEQ ID NO: human IL-16 (IL-l_delta; SEQ ID NO: 10), human IL-lra-R polypeptide (IL-lra-R; SEQ ID NO: human Tango- 77 (Tango-77; SEQ ID NO: 11), human Zilla4 (Zilla4; SEQ ID NO: 12), human IL-l (IL-l_zeta; SEQ ID NO: 13), human IL-1 receptor antagonist p (IL- IRAbeta; SEQ ID NO: 14), human SPOIL II (Spoil II; SEQ ID NO: 15), human IL-le (IL-l_epsilon; SEQ ID NO: 16), and human IL-l (IL-l_eta; SEQ ID NO: 17); Figure 5 schematically illustrates the phylogenetic relationship of the IL-Ira gene family.
WO 01/42304 PCT/US00/32940 Figure 6 schematically illustrates the relationship between human IL-Ira-R polypeptide (Mature CS329), the sequence variant of human IL-lra-R polypeptide (Mature CS329 Variant protein), and the splice variant of human IL-lra-R polypeptide (Omega 329 protein); Figure 7 illustrates the nucleotide sequence of the murine IL-lra-R gene (SEQ ID NO: 35) and the deduced amino acid sequence of murine IL-lra-R polypeptide (SEQ ID NO: 36); Figure 8 illustrates the amino acid sequence alignment of human IL-lra-R polypeptide (hulL-lra-R; SEQ ID NO: 2) and murine IL-lra-R polypeptide (mulL-lra-R; SEQ ID NO: 36); Figures 9A-9I illustrate the genomic nucleotide sequence for the murine IL-Ira-R gene (SEQ ID NO: 37). The locations of the coding portions of exons 1-4 are indicated (underline); Figures 10A-10C illustrate the expression of murine (Figure 10A) and human (Figures 10B-10C) IL- ra-R mRNA as detected by Northern blot analysis; Figures 1 IA-Il B illustrate the results of Western blot analysis using an anti-IL- I ra-R antibody; Figure 12 illustrates the results of FACS analysis of spleen and bone marrow cells recovered from lethally irradiated recipient mice transplanted with murine bone marrow cells transduced with a retroviral vector containing the IL-lra-R gene.
The percentage of cells within the lymphoid compartment is shown for cells subjected to standard FACS analysis using the indicated cell surface markers; Figures 13A-13B illustrate the results of colony assays performed on spleen and 11 WO 01/42304 PCT/US00/32940 bone marrow cells recovered from lethally irradiated recipient mice transplanted with murine bone marrow cells transduced with a retroviral vector containing the IL-lra-R gene. Bone marrow and spleen cells from transduced and control mice were cultured under standard colony assay conditions and colonies were counted on day 14; Figure 14 illustrates the production ofy-interferon in response to IL-12 treatment in spleen cells recovered from lethally irradiated recipient mice transplanted with murine bone marrow cells transduced with a retroviral vector containing the IL- Ira-R gene. A sample of conditioned media was removed from cultures (2 x 106 cells/well/ml) grown in the presence of IL-12 (1 ng/ml) for 48 hours and IFN-y was quantitated by ELISA; Figure 15 illustrates the production of y-interferon in response to IL-12 and IL-18 treatment in spleen cells recovered from lethally irradiated recipient mice transplanted with murine bone marrow cells transduced with a retroviral vector containing the IL-Ira-R gene. A sample of conditioned media was removed from cultures (2 x 106 cells/well/ml) grown in the presence of IL-12 (1 ng/ml) and IL- 18 (10 ng/ml) for 48 hours and IFN-y was quantitated by ELISA.
Detailed Description of the Invention The section headings used herein are for organizational purposes only and are not to be construed as limiting the subject matter described. All references cited in this application are expressly incorporated by reference herein.
Definitions The terms "IL-lra-R gene" or "IL-lra-R nucleic acid molecule" or "IL- Ira-R polynucleotide" refer to a nucleic acid molecule comprising or consisting of a nucleotide sequence as set forth in any of SEQ ID NO: 1, SEQ ID NO: 3, SEQ ID NO: 5, or SEQ ID NO: 35, a nucleotide sequence encoding the polypeptide as set forth in any of SEQ ID NO: 2, SEQ ID NO: 4, SEQ ID NO: 6, or SEQ ID NO: 12 WO 01/42304 PCT/US00/32940 36, a nucleotide sequence of the DNA insert in ATCC Deposit No. PTA-1423, and nucleic acid molecules as defined herein.
The term "IL-lra-R polypeptide allelic variant" refers to one of several possible naturally occurring alternate forms of a gene occupying a given locus on a chromosome of an organism or a population of organisms.
The term "IL-lra-R polypeptide splice variant" refers to a nucleic acid molecule, usually RNA, which is generated by alternative processing of intron sequences in an RNA transcript of IL-lra-R polypeptide amino acid sequence as set forth in any of SEQ ID NO: 2, SEQ ID NO: 4, SEQ ID NO: 6, or SEQ ID NO: 36.
The term "isolated nucleic acid molecule" refers to a nucleic acid molecule of the invention that has been separated from at least about 50 percent of proteins, lipids, carbohydrates, or other materials with which it is naturally found when total nucleic acid is isolated from the source cells, is not linked to all or a portion of a polynucleotide to which the "isolated nucleic acid molecule" is linked in nature, is operably linked to a polynucleotide which it is not linked to in nature, or does not occur in nature as part of a larger polynucleotide sequence.
Preferably, the isolated nucleic acid molecule of the present invention is substantially free from any other contaminating nucleic acid molecule(s) or other contaminants that are found in its natural environment that would interfere with its use in polypeptide production or its therapeutic, diagnostic, prophylactic or research use.
The term "nucleic acid sequence" or "nucleic acid molecule" refers to a DNA or RNA sequence. The term encompasses molecules formed from any of the known base analogs of DNA and RNA such as, but not limited to 4acetylcytosine, 8-hydroxy-N6-methyladenosine, aziridinyl-cytosine, pseudoisocytosine, 5-(carboxyhydroxylmethyl) uracil, 5-fluorouracil, bromouracil, 5-carboxymethylaminomethyl-2-thiouracil, methylaminomethyluracil, dihydrouracil, inosine, N6-iso-pentenyladenine, 1methyladenine, I-methylpseudouracil, I-methylguanine, I-methylinosine, 2,2dimethyl-guanine, 2-methyladenine, 2-methylguanine, 3-methylcytosine, 13 WO 01/42304 PCT/US00/32940 methylcytosine, N6-methyladenine, 7-methylguanine, methylaminomethyluracil, 5-methoxyamino-methyl-2-thiouracil, beta-Dmannosylqueosine, 5' -methoxycarbonyl-methyluracil, 5-methoxyuracil, 2methylthio-N6-isopentenyladenine, uracil-5-oxyacetic acid methylester, oxyacetic acid, oxybutoxosine, pseudouracil, queosine, 2-thiocytosine, 2-thiouracil, 2-thiouracil, 4-thiouracil, 5-methyluracil, N-uracil-5-oxyacetic acid methylester, uracil-5-oxyacetic acid, pseudouracil, queosine, 2-thiocytosine, and 2,6-diaminopurine.
The term "vector" is used to refer to any molecule nucleic acid, plasmid, or virus) used to transfer coding information to a host cell.
The term "expression vector" refers to a vector that is suitable for transformation of a host cell and contains nucleic acid sequences that direct and/or control the expression of inserted heterologous nucleic acid sequences.
Expression includes, but is not limited to, processes such as transcription, translation, and RNA splicing, ifintrons are present.
The term "operably linked" is used herein to refer to an arrangement of flanking sequences wherein the flanking sequences so described are configured or assembled so as to perform their usual function. Thus, a flanking sequence operably linked to a coding sequence may be capable of effecting the replication, transcription and/or translation of the coding sequence. For example, a coding sequence is operably linked to a promoter when the promoter is capable of directing transcription of that coding sequence. A flanking sequence need not be contiguous with the coding sequence, so long as it functions correctly. Thus, for example, intervening untranslated yet transcribed sequences can be present between a promoter sequence and the coding sequence and the promoter sequence can still be considered "operably linked" to the coding sequence.
The term "host cell" is used to refer to a cell which has been transformed, or is capable of being transformed with a nucleic acid sequence and then of expressing a selected gene of interest The term includes the progeny of the parent cell, whether or not the progeny is identical in morphology or in genetic make-up to the original parent, so long as the selected gene is present.
WO 01/42304 PCT/US00/32940 The term "IL-lra-R polypeptide" refers to a polypeptide comprising the amino acid sequence of any of SEQ ID NO: 2, SEQ ID NO: 4, SEQ ID NO: 6, or SEQ ID NO: 36 and related polypeptides. Related polypeptides include IL-lra-R polypeptide fragments, IL-lra-R polypeptide orthologs, IL-lra-R polypeptide variants, and IL-lra-R polypeptide derivatives, which possess at least one activity of the polypeptide as set forth in any of SEQ ID NO: 2, SEQ ID NO: 4, SEQ ID NO: 6, or SEQ ID NO: 36. IL-Ira-R polypeptides may be mature polypeptides, as defined herein, and may or may not have an amino-terminal methionine residue, depending on the method by which they are prepared.
The term "IL-lra-R polypeptide fragment" refers to a polypeptide that comprises a truncation at the amino-terminus (with or without a leader sequence) and/or a truncation at the carboxyl-terminus of the polypeptide as set forth in any of SEQ ID NO: 2, SEQ ID NO: 4, SEQ ID NO: 6, or SEQ ID NO: 36. The term "IL-lra-R polypeptide fragment" also refers to amino-terminal and/or carboxylterminal truncations of IL-lra-R polypeptide orthologs, IL-lra-R polypeptide derivatives, or IL-lra-R polypeptide variants, or to amino-terminal and/or carboxyl-terminal truncations of the polypeptides encoded by IL-lra-R polypeptide allelic variants or IL-lra-R polypeptide splice variants. IL-lra-R polypeptide fragments may result from alternative RNA splicing or from in vivo protease activity. Membrane-bound forms of an IL-lra-R polypeptide are also contemplated by the present invention. In preferred embodiments, truncations and/or deletions comprise about 10 amino acids, or about 20 amino acids, or about amino acids, or about 75 amino acids, or about 100 amino acids, or more than about 100 amino acids. The polypeptide fragments so produced will comprise about 25 contiguous amino acids, or about 50 amino acids, or about 75 amino acids, or about 100 amino acids, or about 125 amino acids. Such IL-lra-R polypeptide fragments may optionally comprise an amino-terminal methionine residue. It will be appreciated that such fragments can be used, for example, to generate antibodies to IL-lra-R polypeptides.
The term "IL-lra-R polypeptide ortholog" refers to a polypeptide from another species that corresponds to IL-Ira-R polypeptide amino acid sequence as WO 01/42304 PCT/US00/32940 set forth in any of SEQ ID NO: 2, SEQ ID NO: 4, SEQ ID NO: 6, or SEQ ID NO: 36. For example, mouse and human IL-lra-R polypeptides are considered orthologs of each other.
The term "IL-Ira-R polypeptide variants" refers to IL-lra-R polypeptides comprising amino acid sequences having one or more amino acid sequence substitutions, deletions (such as internal deletions and/or IL-lra-R polypeptide fragments), and/or additions (such as internal additions and/or IL-lra-R fusion polypeptides) as compared to the IL-lra-R polypeptide amino acid sequence set forth in any of SEQ ID NO: 2, SEQ ID NO: 4, SEQ ID NO: 6, or SEQ ID NO: 36 (with or without a leader sequence). Variants may be naturally occurring IL-lra-R polypeptide allelic variants, IL-lra-R polypeptide orthologs, and IL-Ira- R polypeptide splice variants) or artificially constructed. Such IL-lra-R polypeptide variants may be prepared from the corresponding nucleic acid molecules having a DNA sequence that varies accordingly from the DNA sequence as set forth in any of SEQ ID NO: 1, SEQ ID NO: 3, SEQ ID NO: 5, or SEQ ID NO: 35. In preferred embodiments, the variants have from 1 to 3, or from 1 to 5, or from 1 to 10, or from 1 to 15, or from 1 to 20, or from 1 to 25, or from I to 50, or from 1 to 75, or from 1 to 100, or more than 100 amino acid substitutions, insertions, additions and/or deletions, wherein the substitutions may be conservative, or non-conservative, or any combination thereof.
The term "IL-lra-R polypeptide derivatives" refers to the polypeptide as set forth in any of SEQ ID NO: 2, SEQ ID NO: 4, SEQ ID NO: 6, or SEQ ID NO: 36, IL-lra-R polypeptide fragments, IL-lra-R polypeptide orthologs, or IL-lra-R polypeptide variants, as defined herein, that have been chemically modified. The term "IL-lra-R polypeptide derivatives" also refers to the polypeptides encoded by IL-lra-R polypeptide allelic variants or IL-lra-R polypeptide splice variants, as defined herein, that have been chemically modified.
The term "mature IL-lra-R polypeptide" refers to an IL-lra-R polypeptide lacking a leader sequence. A mature IL-lra-R polypeptide may also include other modifications such as proteolytic processing of the amino-terminus (with or without a leader sequence) and/or the carboxyl-terminus, cleavage of a smaller 16 WO 01/42304 PCT/US00/32940 polypeptide from a larger precursor, N-linked and/or O-linked glycosylation, and the like.
The term "IL-lra-R fusion polypeptide" refers to a fusion of one or more amino acids (such as a heterologous protein or peptide) at the amino- or carboxylterminus of the polypeptide as set forth in any of SEQ ID NO: 2, SEQ ID NO: 4, SEQ ID NO: 6, or SEQ ID NO: 36, IL-lra-R polypeptide fragments, IL-lra-R polypeptide orthologs, IL-lra-R polypeptide variants, or IL-lra-R derivatives, as defined herein. The term "IL-lra-R fusion polypeptide" also refers to a fusion of one or more amino acids at the amino- or carboxyl-terminus of the polypeptide encoded by IL-lra-R polypeptide allelic variants or IL-lra-R polypeptide splice variants, as defined herein.
The term "biologically active IL-lra-R polypeptides" refers to IL-lra-R polypeptides having at least one activity characteristic of the polypeptide comprising the amino acid sequence of any of SEQ ID NO: 2, SEQ ID NO: 4, SEQ ID NO: 6, or SEQ ID NO: 36. In addition, an IL-lra-R polypeptide may be active as an immunogen; that is, the IL-lra-R polypeptide contains at least one epitope to which antibodies may be raised.
The term "isolated polypeptide" refers to a polypeptide of the present invention that has been separated from at least about 50 percent of polynucleotides, lipids, carbohydrates, or other materials with which it is naturally found when isolated from the source cell is not linked (by covalent or noncovalent interaction) to all or a portion of a polypeptide to which the "isolated polypeptide" is linked in nature, is operably linked (by covalent or noncovalent interaction) to a polypeptide with which it is not linked in nature, or does not occur in nature. Preferably, the isolated polypeptide is substantially free from any other contaminating polypeptides or other contaminants that are found in its natural environment that would interfere with its therapeutic, diagnostic, prophylactic or research use.
The term "identity," as known in the art, refers to a relationship between the sequences of two or more polypeptide molecules or two or more nucleic acid molecules, as determined by comparing the sequences. In the art, "identity" also 17 WO 01/42304 PCT/US00/32940 means the degree of sequence relatedness between nucleic acid molecules or polypeptides, as the case may be, as determined by the match between strings of two or more nucleotide or two or more amino acid sequences. "Identity" measures the percent of identical matches between the smaller of two or more sequences with gap alignments (if any) addressed by a particular mathematical model or computer program "algorithms").
The term "similarity" is a related concept, but in contrast to "identity," "similarity" refers to a measure of relatedness which includes both identical matches and conservative substitution matches. If two polypeptide sequences have, for example, 10/20 identical amino acids, and the remainder are all nonconservative substitutions, then the percent identity and similarity would both be If in the same example, there are five more positions where there are conservative substitutions, then the percent identity remains 50%, but the percent similarity would be 75% (15/20). Therefore, in cases where there are conservative substitutions, the percent similarity between two polypeptides will be higher than the percent identity between those two polypeptides.
The term "naturally occurring" or "native" when used in connection with biological materials such as nucleic acid molecules, polypeptides, host cells, and the like, refers to materials which are found in nature and are not manipulated by man. Similarly, "non-naturally occurring" or "non-native" as used herein refers to a material that is not found in nature or that has been structurally modified or synthesized by man.
The terms "effective amount" and "therapeutically effective amount" each refer to the amount of an IL-lra-R polypeptide or IL-lra-R nucleic acid molecule used to support an observable level of one or more biological activities of the IL- Ira-R polypeptides as set forth herein.
The term "pharmaceutically acceptable carrier" or "physiologically acceptable carrier" as used herein refers to one or more formulation materials suitable for accomplishing or enhancing the delivery of the IL-lra-R polypeptide, IL-lra-R nucleic acid molecule, or IL-lra-R selective binding agent as a pharmaceutical composition.
WO 01/42304 PCT/US00/32940 The term "antigen" refers to a molecule or a portion of a molecule capable of being bound by a selective binding agent, such as an antibody, and additionally capable of being used in an animal to produce antibodies capable of binding to an epitope of that antigen. An antigen may have one or more epitopes.
The term "selective binding agent" refers to a molecule or molecules having specificity for an IL-lra-R polypeptide. As used herein, the terms, "specific" and "specificity" refer to the ability of the selective binding agents to bind to human IL-lra-R polypeptides and not to bind to human non-IL-lra-R polypeptides. It will be appreciated, however, that the selective binding agents may also bind orthologs of the polypeptide as set forth in any of SEQ ID NO: 2, SEQ ID NO: 4, SEQ ID NO: 6, or SEQ ID NO: 36, that is, interspecies versions thereof, such as mouse and rat IL-lra-R polypeptides.
The term "transduction" is used to refer to the transfer of genes from one bacterium to another, usually by a phage. "Transduction" also refers to the acquisition and transfer of cukaryotic cellular sequences by retroviruses.
The term "transfection" is used to refer to the uptake of foreign or exogenous DNA by a cell, and a cell has been "transfected" when the exogenous DNA has been introduced inside the cell membrane. A number of transfection techniques are well known in the art and are disclosed herein. See, Graham et al., 1973, Virology 52:456; Sambrook et al., Molecular Cloning, A Laboratory Manual (Cold Spring Harbor Laboratories, 1989); Davis et al., Basic Methods in Molecular Biology (Elsevier, 1986); and Chu et al., 1981, Gene 13:197. Such techniques can be used to introduce one or more exogenous DNA moieties into suitable host cells.
The term "transformation" as used herein refers to a change in a cell's genetic characteristics, and a cell has been transformed when it has been modified to contain a new DNA. For example, a cell is transformed where it is genetically modified from its native state. Following transfection or transduction, the transforming DNA may recombine with that of the cell by physically integrating into a chromosome of the cell, may be maintained transiently as an episomal element without being replicated, or may replicate independently as a plasmid. A 19 WO 01/42304 PCT/US0O/32940 cell is considered to have been stably transformed when the DNA is replicated with the division of the cell.
Relatedness of Nucleic Acid Molecules and/or Polypeptides It is understood that related nucleic acid molecules include allelic or splice variants of the nucleic acid molecule of any of SEQ ID NO: 1, SEQ ID NO: 3, SEQ ID NO: 5, or SEQ ID NO: 35, and include sequences which are complementary to any of the above nucleotide sequences. Related nucleic acid molecules also include a nucleotide sequence encoding a polypeptide comprising or consisting essentially of a substitution, modification, addition and/or deletion of one or more amino acid residues compared to the polypeptide in any of SEQ ID NO: 2, SEQ ID NO: 4, SEQ ID NO: 6, or SEQ ID NO: 36. Such related IL-lra-R polypeptides may comprise, for example, an addition and/or a deletion of one or more N-linked or O-linked glycosylation sites or an addition and/or a deletion of one or more cysteine residues.
Related nucleic acid molecules also include fragments of IL-Ira-R nucleic acid molecules which encode a polypeptide of at least about 25 contiguous amino acids, or about 50 amino acids, or about 75 amino acids, or about 100 amino acids, or about 125 amino acids, or more than 125 amino acid residues of the IL- Ira-R polypeptide of any of SEQ ID NO: 2, SEQ ID NO: 4, SEQ ID NO: 6, or SEQ ID NO: 36.
In addition, related IL-lra-R nucleic acid molecules also include those molecules which comprise nucleotide sequences which hybridize under moderately or highly stringent conditions as defined herein with the fully complementary sequence of the IL-1 ra-R nucleic acid molecule of any of SEQ ID NO: 1, SEQ ID NO: 3, SEQ ID NO: 5, or SEQ ID NO: 35, or of a molecule encoding a polypeptide, which polypeptide comprises the amino acid sequence as shown in any of SEQ ID NO: 2, SEQ ID NO: 4, SEQ ID NO: 6, or SEQ ID NO: 36, or of a nucleic acid fragment as defined herein, or of a nucleic acid fragment encoding a polypeptide as defined herein. Hybridization probes may be prepared using the IL-lra-R sequences provided herein to screen cDNA, genomic or WO 01/42304 PCT/US00/32940 synthetic DNA libraries for related sequences. Regions of the DNA and/or amino Sacid sequence of IL-lra-R polypeptide that exhibit significant identity to known sequences are readily determined using sequence alignment algorithms as described herein and those regions may be used to design probes for screening.
The term "highly stringent conditions" refers to those conditions that are designed to permit hybridization of DNA strands whose sequences are highly complementary, and to exclude hybridization of significantly mismatched DNAs.
Hybridization stringency is principally determined by temperature, ionic strength, and the concentration of denaturing agents such as formamide. Examples of "highly stringent conditions" for hybridization and washing are 0.015 M sodium chloride, 0.0015 M sodium citrate at 65-68 0 C or 0.015 M sodium chloride, 0.0015 M sodium citrate, and 50% formamide at 42 0 C. See Sambrook, Fritsch Maniatis, Molecular Cloning: A Laboratory Manual (2nd ed., Cold Spring Harbor Laboratory, 1989); Anderson et al., Nucleic Acid Hybridisation: A Practical Approach Ch. 4 (IRL Press Limited).
More stringent conditions (such as higher temperature, lower ionic strength, higher formamide, or other denaturing agent) may also be used however, the rate of hybridization will be affected. Other agents may be included in the hybridization and washing buffers for the purpose of reducing non-specific and/or background hybridization. Examples are 0.1% bovine serum albumin, 0.1% polyvinyl-pyrrolidone, 0.1% sodium pyrophosphate, 0.1% sodium dodecylsulfate, NaDodSO 4 (SDS), ficoll, Denhardt's solution, sonicated salmon sperm DNA (or another non-complementary DNA), and dextran sulfate, although other suitable agents can also be used. The concentration and types of these additives can be changed without substantially affecting the stringency of the hybridization conditions. Hybridization experiments are usually carried out at pH 6.8-7.4; however, at typical ionic strength conditions, the rate of hybridization is nearly independent of pH. See Anderson et al., Nucleic Acid Hybridisation: A Practical Approach Ch. 4 (IRL Press Limited).
3 0 Factors affecting the stability of DNA duplex include base composition, length, and degree of base pair mismatch. Hybridization conditions can be 21 WO 01/42304 PCT/US00/32940 adjusted by one skilled in the art in order to accommodate these variables and allow DNAs of different sequence relatedness to form hybrids. The melting temperature of a perfectly matched DNA duplex can be estimated by the following equation: Tm(°C) 81.5 16.6(log[Na+]) 0.41(%G+C) 600/N 0.72(%formamide) where N is the length of the duplex formed, is the molar concentration of the sodium ion in the hybridization or washing solution, %G+C is the percentage of (guanine+cytosine) bases in the hybrid. For imperfectly matched hybrids, the melting temperature is reduced by approximately 1°C for each 1% mismatch.
The term "moderately stringent conditions" refers to conditions under which a DNA duplex with a greater degree of base pair mismatching than could occur under "highly stringent conditions" is able to form. Examples of typical "moderately stringent conditions" are 0.015 M sodium chloride, 0.0015 M sodium citrate at 50-65 0 C or 0.015 M sodium chloride, 0.0015 M sodium citrate, and formamide at 37-50 0 C. By way of example, "moderately stringent conditions" of in 0.015 M sodium ion will allow about a 21% mismatch.
It will be appreciated by those skilled in the art that there is no absolute distinction between "highly stringent conditions" and "moderately stringent conditions." For example, at 0.015 M sodium ion (no formamide), the melting temperature of perfectly matched long DNA is about 71 0 C. With a wash at 65 0
C
(at the same ionic strength), this would allow for approximately a 6% mismatch.
To capture more distantly related sequences, one skilled in the art can simply lower the temperature or raise the ionic strength.
A good estimate of the melting temperature in 1M NaCI* for oligonucleotide probes up to about 20nt is given by: Tm 2 0 C per A-T base pair 4 0 C per G-C base pair *The sodium ion concentration in 6X salt sodium citrate (SSC) is 1 M. See Suggs et al., Developmental Biology Using Purified Genes 683 (Brown and Fox, eds., 1981).
High stringency washing conditions for oligonucleotides are usually at a temperature of 0-5 0 C below the Tm of the oligonucleotide in 6X SSC, 0.1% SDS.
22 WO 01/42304 PCT/US00/32940 In another embodiment, related nucleic acid molecules comprise or consist of a nucleotide sequence that is at least about 70 percent identical to the nucleotide sequence as shown in any of SEQ ID NO: 1, SEQ ID NO: 3, SEQ ID NO: 5, or SEQ ID NO: 35, or comprise or consist essentially of a nucleotide sequence encoding a polypeptide that is at least about 70 percent identical to the polypeptide as set forth in any of SEQ ID NO: 2, SEQ ID NO: 4, SEQ ID NO: 6, or SEQ ID NO: 36. In preferred embodiments, the nucleotide sequences are about percent, or about 80 percent, or about 85 percent, or about 90 percent, or about 96, 97, 98, or 99 percent identical to the nucleotide sequence as shown in any of SEQ ID NO: 1, SEQ ID NO: 3, SEQ ID NO: 5, or SEQ ID NO: 35, or the nucleotide sequences encode a polypeptide that is about 75 percent, or about percent, or about 85 percent, or about 90 percent, or about 95, 96, 97, 98, or 99 percent identical to the polypeptide sequence as set forth in any of SEQ ID NO: 2, SEQ ID NO: 4, SEQ ID NO: 6, or SEQ ID NO: 36. Related nucleic acid molecules encode polypeptides possessing at least one activity of the polypeptide set forth in any of SEQ ID NO: 2, SEQ ID NO: 4, SEQ ID NO: 6, or SEQ ID NO: 36.
Differences in the nucleic acid sequence may result in conservative and/or non-conservative modifications of the amino acid sequence relative to the amino acid sequence of any of SEQ ID NO: 2, SEQ ID NO: 4, SEQ ID NO: 6, or SEQ ID NO: 36.
Conservative modifications to the amino acid sequence of any of SEQ ID NO: 2, SEQ ID NO: 4, SEQ ID NO: 6, or SEQ ID NO: 36 (and the corresponding modifications to the encoding nucleotides) will produce a polypeptide having functional and chemical characteristics similar to those of IL-lra-R polypeptides.
In contrast, substantial modifications in the functional and/or chemical characteristics of IL-lra-R polypeptides may be accomplished by selecting substitutions in the amino acid sequence of any of SEQ ID NO: 2, SEQ ID NO: 4, SEQ ID NO: 6, or SEQ ID NO: 36 that differ significantly in their effect on maintaining the structure of the molecular backbone in the area of the substitution, for example, as a sheet or helical conformation, the charge or 23 WO 01/42304 PCT/US00/32940 hydrophobicity of the molecule at the target site, or the bulk of the side chain.
For example, a "conservative amino acid substitution" may involve a substitution of a native amino acid residue with a nonnative residue such that there is little or no effect on the polarity or charge of the amino acid residue at that position. Furthermore, any native residue in the polypeptide may also be substituted with alanine, as has been previously described for "alanine scanning mutagenesis." Conservative amino acid substitutions also encompass non-naturally occurring amino acid residues that are typically incorporated by chemical peptide synthesis rather than by synthesis in biological systems. These include peptidomimetics, and other reversed or inverted forms of amino acid moieties.
Naturally occurring residues may be divided into classes based on common side chain properties: 1) hydrophobic: norleucine, Met, Ala, Val, Leu, lie; 2) neutral hydrophilic: Cys, Ser, Thr; 3) acidic: Asp, Glu; 4) basic: Asn, Gin, His, Lys, Arg; residues that influence chain orientation: Gly, Pro; and 6) aromatic: Trp, Tyr, Phe.
For example, non-conservative substitutions may involve the exchange of a member of one of these classes for a member from another class. Such substituted residues may be introduced into regions of the human IL-lra-R polypeptide that are homologous with non-human IL-lra-R polypeptides, or into the non-homologous regions of the molecule.
In making such changes, the hydropathic index of amino acids may be considered. Each amino acid has been assigned a hydropathic index on the basis of its hydrophobicity and charge characteristics. The hydropathic indices are: isoleucine valine leucine phenylalanine cysteine/cystine methionine alanine glycine threonine serine tryptophan tyrosine proline histidine glutamate glutamine aspartate asparagine WO 01/42304 PCT/US00/32940 lysine and arginine The importance of the hydropathic amino acid index in conferring interactive biological function on a protein is generally understood in the art (Kyte et al., 1982, J. Mol. Biol. 157:105-31). It is known that certain amino acids may be substituted for other amino acids having a similar hydropathic index or score and still retain a similar biological activity. In making changes based upon the hydropathic index, the substitution of amino acids whose hydropathic indices are within ±2 is preferred, those which are within 1 are particularly preferred, and those within ±0.5 are even more particularly preferred.
It is also understood in the art that the substitution of like amino acids can be made effectively on the basis of hydrophilicity, particularly where the biologically functionally equivalent protein or peptide thereby created is intended for use in immunological embodiments, as in the present case. The greatest local average hydrophilicity of a protein, as governed by the hydrophilicity of its adjacent amino acids, correlates with its immunogenicity and antigenicity, i.e., with a biological property of the protein.
The following hydrophilicity values have been assigned to these amino acid residues: arginine lysine aspartate glutamate serine asparagine glutamine glycine threonine proline alanine histidine cysteine methionine valine leucine isoleucine tyrosine phenylalanine and tryptophan In making changes based upon similar hydrophilicity values, the substitution of amino acids whose hydrophilicity values are within ±2 is preferred, those which are within ±1 are particularly preferred, and those within ±0.5 are even more particularly preferred. One may also identify epitopes from primary amino acid sequences on the basis of hydrophilicity. These regions are also referred to as "epitopic core regions." Desired amino acid substitutions (whether conservative or nonconservative) can be determined by those skilled in the art at the time such substitutions are desired. For example, amino acid substitutions can be used to identify important residues of the IL-lra-R polypeptide, or to increase or decrease WO 01/42304 WO 0142304PCTIUSOO/32940 the affinity of the IL- I ra-R polypeptides described herein. Exemplary amino acid substitutions are set forth in Table 1.
Table I Amino Acid Substitutions Original Residues Exemplary Substitutions Preferred Substitutions Ala Val, Leu, Ile Val Arg Lys, Gin, Asn Lys Asn Gin Gin Asp Glu Glu Cys Ser, Ala Ser Gin Asri Asn Giu Asp Asp Giy Pro, Ala Ala His Asn, Gin, Lys, Arg Arg Ile Leu, Val, Met, Ala, Leu Phe, Norleucine Leu Norleucine, Ile, Ie Val, Met, Ala, Phe Lys Arg, 1,4 Diamino-butyric Arg Acid, Gin, Asn Met Leu, Phe, Ilie Leu Phe Leu, Val, Ile, Ala, Leu Tyr Pro Ala Gly Ser Thr, Aia, Cys Thr Thr Ser Ser Trp Tyr, Phe Tyr Tyr Trp, Phe, Thr, Ser Phe WO 01/42304 PCT/US00/32940 Val lie, Met, Leu, Phe, Leu Ala, Norleucine A skilled artisan will be able to determine suitable variants of the polypeptide as set forth in any of SEQ ID NO: 2, SEQ ID NO: 4, SEQ ID NO: 6, or SEQ ID NO: 36 using well-known techniques. For identifying suitable areas of the molecule that may be changed without destroying biological activity, one skilled in the art may target areas not believed to be important for activity. For example, when similar polypeptides with similar activities from the same species or from other species arc known, one skilled in the art may compare the amino acid sequence of an IL-Ira-R polypeptide to such similar polypeptides. With such a comparison, one can identify residues and portions of the molecules that are conserved among similar polypeptides. It will be appreciated that changes in areas of the IL-lra-R molecule that are not conserved relative to such similar polypeptides would be less likely to adversely affect the biological activity and/or structure of an IL-lra-R polypeptide. One skilled in the art. would also know that, even in relatively conserved regions, one may substitute chemically similar amino acids for the naturally occurring residues while retaining activity (conservative amino acid residue substitutions). Therefore, even areas that may be important for biological activity or for structure may be subject to conservative amino acid substitutions without destroying the biological activity or without adversely affecting the polypeptide structure.
Additionally, one skilled in the art can review structure-function studies identifying residues in similar polypeptides that are important for activity or structure. In view of such a comparison, one can predict the importance of amino acid residues in an IL-lra-R polypeptide that correspond to amino acid residues that are important for activity or structure in similar polypeptides. One skilled in the art may opt for chemically similar amino acid substitutions for such predicted important amino acid residues of IL-Ira-R polypeptides.
One skilled in the art can also analyze the three-dimensional structure and amino acid sequence in relation to that structure in similar polypeptides. In view 27 WO 01/42304 PCT/US00/32940 of such information, one skilled in the art may predict the alignment of amino acid residues of IL-lra-R polypeptide with respect to its three dimensional structure.
One skilled in the art may choose not to make radical changes to amino acid residues predicted to be on the surface of the protein, since such residues may be involved in important interactions with other molecules. Moreover, one skilled in the art may generate test variants containing a single amino acid substitution at each amino acid residue. The variants could be screened using activity assays known to those with skill in the art. Such variants could be used to gather information about suitable variants. For example, if one discovered that a change to a particular amino acid residue resulted in destroyed, undesirably reduced, or unsuitable activity, variants with such a change would be avoided. In other words, based on information gathered from such routine experiments, one skilled in the art can readily determine the amino acids where further substitutions should be avoided either alone or in combination with other mutations.
A number of scientific publications have been devoted to the prediction of secondary structure. See Moult, 1996, Curr. Opin. Biotechnol. 7:422-27; Chou et al.. 1974, Biochemistry 13:222-45; Chou et al., 1974, Biochemistry 113:211-22; Chou et al., 1978, Adv. Enzymol. Relat. Areas Mol. Biol. 47:45-48; Chou et al., 1978, Ann. Rev. Biochem. 47:251-276; and Chou et al., 1979, Biophys. J. 26:367- 84. Moreover, computer programs are currently available to assist with predicting secondary structure. One method of predicting secondary structure is based upon homology modeling. For example, two polypeptides or proteins which have a sequence identity of greater than 30%, or similarity greater than 40%, often have similar structural topologies. The recent growth of the protein structural database (PDB) has provided enhanced predictability of secondary structure, including the potential number of folds within the structure of a polypeptide or protein. See Holm et al., 1999, Nucleic Acids Res. 27:244-47. It has been suggested that there are a limited number of folds in a given polypeptide or protein and that once a critical number of structures have been resolved, structural prediction will become dramatically more accurate (Brenner et al., 1997, Curr. Opin. Struct. Biol. 7:369- 76).
WO 01/42304 PCT/US00/32940 Additional methods of predicting secondary structure include "threading" (Jones, 1997, Curr. Opin. Struct. Biol. 7:377-87; Sippl et al., 1996, Structure 4:15-19), "profile analysis" (Bowie et al., 1991, Science, 253:164-70; Gribskov et al., 1990, Methods Enzymol. 183:146-59; Gribskov et al., 1987, Proc. Nat. Acad.
Sci. U.S.A. 84:4355-58), and "evolutionary linkage" (See Holm et al., supra, and Brenner et al., supra).
Preferred IL-lra-R polypeptide variants include glycosylation variants wherein the number and/or type of glycosylation sites have been altered compared to the amino acid sequence set forth in any of SEQ ID NO: 2, SEQ ID NO: 4, SEQ ID NO: 6, or SEQ ID NO: 36. In one embodiment, IL-lra-R polypeptide variants comprise a greater or a lesser number of N-linked glycosylation sites than the amino acid sequence set forth in any of SEQ ID NO: 2, SEQ ID NO: 4, SEQ ID NO: 6, or SEQ ID NO: 36. An N-linked glycosylation site is characterized by the sequence: Asn-X-Ser or Asn-X-Thr, wherein the amino acid residue designated as X may be any amino acid residue except proline. The substitution of amino acid residues to create this sequence provides a potential new site for the addition of an N-linked carbohydrate chain. Alternatively, substitutions that eliminate this sequence will remove an existing N-linked carbohydrate chain.
Also provided is a rearrangement of N-linked carbohydrate chains wherein one or more N-linked glycosylation sites (typically those that are naturally occurring) are eliminated and one or more new N-linked sites are created. Additional preferred IL-lra-R variants include cysteine variants, wherein one or more cysteine residues are deleted or substituted with another amino acid scrinc) as compared to the amino acid sequence set forth in any of SEQ ID NO: 2, SEQ ID NO: 4, SEQ ID NO: 6, or SEQ ID NO: 36. Cysteine variants are useful when IL-lra-R polypeptides must be refolded into a biologically active conformation such as after the isolation of insoluble inclusion bodies. Cysteine variants generally have fewer cysteine residues than the native protein, and typically have an even number to minimize interactions resulting from unpaired cysteines.
In other embodiments, related nucleic acid molecules comprise or consist of a nucleotide sequence encoding a polypeptide as set forth in any of SEQ ID 29 WO 01/42304 PCT/US00/32940 NO: 2, SEQ ID NO: 4, SEQ ID NO: 6, or SEQ ID NO: 36 with at least one amino acid insertion and wherein the polypeptide has an activity of the polypeptide set forth in any of SEQ ID NO: 2, SEQ ID NO: 4, SEQ ID NO: 6, or SEQ ID NO: 36, or a nucleotide sequence encoding a polypeptide as set forth in any of SEQ ID NO: 2, SEQ ID NO: 4, SEQ ID NO: 6, or SEQ ID NO: 36 with at least one amino acid deletion and wherein the polypeptide has an activity of the polypeptide set forth in any of SEQ ID NO: 2, SEQ ID NO: 4, SEQ ID NO: 6, or SEQ ID NO: 36.
Related nucleic acid molecules also comprise or consist of a nucleotide sequence encoding a polypeptide as set forth in any of SEQ ID NO: 2, SEQ ID NO: 4, SEQ ID NO: 6, or SEQ ID NO: 36 wherein the polypeptide has a carboxyl- and/or amino-terminal truncation and further wherein the polypeptide has an activity of the polypeptide set forth in any of SEQ ID NO: 2, SEQ ID NO: 4, SEQ ID NO: 6, or SEQ ID NO: 36. Related nucleic acid molecules also comprise or consist of a nucleotide sequence encoding a polypeptide as set forth in any of SEQ ID NO: 2, SEQ ID NO: 4, SEQ ID NO: 6, or SEQ ID NO: 36 with at least one modification selected from the group consisting of amino acid substitutions, amino acid insertions, amino acid deletions, carboxyl-terminal truncations, and aminoterminal truncations and wherein the polypeptide has an activity of the polypeptide set forth in any of SEQ ID NO: 2, SEQ ID NO: 4, SEQ ID NO: 6, or SEQ ID NO: 36.
In addition, the polypeptide comprising the amino acid sequence of any of SEQ ID NO: 2, SEQ ID NO: 4, SEQ ID NO: 6, or SEQ ID NO: 36, or other IL- Ira-R polypeptide, may be fused to a homologous polypeptide to form a homodimer or to a heterologous polypeptide to form a heterodimer. Heterologous peptides and polypeptides include, but are not limited to: an epitope to allow for the detection and/or isolation of an IL-Ira-R fusion polypeptide; a transmembrane receptor protein or a portion thereof, such as an extracellular domain or a transmembrane and intracellular domain; a ligand or a portion thereof which binds to a transmembrane receptor protein; an enzyme or portion thereof which is catalytically active; a polypeptide or peptide which promotes oligomerization, such as a leucine zipper domain; a polypeptide or peptide which increases WO 01/42304 PCT/US00/32940 stability, such as an immunoglobulin constant region; and a polypeptide which has a therapeutic activity different from the polypeptide comprising the amino acid sequence as set forth in any of SEQ ID NO: 2, SEQ ID NO: 4, SEQ ID NO: 6, or SEQ ID NO: 36, or other IL- ra-R polypeptide.
Fusions can be made either at the amino-terminus or at the carboxylterminus of the polypeptide comprising the amino acid sequence set forth in any of SEQ ID NO: 2, SEQ ID NO: 4, SEQ ID NO: 6, or SEQ ID NO: 36, or other IL- Ira-R polypeptide. Fusions may be direct with no linker or adapter molecule or may be through a linker or adapter molecule. A linker or adapter molecule may be one or more amino acid residues, typically from about 20 to about 50 amino acid residues. A linker or adapter molecule may also be designed with a cleavage site for a DNA restriction endonuclease or for a protease to allow for the separation of the fused moieties. It will be appreciated that once constructed, the fusion polypeptides can be derivatized according to the methods described herein.
In a further embodiment of the invention, the polypeptide comprising the amino acid sequence of any of SEQ ID NO: 2, SEQ ID NO: 4, SEQ ID NO: 6, or SEQ ID NO: 36, or other IL-lra-R polypeptide, is fused to one or more domains of an Fc region of human IgG. Antibodies comprise two functionally independent parts, a variable domain known as "Fab," that binds an antigen, and a constant domain known as that is involved in effector functions such as complement activation and attack by phagocytic cells. An Fc has a long serum half-life, whereas an Fab is short-lived. Capon et al., 1989, Nature 337:525-31. When constructed together with a therapeutic protein, an Fc domain can provide longer half-life or incorporate such functions as Fc receptor binding, protein A binding, complement fixation, and perhaps even placental transfer. Id. Table II summarizes the use of certain Fc fusions known in the art.
Table II Fc Fusion with Therapeutic Proteins Form of Fc I Fusion partner Therapeutic implications Reference WO 01/42304 PCT/USO0/32940 IgGI N-terminus of Hodgkin's disease; U.S. Patent No.
anaplastic lymphoma; T- 5,480,981 cell leukemia Murine Fcy2a IL-10 anti-inflammatory; Zheng et al., 1995, J.
transplant rejection Immunol. 154:5590-600 IgGI TNF receptor septic shock Fisher et al., 1996, N.
Engl. J. Med. 334:1697- 1702; Van Zee et a., 1996, J. Immunol.
156:2221-30 IgG, IgA, IgM, TNF receptor inflammation, U.S. Patent No.
or IgE autoimmune disorders 5,808,029 (excluding the first domain) IgG I CD4 receptor AIDS Capon et al., 1989, Nature 337: 525-31 IgG1, N-terminus anti-cancer, antiviral Harvill et al., 1995, IgG3 of IL-2 Immunotech. 1:95-105 IgGI C-terminus of osteoarthritis; WO 97/23614 OPG bone density IgG 1 N-terminus of anti-obesity PCT/US 97/23183, filed leptin December 11, 1997 Human Ig Cy I CTLA-4 autoimmune disorders Linsley, 1991, J. Exp.
Med., 174:561-69 In one example, a human IgG hinge, CH2, and CH3 region may be fused at either the amino-terminus or carboxyl-terminus of the IL-I ra-R polypeptides using methods known to the skilled artisan. In another example, a human IgG hinge, CH2, and CH3 region may be fused at either the amino-terminus or carboxyl-terminus of an IL-lra-R polypeptide fragment the predicted extracellular portion of IL- ra-R polypeptide).
The resulting IL-lra-R fusion polypeptide may be purified by use of a Protein A affinity column. Peptides and proteins fused to an Fc region have been found to exhibit a substantially greater half-life in vivo than the unfused counterpart. Also, a fusion to an Fc region allows for dimerization/multimerization of the fusion polypeptide. The Fe region may be a naturally occurring Fc region, or may be altered to improve certain qualities, such as therapeutic qualities, circulation time, or reduced aggregation.
WO 01/42304 PCT/US00/32940 Identity and similarity of related nucleic acid molecules and polypeptides are readily calculated by known methods. Such methods include, but are not limited to those described in Computational Molecular Biology Lesk, ed., Oxford University Press 1988); Biocomputing: Informatics and Genome.Projects Smith, ed., Academic Press 1993); Computer Analysis of Sequence Data (Part 1, A.M. Griffin and H.G. Griffin, eds., Humana Press 1994); G. von Heinle, Sequence Analysis in Molecular Biology (Academic Press 1987); Sequence Analysis Primer Gribskov and J. Devereux, eds., M. Stockton Press 1991); and Carillo et al., 1988, SIAM J. Applied Math., 48:1073.
Preferred methods to determine identity and/or similarity are designed to give the largest match between the sequences tested. Methods to determine identity and similarity are described in publicly available computer programs.
Preferred computer program methods to determine identity and similarity between two sequences include, but are not limited to, the GCG program package, including GAP (Devereux et al., 1984, Nucleic Acids Res. 12:387; Genetics Computer Group, University of Wisconsin, Madison, WI), BLASTP, BLASTN, and FASTA (Altschul et al., 1990, J. Mol. Biol. 215:403-10). The BLASTX program is publicly available from the National Center for Biotechnology Information (NCBI) and other sources (Altschul et al., BLAST Manual (NCB NLM NIH, Bethesda, MD); Altschul et al., 1990, supra). The well-known Smith Waterman algorithm may also be used to determine identity.
Certain alignment schemes for aligning two amino acid sequences may result in the matching of only a short region of the two sequences, and this small aligned region may have very high sequence identity even though there is no significant relationship between the two full-length sequences. Accordingly, in a preferred embodiment, the selected alignment method (GAP program) will result in an alignment that spans at least 50 contiguous amino acids of the claimed polypeptide.
For example, using the computer algorithm GAP (Genetics Computer Group, University of Wisconsin, Madison, WI), two polypeptides for which the percent sequence identity is to be determined are aligned for optimal matching of 33 WO 01/42304 PCT/US0O/32940 their respective amino acids (the "matched span," as determined by the algorithm). A gap opening penalty (which is calculated as 3X the average diagonal; the "average diagonal" is the average of the diagonal of the comparison matrix being used; the "diagonal" is the score or number assigned to each perfect amino acid match by the particular comparison matrix) and a gap extension penalty (which is usually 0. 1X the gap opening penalty), as well as a comparison matrix such as PAM 250 or BLOSUM 62 are used in conjunction with the algorithm. A standard comparison matrix is also used by the algorithm (see Dayhoff et al., 5 Atlas of Protein Sequence and Structure (Supp. 3 1978)(PAM250 comparison matrix); Henikoff et al., 1992, Proc. Natl. Acad. Sci USA 89:10915-19 (BLOSUM 62 comparison matrix)).
Preferred parameters for polypeptide sequence comparison include the following: Algorithm: Needleman and Wunsch, 1970, J. Mol. Biol. 48:443-53; Comparison matrix: BLOSUM 62 (Henikoffet al., supra); Gap Penalty: 12 Gap Length Penalty: 4 Threshold of Similarity: 0 The GAP program is useful with the above parameters. The aforementioned parameters are the default parameters for polypeptide comparisons (along with no penalty for end gaps) using the GAP algorithm.
Preferred parameters for nucleic acid molecule sequence comparison include the following: Algorithm: Needleman and Wunsch, supra; Comparison matrix: matches +10, mismatch 0 Gap Penalty: Gap Length Penalty: 3 WO 01/42304 PCT/USOO/32940 The GAP program is also useful with the above parameters. The aforementioned parameters are the default parameters for nucleic acid molecule comparisons.
Other exemplary algorithms, gap opening penalties, gap extension penalties, comparison matrices, and thresholds of similarity may be used, including those set forth in the Program Manual, Wisconsin Package, Version 9, September, 1997. The particular choices to be made will be apparent to those of skill in the art and will depend on the specific comparison to be made, such as DNA-to-DNA, protein-to-protein, protein-to-DNA; and additionally, whether the comparison is between given pairs of sequences (in which case GAP or BestFit are generally preferred) or between one sequence and a large database of sequences (in which case FASTA or BLASTA are preferred).
Nucleic Acid Molecules The nucleic acid molecules encoding a polypeptide comprising the amino acid sequence of an IL-lra-R polypeptide can readily be obtained in a variety of ways including, without limitation, chemical synthesis, cDNA or genomic library screening, expression library screening, and/or PCR amplification of cDNA.
Recombinant DNA methods used herein are generally those set forth in Sambrook et al., Molecular Cloning: A Laboratory Manual (Cold Spring Harbor Laboratory Press, 1989) and/or Current Protocols in Molecular Biology (Ausubel et al., eds., Green Publishers Inc. and Wiley and Sons 1994). The invention provides for nucleic acid molecules as described herein and methods for obtaining such molecules.
Where a gene encoding the amino acid sequence of an IL-lra-R polypeptide has been identified from one species, all or a portion of that gene may be used as a probe to identify orthologs or related genes from the same species.
The probes or primers may be used to screen cDNA libraries from various tissue sources believed to express the IL-lra-R polypeptide. In addition, part or all of a nucleic acid molecule having the sequence as set forth in any of SEQ ID NO: 1, SEQ ID NO: 3, SEQ ID NO: 5, or SEQ ID NO: 35 may be used to screen a genomic library to identify and isolate a gene encoding the amino acid sequence WO 01/42304 PCT/US00/32940 of an IL-Ira-R polypeptide. Typically, conditions of moderate or high stringency will be employed for screening to minimize the number of false positives obtained from the screening.
Nucleic acid molecules encoding the amino acid sequence of IL-Ira-R polypeptides may also be identified by expression cloning which employs the detection of positive clones based upon a property of the expressed protein.
Typically, nucleic acid libraries are screened by the binding an antibody or other binding partner receptor or ligand) to cloned proteins that are expressed and displayed on a host cell surface. The antibody or binding partner is modified with a detectable label to identify those cells expressing the desired clone.
Recombinant expression techniques conducted in accordance with the descriptions set forth below may be followed to produce these polynucleotides and to express the encoded polypeptides. For example, by inserting a nucleic acid sequence that encodes the amino acid sequence of an IL-lra-R polypeptide into an appropriate vector, one skilled in the art can readily produce large quantities of the desired nucleotide sequence. The sequences can then be used to generate detection probes or amplification primers. Alternatively, a polynucleotide encoding the amino acid sequence of an IL- ra-R polypeptide can be inserted into an expression vector. By introducing the expression vector into an appropriate host, the encoded IL-Ira-R polypeptide may be produced in large amounts.
Another method for obtaining a suitable nucleic acid sequence is the polymerase chain reaction (PCR). In this method, cDNA is prepared from poly(A)+RNA or total RNA using the enzyme reverse transcriptase. Two primers, typically complementary to two separate regions of cDNA encoding the amino acid sequence of an IL-lra-R polypeptide, are then added to the cDNA along with a polymerase such as Taq polymerase, and the polymerase amplifies the cDNA region between the two primers.
Another means of preparing a nucleic acid molecule encoding the amino acid sequence of an IL-lra-R polypeptide is chemical synthesis using methods well known to the skilled artisan such as those described by Engels et al., 1989, Angew. Chem. Intl. Ed. 28:716-34. These methods include, inter alia, the 36 WO 01/42304 PCT/US00/32940 phosphotriester, phosphoramidite, and H-phosphonate methods for nucleic acid synthesis. A preferred method for such chemical synthesis is polymer-supported synthesis using standard phosphoramidite chemistry. Typically, the DNA encoding the amino acid sequence of an IL-lra-R polypeptide will be several hundred nucleotides in length. Nucleic acids larger than about 100 nucleotides can be synthesized as several fragments using these methods. The fragments can then be ligated together to form the full-length nucleotide sequence of an IL-Ira-R gene. Usually, the DNA fragment encoding the amino-terminus of the polypeptide will have an ATG, which encodes a methionine residue. This methionine may or may not be present on the mature form of the IL-lra-R polypeptide, depending on whether the polypeptide produced in the host cell is designed to be secreted from that cell. Other methods known to the skilled artisan may be used as well.
In certain embodiments, nucleic acid variants contain codons which have been altered for optimal expression of an IL-lra-R polypeptide in a given host cell. Particular codon alterations will depend upon the IL-lra-R polypeptide and host cell selected for expression. Such "codon optimization" can be carried out by a variety of methods, for example, by selecting codons which are preferred for use in highly expressed genes in a given host cell. Computer algorithms which incorporate codon frequency tables such as "Eco_high.Cod" for codon preference of highly expressed bacterial genes may be used and are provided by the University of Wisconsin Package Version 9.0 (Genetics Computer Group, Madison, WI). Other useful codon frequency tables include "Celcganshigh.cod," "Celegans_low.cod," "Drosophila_high.cod," "Humanhigh.cod," "Maizehigh.cod," and "Yeasthigh.cod." In some cases, it may be desirable to prepare nucleic acid molecules encoding IL-lra-R polypeptide variants. Nucleic acid molecules encoding variants may be produced using site directed mutagenesis, PCR amplification, or other appropriate methods, where the primer(s) have the desired point mutations (see Sambrook et al., supra, and Ausubel et al, supra, for descriptions of mutagenesis techniques). Chemical synthesis using methods described by Engels 37 WO 01/42304 PCT/US0/32940 el al., supra, may also be used to prepare such variants. Other methods known to the skilled artisan may be used as well.
Vectors and Host Cells A nucleic acid molecule encoding the amino acid sequence of an IL-lra-R polypeptide is inserted into an appropriate expression vector using standard ligation techniques. The vector is typically selected to be functional in the particular host cell employed the vector is compatible with the host cell machinery such that amplification of the gene and/or expression of the gene can occur). A nucleic acid molecule encoding the amino acid sequence of an IL-lra-R polypeptide may be amplified/expressed in prokaryotic, yeast, insect (baculovirus systems) and/or eukaryotic host cells. Selection of the host cell will depend in part on whether an IL-lra-R polypeptide is to be post-translationally modified glycosylated and/or phosphorylated). If so, yeast, insect, or mammalian host cells are preferable. For a review of expression vectors, see Meth. Enz., vol. 185 Goeddel, ed., Academic Press 1990).
Typically, expression vectors used in any of the host cells will contain sequences for plasmid maintenance and for cloning and expression of exogenous nucleotide sequences. Such sequences, collectively referred to as "flanking sequences" in certain embodiments will typically include one or more of the following nucleotide sequences: a promoter, one or more enhancer sequences, an origin of replication, a transcriptional termination sequence, a complete intron sequence containing a donor and acceptor splice site, a sequence encoding a leader sequence for polypeptide secretion, a ribosome binding site, a polyadenylation sequence, a polylinker region for inserting the nucleic acid encoding the polypeptide to be expressed, and a selectable marker element. Each of these sequences is discussed below.
Optionally, the vector may contain a "tag"-encoding sequence, an oligonucleotide molecule located at the 5' or 3' end of the IL-lra-R polypeptide coding sequence; the oligonucleotide sequence encodes polyHis (such as hexaHis), or another "tag" such as FLAG, HA (hemaglutinin influenza virus), or 38 WO 01/42304 PCT/US00/32940 myc for which commercially available antibodies exist. This tag is typically fused to the polypeptide upon expression of the polypeptide, and can serve as a means for affinity purification of the IL-lra-R polypeptide from the host cell. Affinity purification can be accomplished, for example, by column chromatography using antibodies against the tag as an affinity matrix. Optionally, the tag can subsequently be removed from the purified IL-lra-R polypeptide by various means such as using certain peptidases for cleavage.
Flanking sequences may be homologous from the same species and/or strain as the host cell), heterologous from a species other than the host cell species or strain), hybrid a combination of flanking sequences from more than one source), or synthetic, or the flanking sequences may be native sequences which normally function to regulate IL-lra-R polypcptide expression. As such, the source of a flanking sequence may be any prokaryotic or eukaryotic organism, any vertebrate or invertebrate organism, or any plant, provided that the flanking sequence is functional in, and can be activated by, the host cell machinery.
Flanking sequences useful in the vectors of this invention may be obtained by any of several methods well known in the art. Typically, flanking sequences useful herein other than the IL-lra-R gene flanking sequences will have been previously identified by mapping and/or by restriction endonuclease digestion and can thus be isolated from the proper tissue source using the appropriate restriction endonucleases. In some cases, the full nucleotide sequence of a flanking sequence may be known. Here, the flanking sequence may be synthesized using the methods described herein for nucleic acid synthesis or cloning.
Where all or only a portion of the flanking sequence is known, it may be obtained using PCR and/or by screening a genomic library with a suitable oligonucleotide and/or flanking sequence fragment from the same or another species. Where the flanking sequence is not known, a fragment of DNA containing a flanking sequence may be isolated from a larger piece of DNA that may contain, for example, a coding sequence or even another gene or genes.
Isolation may be accomplished by restriction endonuclease digestion to produce the proper DNA fragment followed by isolation using agarose gel purification, 39 WO 01/42304 PCT/US00/32940 Qiagen® column chromatography (Chatsworth, CA), or other methods known to the skilled artisan. The selection of suitable enzymes to accomplish this purpose will be readily apparent to one of ordinary skill in the art.
An origin of replication is typically a part of those prokaryotic expression vectors purchased commercially, and the origin aids in the amplification of the vector in a host cell. Amplification of the vector to a certain copy number can, in some cases, be important for the optimal expression of an IL-lra-R polypeptide.
If the vector of choice does not contain an origin of replication site, one may be chemically synthesized based on a known sequence, and ligated into the vector.
For example, the origin of replication from the plasmid pBR322 (New England Biolabs, Beverly, MA) is suitable for most gram-negative bacteria and various origins SV40, polyoma, adenovirus, vesicular stomatitus virus (VSV), or papillomaviruses such as HPV or BPV) are useful for cloning vectors in mammalian cells. Generally, the origin of replication component is not needed for mammalian expression vectors (for example, the SV40 origin is often used only because it contains the early promoter).
A transcription termination sequence is typically located 3' of the end of a polypeptide coding region and serves to terminate transcription. Usually, a transcription termination sequence in prokaryotic cells is a G-C rich fragment followed by a poly-T sequence. While the sequence is easily cloned from a library or even purchased commercially as part of a vector, it can also be readily synthesized using methods for nucleic acid synthesis such as those described herein.
A selectable marker gene element encodes a protein necessary for the survival and growth of a host cell grown in a selective culture medium. Typical selection marker genes encode proteins that confer resistance to antibiotics or other toxins, ampicillin, tetracycline, or kanamycin for prokaryotic host cells; complement auxotrophic deficiencies of the cell; or supply critical nutrients not available from complex media. Preferred selectable markers are the kanamycin resistance gene, the ampicillin resistance gene, and the tetracycline resistance gene. A neomycin resistance gene may also be used for selection in WO 01/42304 PCT/US00/32940 prokaryotic and eukaryotic host cells.
Other selection genes may be used to amplify the gene that will be expressed. Amplification is the process wherein genes that are in greater demand for the production of a protein critical for growth are reiterated in tandem within the chromosomes of successive generations of recombinant cells. Examples of suitable selectable markers for mammalian cells include dihydrofolate reductase (DHFR) and thymidine kinase. The mammalian cell transformants are placed under selection pressure wherein only the transformants are uniquely adapted to survive by virtue of the selection gene present in the vector. Selection pressure is imposed by culturing the transformed cells under conditions in which the concentration of selection agent in the medium is successively changed, thereby leading to the amplification of both the selection gene and the DNA that encodes an IL-lra-R polypeptide. As a result, increased quantities of IL-lra-R polypeptide are synthesized from the amplified DNA.
A ribosome binding site is usually necessary for translation initiation of mRNA and is characterized by a Shine-Dalgarno sequence (prokaryotes) or a Kozak sequence (eukaryotes). The element is typically located 3' to the promoter and 5' to the coding sequence of an IL-lra-R polypeptide to be expressed. The Shine-Dalgarno sequence is varied but is typically a polypurine having a high A-G content). Many Shine-Dalgamo sequences have been identified, each of which can be readily synthesized using methods set forth herein and used in a prokaryotic vector.
A leader, or signal, sequence may be used to direct an IL-lra-R polypeptide out of the host cell. Typically, a nucleotide sequence encoding the signal sequence is positioned in the coding region of an IL-lra-R nucleic acid molecule, or directly at the 5' end of an IL-lra-R polypeptide coding region.
Many signal sequences have been identified, and any of those that are functional in the selected host cell may be used in conjunction with an IL-lra-R nucleic acid molecule. Therefore, a signal sequence may be homologous (naturally occurring) or heterologous to the IL-lra-R nucleic acid molecule. Additionally, a signal sequence may be chemically synthesized using methods described herein. In most 41 WO 01/42304 PCT/US00/32940 cases, the secretion of an IL-lra-R polypeptide from the host cell via the presence of a signal peptide will result in the removal of the signal peptide from the secreted IL-lra-R polypeptide. The signal sequence may be a component of the vector, or it may be a part of an IL-lra-R nucleic acid molecule that is inserted into the vector.
Included within the scope of this invention is the use of either a nucleotide sequence encoding a native IL-Ira-R polypeptide signal sequence joined to an IL- Ira-R polypeptide coding region or a nucleotide sequence encoding a heterologous signal sequence joined to an IL-lra-R polypeptide coding region.
The heterologous signal sequence selected should be one that is recognized and processed, cleaved by a signal peptidase, by the host cell. For prokaryotic host cells that do not recognize and process the native IL-lra-R polypeptide signal sequence, the signal sequence is substituted by a prokaryotic signal sequence selected, for example, from the group of the alkaline phosphatase, penicillinase, or heat-stable enterotoxin II leaders. For yeast secretion, the native IL-lra-R polypeptide signal sequence may be substituted by the yeast invertase, alpha factor, or acid phosphatase leaders. In mammalian cell expression the native signal sequence is satisfactory, although other mammalian signal sequences may be suitable.
In some cases, such as where glycosylation is desired in a eukaryotic host cell expression system, one may manipulate the various prescquences to improve glycosylation or yield. For example, one may alter the peptidase cleavage site of a particular signal peptide, or add pro-sequences, which also may affect glycosylation. The final protein product may have, in the -1 position (relative to the first amino acid of the mature protein) one or more additional amino acids incident to expression, which may not have been totally removed. For example, the final protein product may have one or two amino acid residues found in the peptidase cleavage site, attached to the amino-terminus. Alternatively, use of some enzyme cleavage sites may result in a slightly truncated form of the desired IL-lra-R polypeptide, if the enzyme cuts at such area within the mature polypeptide.
WO 01/42304 PCT/US00/32940 In many cases, transcription of a nucleic acid molecule is increased by the presence of one or more introns in the vector; this is particularly true where a polypeptide is produced in cukaryotic host cells, especially mammalian host cells.
The introns used may be naturally occurring within the IL-lra-R gene especially where the gene used is a full-length genomic sequence or a fragment thereof.
Where the intron is not naturally occurring within the gene (as for most cDNAs), the intron may be obtained from another source. The position of the intron with respect to flanking sequences and the IL-lra-R gene is generally important, as the intron must be transcribed to be effective. Thus, when an IL-lra-R cDNA molecule is being transcribed, the preferred position for the intron is 3' to the transcription start site and 5' to the poly-A transcription termination sequence.
Preferably, the intron or introns will be located on one side or the other 5' or of the cDNA such that it does not interrupt the coding sequence. Any intron from any source, including viral, prokaryotic and eukaryotic (plant or animal) organisms, may be used to practice this invention, provided that it is compatible with the host cell into which it is inserted. Also included herein are synthetic introns. Optionally, more than one intron may be used in the vector.
The expression and cloning vectors of the present invention will typically contain a promoter that is recognized by the host organism and operably linked to the molecule encoding the IL-lra-R polypcptide. Promoters are untranscribed sequences located upstream to the start codon of a structural gene (generally within about 100 to 1000 bp) that control the transcription of the structural gene. Promoters are conventionally grouped into one of two classes: inducible promoters and constitutive promoters. Inducible promoters initiate increased levels of transcription from DNA under their control in response to some change in culture conditions, such as the presence or absence of a nutrient or a change in temperature. Constitutive promoters, on the other hand, initiate continual gene product production; that is, there is little or no control over gene expression. A large number of promoters, recognized by a variety of potential host cells, are well known. A suitable promoter is operably linked to the DNA encoding IL-lra-R polypeptide by removing the promoter from the source DNA 43 WO 01/42304 PCT/US00/32940 by restriction enzyme digestion and inserting the desired promoter sequence into the vector. The native IL-Ira-R promoter sequence may be used to direct amplification and/or expression of an IL-lra-R nucleic acid molecule. A heterologous promoter is preferred, however, if it permits greater transcription and higher yields of the expressed protein as compared to the native promoter, and if it is compatible with the host cell system that has been selected for use.
Promoters suitable for use with prokaryotic hosts include the betalactamase and lactose promoter systems; alkaline phosphatase; a tryptophan (trp) promoter system; and hybrid promoters such as the tac promoter. Other known bacterial promoters are also suitable. Their sequences have been published, thereby enabling one skilled in the art to ligate them to the desired DNA sequence, using linkers or adapters as needed to supply any useful restriction sites.
Suitable promoters for use with yeast hosts are also well known in the art.
Yeast enhancers are advantageously used with yeast promoters. Suitable promoters for use with mammalian host cells are well known and include, but are not limited to, those obtained from the genomes of viruses such as polyoma virus, fowlpox virus, adenovirus (such as Adenovirus bovine papilloma virus, avian sarcoma virus, cytomegalovirus, retroviruses, hepatitis-B virus and most preferably Simian Virus 40 (SV40). Other suitable mammalian promoters include heterologous mammalian promoters, for example, heat-shock promoters and the actin promoter.
Additional promoters which may be of interest in controlling IL-lra-R gene expression include, but are not limited to: the SV40 early promoter region (Bemoist and Chambon, 1981, Nature 290:304-10); the CMV promoter; the promoter contained in the 3' long terminal repeat of Rous sarcoma virus (Yamamoto, et al., 1980, Cell 22:787-97); the herpes thymidine kinase promoter (Wagner et al., 1981, Proc. Natl. Acad. Sci. U.S.A. 78:1444-45); the regulatory sequences of the metallothionine gene (Brinster et al., 1982, Nature 296:39-42); prokaryotic expression vectors such as the beta-lactamase promoter (Villa- Kamaroff et al., 1978, Proc. Nail. Acad. Sci. 75:3727-31); or the tac promoter (DeBoer et al., 1983, Proc. Natl. Acad. Sci. 80:21-25). Also of WO 01/42304 PCT/US00/32940 interest are the following animal transcriptional control regions, which exhibit tissue specificity and have been utilized in transgenic animals: the elastase I gene control region which is active in pancreatic acinar cells (Swift et al, 1984, Cell 38:639-46; Omitz et al., 1986, Cold Spring Harbor Symp. Quant. Biol. -50:399- 409 (1986); MacDonald, 1987, Hepatology 7:425-515); the insulin gene control region which is active in pancreatic beta cells (Hanahan, 1985, Nature 315:115- 22); the immunoglobulin gene control region which is active in lymphoid cells (Grosschedl etal., 1984, Cell 38:647-58; Adames et al., 1985, Nature 318:533-38; Alexander et al., 1987, Mol. Cell. Biol., 7:1436-44); the mouse mammary tumor virus control region which is active in testicular, breast, lymphoid and mast cells (Leder et al., 1986, Cell 45:485-95); the albumin gene control region which is active in liver (Pinkert et al., 1987, Genes and Devel. 1:268-76); the alpha-fetoprotein gene control region which is active in liver (Krumlauf et al., 1985, Mol.
Cell. Biol, 5:1639-48; Hammer et al., 1987, Science 235:53-58); the alpha 1antitrypsin gene control region which is active in the liver (Kelsey et al., 1987, Genes and Devel. 1:161-71); the beta-globin gene control region which is active in myeloid cells (Mogram et al., 1985, Nature 315:338-40; Kollias et al., 1986, Cell 46:89-94); the myelin basic protein gene control region which is active in oligodendrocyte cells in the brain (Readhead et al., 1987, Cell 48:703-12); the myosin light chain-2 gene control region which is active in skeletal muscle (Sani, 1985, Nature 314:283-86); and the gonadotropic releasing hormone gene control region which is active in the hypothalamus (Mason et al., 1986, Science 234:1372- 78).
An enhancer sequence may be inserted into the vector to increase the transcription of a DNA encoding an IL-lra-R polypeptide of the present invention by higher eukaryotes. Enhancers are cis-acting elements of DNA, usually about 10-300 bp in length, that act on the promoter to increase transcription. Enhancers are relatively orientation and position independent. They have been found 5' and 3' to the transcription unit. Several enhancer sequences available from mammalian genes are known globin, elastase, albumin, alpha-feto-protein and insulin). Typically, however, an enhancer from a virus will be used. The WO 01/42304 PCT/US00/32940 enhancer, the cytomegalovirus early promoter enhancer, the polyoma enhancer, and adenovirus enhancers are exemplary enhancing elements for the activation of eukaryotic promoters. While an enhancer may be spliced into the vector at a position 5' or 3' to an IL-lra-R nucleic acid molecule, it is typically located at a site 5' from the promoter.
Expression vectors of the invention may be constructed from a starting vector such as a commercially available vector. Such vectors may or may not contain all of the desired flanking sequences. Where one or more of the flanking sequences described herein are not already present in the vector, they may be individually obtained and ligated into the vector. Methods used for obtaining each of the flanking sequences are well known to one skilled in the art.
Preferred vectors for practicing this invention are those which are compatible with bacterial, insect, and mammalian host cells. Such vectors include, inter alia, pCRII, pCR3, and pcDNA3.1 (Invitrogen, San Diego, CA), pBSII (Stratagene, La Jolla, CA), pET15 (Novagen, Madison, WI), pGEX (Pharmacia Biotech, Piscataway, NJ), pEGFP-N2 (Clontech, Palo Alto, CA), pETL (BlueBacII, Invitrogen), pDSR-alpha (PCT Pub. No. WO 90/14363) and pFastBacDual (Gibco-BRL, Grand Island, NY).
Additional suitable vectors include, but are not limited to, cosmids, plasmids, or modified viruses, but it will be appreciated that the vector system must be compatible with the selected host cell. Such vectors include, but are not limited to plasmids such as Bluescripto plasmid derivatives (a high copy number ColEl-based phagemid, Stratagene Cloning Systems, La Jolla CA), PCR cloning plasmids designed for cloning Taq-amplified PCR products TOPOTM TA Cloning' Kit, PCR2.10 plasmid derivatives, Invitrogen, Carlsbad, CA), and mammalian, yeast or virus vectors such as a baculovirus expression system (pBacPAK plasmid derivatives, Clontech, Palo Alto, CA).
After the vector has been constructed and a nucleic acid molecule encoding an IL-lra-R polypeptide has been inserted into the proper site of the vector, the completed vector may be inserted into a suitable host cell for WO 01/42304 PCT/US00/32940 amplification and/or polypeptide expression. The transformation of an expression vector for an IL-lra-R polypeptide into a selected host cell may be accomplished by well known methods including methods such as transfection, infection, calcium chloride, electroporation, microinjection, lipofection, DEAE-dextran method, or other known techniques. The method selected will in part be a function of the type of host cell to be used. These methods and other suitable methods are well known to the skilled artisan, and are set forth, for example, in Sambrook et al., supra.
Host cells may be prokaryotic host cells (such as E. coli) or eukaryotic host cells (such as a yeast, insect, or vertebrate cell). The host cell, when cultured under appropriate conditions, synthesizes an IL-lra-R polypeptide which can subsequently be collected from the culture medium (if the host cell secretes it into the medium) or directly from the host cell producing it (if it is not secreted). The selection of an appropriate host cell will depend upon various factors, such as desired expression levels, polypeptide modifications that are desirable or necessary for activity (such as glycosylation or phosphorylation) and ease of folding into a biologically active molecule.
A number of suitable host cells are known in the art and many are available from the American Type Culture Collection (ATCC), Manassas, VA.
Examples include, but are not limited to, mammalian cells, such as Chinese hamster ovary cells (CHO), CHO DHFR(-) cells (Urlaub et al., 1980, Proc. Natl.
Acad. Sci. US.A. 97:4216-20), human embryonic kidney (HEK) 293 or 293T cells, or 3T3 cells. The selection of suitable mammalian host cells and methods for transformation, culture, amplification, screening, product production, and purification are known in the art. Other suitable mammalian cell lines, are the monkey COS-I and COS-7 cell lines, and the CV-I cell line. Further exemplary mammalian host cells include primate cell lines and rodent cell lines, including transformed cell lines. Normal diploid cells, cell strains derived from in vitro culture of primary tissue, as well as primary explants, are also suitable. Candidate cells may be genotypically deficient in the selection gene, or may contain a dominantly acting selection gene. Other suitable mammalian cell lines include but 47 WO 01/42304 PCT/US00/32940 are not limited to, mouse neuroblastoma N2A cells, HeLa, mouse L-929 cells, 3T3 lines derived from Swiss, Balb-c or NIH mice, BHK or HaK hamster cell lines.
Each of these cell lines is known by and available to those skilled in the art of protein expression.
Similarly useful as host cells suitable for the present invention are bacterial cells. For example, the various strains of E. coli HBI01, and MC1061) are well-known as host cells in the field of biotechnology.
Various strains of B. subtilis, Pseudomonas spp., other Bacillus spp., Streptomyces spp., and the like may also be employed in this method.
Many strains of yeast cells known to those skilled in the art are also available as host cells for the expression of the polypeptides of the present invention. Preferred yeast cells include, for example, Saccharomyces cerivisae and Pichia pastoris.
Additionally, where desired, insect cell systems may be utilized in the methods of the present invention. Such systems are described, for example, in Kitts et al., 1993, Biotechniques, 14:810-17; Lucklow, 1993, Curr. Opin.
Biotechnol. 4:564-72; and Lucklow et al., 1993, J. Virol., 67:4566-79. Preferred insect cells are Sf-9 and Hi5 (Invitrogen).
One may also use transgenic animals to express glycosylated IL-lra-R polypeptides. For example, one may use a transgenic milk-producing animal (a cow or goat, for example) and obtain the present glycosylated polypeptide in the animal milk. One may also use plants to produce IL-lra-R polypeptides, however, in general, the glycosylation occurring in plants is different from that produced in mammalian cells, and may result in a glycosylated product which is not suitable for human therapeutic use.
Polypeptide Production Host cells comprising an IL-lra-R polypeptide expression vector may be cultured using standard media well known to the skilled artisan. The media will usually contain all nutrients necessary for the growth and survival of the cells.
Suitable media for culturing E. coli cells include, for example, Luria Broth (LB) 48 WO 01/42304 PCT/US00/32940 and/or Terrific Broth Suitable media for culturing eukaryotic cells include Roswell Park Memorial Institute medium 1640 (RPMI 1640), Minimal Essential Medium (MEM) and/or Dulbecco's Modified Eagle Medium (DMEM), all of which may be supplemented with serum and/or growth factors as necessary for the particular cell line being cultured. A suitable medium for insect cultures is Grace's medium supplemented with yeastolate, lactalbumin hydrolysate, and/or fetal calf serum as necessary.
Typically, an antibiotic or other compound useful for selective growth of transfected or transformed cells is added as a supplement to the media. The compound to be used will be dictated by the selectable marker element present on the plasmid with which the host cell was transformed. For example, where the selectable marker element is kanamycin resistance, the compound added to the culture medium will be kanamycin. Other compounds for selective growth include ampicillin, tetracycline, and neomycin.
The amount of an IL-lra-R polypeptide produced by a host cell can be evaluated using standard methods known in the art. Such methods include, without limitation, Western blot analysis, SDS-polyacrylamide gel electrophoresis, non-denaturing gel electrophoresis, High Performance Liquid Chromatography (HPLC) separation, immunoprecipitation, and/or activity assays such as DNA binding gel shift assays.
If an IL-Ira-R polypeptide has been designed to be secreted from the host cells, the majority of polypeptide may be found in the cell culture medium. If however, the IL-lra-R polypeptide is not secreted from the host cells, it will be present in the cytoplasm and/or the nucleus (for eukaryotic host cells) or in the cytosol (for gram-negative bacteria host cells).
For an IL-lra-R polypeptide situated in the host cell cytoplasm and/or nucleus (for eukaryotic host cells) or in the cytosol (for bacterial host cells), the intracellular material (including inclusion bodies for gram-negative bacteria) can be extracted from the host cell using any standard technique known to the skilled artisan. For example, the host cells can be lysed to release the contents of the periplasm/cytoplasm by French press, homogenization, and/or sonication 49 WO 01/42304 PCT/US00/32940 followed by centrifugation.
If an IL-lra-R polypeptide has formed inclusion bodies in the cytosol, the inclusion bodies can often bind to the inner and/or outer cellular membranes and thus will be found primarily in the pellet material after centrifugation. The pellet material can then be treated at pH extremes or with a chaotropic agent such as a detergent, guanidine, guanidine derivatives, urea, or urea derivatives in the presence of a reducing agent such as dithiothreitol at alkaline pH or tris carboxyethyl phosphine at acid pH to release, break apart, and solubilize the inclusion bodies. The solubilized IL-lra-R polypeptide can then be analyzed using gel electrophoresis, immunoprecipitation, or the like. If it is desired to isolate the IL-lra-R polypeptide, isolation may be accomplished using standard methods such as those described herein and in Marston et al., 1990, Meth. Enz., 182:264-75.
In some cases, an IL-lra-R polypeptide may not be biologically active upon isolation. Various methods for "refolding" or converting the polypeptide to its tertiary structure and generating disulfide linkages can be used to restore biological activity. Such methods include exposing the solubilized polypeptide to a pH usually above 7 and in the presence of a particular concentration of a chaotrope. The selection of chaotrope is very similar to the choices used for inclusion body solubilization, but usually the chaotrope is used at a lower concentration and is not necessarily the same as chaotropes used for the solubilization. In most cases the refolding/oxidation solution will also contain a reducing agent or the reducing agent plus its oxidized form in a specific ratio to generate a particular redox potential allowing for disulfide shuffling to occur in the formation of the protein's cysteine bridges. Some of the commonly used redox couples include cysteine/cystamine, glutathione (GSH)/dithiobis GSH, cupric chloride, dithiothreitol(DTT)/dithiane DTT, and 2-2mercaptoethanol(bME)/dithio-b(ME). In many instances, a cosolvent may be used or may be needed to increase the efficiency of the refolding, and the more common reagents used for this purpose include glycerol, polyethylene glycol of various molecular weights, arginine and the like.
WO 01/42304 PCT/US00/32940 If inclusion bodies are not formed to a significant degree upon expression of an IL-Ira-R polypeptide, then the polypeptide will be found primarily in the supernatant after centrifugation of the cell homogenate. The polypeptide may be further isolated from the supernatant using methods such as those described herein.
The purification of an IL-lra-R polypeptide from solution can be accomplished using a variety of techniques. If the polypeptide has been synthesized such that it contains a tag such as Hexahistidine (IL-lra-R polypeptide/hexaHis) or other small peptide such as FLAG (Eastman Kodak Co., New Haven, CT) or myc (Invitrogen, Carlsbad, CA) at either its carboxyl- or amino-terminus, it may be purified in a one-step process by passing the solution through an affinity column where the column matrix has a high affinity for the tag.
For example, polyhistidine binds with great affinity and specificity to nickel. Thus, an affinity column of nickel (such as the Qiagen® nickel columns) can be used for purification of IL-lra-R polypeptide/polyHis. See, Current Protocols in Molecular Biology 10.11.8 (Ausubel et al., eds., Green Publishers Inc. and Wiley and Sons 1993).
Additionally, IL- RA-R polypeptides may be purified through the use of a monoclonal antibody that is capable of specifically recognizing and binding to an IL-Ira-R polypeptide.
Other suitable procedures for purification include, without limitation, affinity chromatography, immunoaffinity chromatography, ion exchange chromatography, molecular sieve chromatography, HPLC, electrophoresis (including native gel electrophoresis) followed by gel elution, and preparative isoelectric focusing ("Isoprime" machine/technique, Hoefcr Scientific, San Francisco, CA). In some cases, two or more purification techniques may be combined to achieve increased purity.
IL-lra-R polypeptides may also be prepared by chemical synthesis methods (such as solid phase peptide synthesis) using techniques known in the art such as those set forth by Merrifield et al., 1963, J. Am. Chem. Soc. 85:2149; WO 01/42304 PCT/US00/32940 Houghten et al., 1985, Proc Nati Acad. Sci. USA 82:5132; and Stewart and Young, Solid Phase Peptide Synthesis (Pierce Chemical Co. 1984). Such polypeptides may be synthesized with or without a methionine on the aminoterminus. Chemically synthesized IL-Ira-R polypeptides may be oxidized using methods set forth in these references to form disulfide bridges. Chemically synthesized IL-lra-R polypeptides are expected to have comparable biological activity to the corresponding IL-lra-R polypeptides produced recombinantly or purified from natural sources, and thus may be used interchangeably with a recombinant or natural IL-lra-R polypeptide.
Another means of obtaining IL-lra-R polypeptide is via purification from biological samples such as source tissues and/or fluids in which the IL-lra-R polypeptide is naturally found. Such purification can be conducted using methods for protein purification as described herein. The presence of the IL-lra-R polypeptide during purification may be monitored, for example, using an antibody prepared against recombinantly produced IL-lra-R polypeptide or peptide fragments thereof.
A number of additional methods for producing nucleic acids and polypeptides are known in the art, and the methods can be used to produce polypeptides having specificity for IL-lra-R polypeptide. See, Roberts et al., 1997, Proc. Natl. Acad. Sci. U.S.A. 94:12297-303, which describes the production of fusion proteins between an mRNA and its encoded peptide. See also. Roberts, 1999, Curr. Opin. Chem. Biol. 3:268-73. Additionally, U.S. Patent No. 5,824,469 describes methods for obtaining oligonucleotides capable of carrying out a specific biological function. The procedure involves generating a heterogeneous pool of oligonucleotides, each having a 5' randomized sequence, a central preselected sequence, and a 3' randomized sequence. The resulting heterogeneous pool is introduced into a population of cells that do not exhibit the desired biological function. Subpopulations of the cells are then screened for those that exhibit a predetermined biological function. From that subpopulation, oligonucleotides capable of carrying out the desired biological function are isolated.
WO 01/42304 PCT/US00/32940 U.S. Patent Nos. 5,763,192; 5,814,476; 5,723,323; and 5,817,483 describe processes for producing peptides or polypeptides. This is done by producing stochastic genes or fragments thereof, and then introducing these genes into host cells which produce one or more proteins encoded by the stochastic genes. The host cells are then screened to identify those clones producing peptides or polypeptides having the desired activity.
Another method for producing peptides or polypeptides is described in PCT/US98/20094 (W099/15650) filed by Athersys, Inc. Known as "Random Activation of Gene Expression for Gene Discovery" (RAGE-GD), the process involves the activation of endogenous gene expression or over-expression of a gene by in situ recombination methods. For example, expression of an endogenous gene is activated or increased by integrating a regulatory sequence into the target cell which is capable of activating expression of the gene by nonhomologous or illegitimate recombination. The target DNA is first subjected to radiation, and a genetic promoter inserted. The promoter eventually locates a break at the front of a gene, initiating transcription of the gene. This results in expression of the desired peptide or polypeptide.
It will be appreciated that these methods can also be used to create comprehensive IL-lra-R polypeptide expression libraries, which can subsequently be used for high throughput phenotypic screening in a variety of assays, such as biochemical assays, cellular assays, and whole organism assays plant, mouse, etc.).
Synthesis It will be appreciated by those skilled in the art that the nucleic acid and polypeptide molecules described herein may be produced by recombinant and other means.
Selective Binding Agents The term "selective binding agent" refers to a molecule that has specificity for one or more IL-lra-R polypeptides. Suitable selective binding agents include, WO 01/42304 PCT/US00/32940 but are not limited to, antibodies and derivatives thereof, polypeptides, and small molecules. Suitable selective binding agents may be prepared using methods known in the art. An exemplary IL-lRA-R polypeptide selective binding agent of the present invention is capable of binding a certain portion of the IL-IRA-R polypeptide thereby inhibiting the binding of the polypeptide to an IL-lra-R polypeptide receptor.
Selective binding agents such as antibodies and antibody fragments that bind IL-lra-R polypeptides are within the scope of the present invention. The antibodies may be polyclonal including monospecific polyclonal; monoclonal (MAbs); recombinant; chimeric; humanized, such as CDR-grafted; human; single chain; and/or bispecific; as well as fragments; variants; or derivatives thereof.
Antibody fragments include those portions of the antibody that bind to an epitope on the IL-IRA-R polypeptide. Examples of such fragments include Fab and F(ab') fragments generated by enzymatic cleavage of full-length antibodies.
Other binding fragments include those generated by recombinant DNA techniques, such as the expression of recombinant plasmids containing nucleic acid sequences encoding antibody variable regions.
Polyclonal antibodies directed toward an IL-lra-R polypeptide generally are produced in animals rabbits or mice) by means of multiple subcutaneous or intraperitoneal injections of IL-Ira-R polypeptide and an adjuvant. It may be useful to conjugate an IL-lra-R polypeptide to a carrier protein that is immunogenic in the species to be immunized, such as keyhole limpet hemocyanin, serum, albumin, bovine thyroglobulin, or soybean trypsin inhibitor.
Also, aggregating agents such as alum are used to enhance the immune response.
After immunization, the animals are bled and the serum is assayed for anti-IL-lra- R antibody titer.
Monoclonal antibodies directed toward IL-lra-R polypeptides are produced using any method that provides for the production of antibody molecules by continuous cell lines in culture. Examples of suitable methods for preparing monoclonal antibodies include the hybridoma methods of Kohler et al., 1975, Nature 256:495-97 and the human B-cell hybridoma method (Kozbor, 54 WO 01/42304 PCT/US0O/32940 1984, J. Immunol. 133:3001; Brodeur et al., Monoclonal Antibody Production Techniques and Applications 51-63 (Marcel Dekker, Inc., 1987). Also provided by the invention are hybridoma cell lines that produce monoclonal antibodies reactive with 1L- ra-R polypeptides.
Monoclonal antibodies of the invention may be modified for use as therapeutics. One embodiment is a "chimeric" antibody in which a portion of the heavy and/or light chain is identical with or homologous to a corresponding sequence in antibodies derived from a particular species or belonging to a particular antibody class or subclass, while the remainder of the chain(s) is/are identical with or homologous to a corresponding sequence in antibodies derived from another species or belonging to another antibody class or subclass. Also included are fragments of such antibodies, so long as they exhibit the desired biological activity. See U.S. Patent No. 4,816,567; Morrison et al., 1985, Proc. Natl. Acad. Sci. 81:6851-55.
In another embodiment, a monoclonal antibody of the invention is a "humanized" antibody. Methods for humanizing non-human antibodies are well known in the art. See U.S. Patent Nos. 5,585,089 and 5,693,762. Generally, a humanized antibody has one or more amino acid residues introduced into it from a source that is non-human. Humanization can be performed, for example, using methods described in the art (Jones et al., 1986, Nature 321:522-25; Riechmann et al., 1998, Nature 332:323-27; Verhoeyen et al., 1988, Science 239:1534-36), by substituting at least a portion of a rodent complementarity-determining region (CDR) for the corresponding regions of a human antibody.
Also encompassed by the invention are human antibodies that bind IL-Ira- R polypeptides. Using transgenic animals mice) that are capable of producing a repertoire of human antibodies in the absence of endogenous immunoglobulin production such antibodies are produced by immunization with an IL-Ira-R polypeptide antigen having at least 6 contiguous amino acids), optionally conjugated to a carrier. See, Jakobovits et al., 1993, Proc. Natl.
Acad. Sci. 90:2551-55; Jakobovits et al., 1993, Nature 362:255-58; Bruggermann et al., 1993, Year in Immuno. 7:33. In one method, such transgenic animals are WO 01/42304 PCT/US00/32940 produced by incapacitating the endogenous loci encoding the heavy and light immunoglobulin chains therein, and inserting loci encoding human heavy and light chain proteins into the genome thereof. Partially modified animals, that is those having less than the full complement of modifications, are then cross-bred to obtain an animal having all of the desired immune system modifications. When administered an immunogen, these transgenic animals produce antibodies with human (rather than, murine) amino acid sequences, including variable regions which are immunospecific for these antigens. See PCT App. Nos.
PCT/US96/05928 and PCT/US93/06926. Additional methods are described in U.S. Patent No. 5,545,807, PCT App. Nos. PCT/US91/245 and PCT/GB89/01207, and in European Patent Nos. 546073B1 and 546073A1. Human antibodies can also be produced by the expression of recombinant DNA in host cells or by expression in hybridoma cells as described herein.
In an alternative embodiment, human antibodies can also be produced from phage-display libraries (Hoogenboom et al., 1991, J. Mol. Biol. 227:381; Marks et al., 1991, J. Mol. Biol. 222:581). These processes mimic immune selection through the display of antibody repertoires on the surface of filamentous bacteriophage, and subsequent selection of phage by their binding to an antigen of choice. One such technique is described in PCT App. No. PCT/US98/17364, which describes the isolation of high affinity and functional agonistic antibodies for MPL- and msk- receptors using such an approach.
Chimeric, CDR grafted, and humanized antibodies are typically produced by recombinant methods. Nucleic acids encoding the antibodies are introduced into host cells and expressed using materials and procedures described herein. In a preferred embodiment, the antibodies are produced in mammalian host cells, such as CHO cells. Monoclonal human) antibodies may be produced by the expression of recombinant DNA in host cells or by expression in hybridoma cells as described herein.
The anti-IL-lra-R antibodies of the invention may be employed in any known assay method, such as competitive binding assays, direct and indirect sandwich assays, and immunoprecipitation assays (Sola, Monoclonal Antibodies: 56 WO 01/42304 PCT/US00/32940 A Manual of Techniques 147-158 (CRC Press, Inc., 1987)) for the detection and quantitation of IL-lra-R polypeptides. The antibodies will bind IL-lra-R polypeptides with an affinity that is appropriate for the assay method being employed.
For diagnostic applications, in certain embodiments, anti-IL-lra-R antibodies may be labeled with a detectable moiety. The detectable moiety can be any one that is capable of producing, either directly or indirectly, a detectable signal. For example, the detectable moiety may be a radioisotope, such as 3 H, 14C, 32 P, 35 S, 9Tc, or 67 Ga; a fluorescent or chemiluminescent compound, such as fluorescein isothiocyanate, rhodamine, or luciferin; or an enzyme, such as alkaline phosphatase, p-galactosidase, or horseradish peroxidase (Bayer, et al., 1990, Meth. Enz. 184:138-63).
Competitive binding assays rely on the ability of a labeled standard an IL-lra-R polypeptide, or an immunologically reactive portion thereof) to compete with the test sample analyte (an IL-lra-R polypeptide) for binding with a limited amount of anti-IL-lra-R antibody. The amount of an IL-lra-R polypeptide in the test sample is inversely proportional to the amount of standard that becomes bound to the antibodies. To facilitate determining the amount of standard that becomes bound, the antibodies typically are insolubilized before or after the competition, so that the standard and analyte that are bound to the antibodies may conveniently be separated from the standard and analyte which remain unbound.
Sandwich assays typically involve the use of two antibodies, each capable of binding to a different immunogenic portion, or epitope, of the protein to be detected and/or quantitated. In a sandwich assay, the test sample analyte is typically bound by a first antibody which is immobilized on a solid support, and thereafter a second antibody binds to the analyte, thus forming an insoluble threepart complex. See, U.S. Patent No. 4,376,110. The second antibody may itself be labeled with a detectable moiety (direct sandwich assays) or may be measured using an anti-immunoglobulin antibody that is labeled with a detectable moiety (indirect sandwich assays). For example, one type of sandwich assay is an 57 WO 01/42304 PCT/US00/32940 enzyme-linked immunosorbent assay (ELISA), in which case the detectable moiety is an enzyme.
The selective binding agents, including anti-IL-Ira-R antibodies, are also useful for in vivo imaging. An antibody labeled with a detectable moiety may be administered to an animal, preferably into the bloodstream, and the presence and location of the labeled antibody in the host assayed. The antibody may be labeled with any moiety that is detectable in an animal, whether by nuclear magnetic resonance, radiology, or other detection means known in the art.
Selective binding agents of the invention, including antibodies, may be used as therapeutics. These therapeutic agents are generally agonists or antagonists, in that they either enhance or reduce, respectively, at least one of the biological activities of an IL-lra-R polypeptide. In one embodiment, antagonist antibodies of the invention are antibodies or binding fragments thereof which are capable of specifically binding to an IL-lra-R polypeptide and which are capable of inhibiting or eliminating the functional activity of an IL-lra-R polypeptide in vivo or in vitro. In preferred embodiments, the selective binding agent, an antagonist antibody, will inhibit the functional activity of an IL-lra-R polypeptide by at least about 50%, and preferably by at least about 80%. In another embodiment, the selective binding agent may be an anti-IL-Ira-R polypeptide antibody that is capable of interacting with an IL-lra-R polypeptide binding partner (a ligand or receptor) thereby inhibiting or eliminating IL-lra-R polypeptide activity in vitro or in vivo. Selective binding agents, including agonist and antagonist anti-IL-lra-R polypeptide antibodies, are identified by screening assays that are well known in the art.
The invention also relates to a kit comprising IL-lra-R selective binding agents (such as antibodies) and other reagents useful for detecting IL-lra-R polypeptide levels in biological samples. Such reagents may include a detectable label, blocking serum, positive and negative control samples, and detection reagents.
Microarrays WO 01/42304 PCT/US00/32940 It will be appreciated that DNA microarray technology can be utilized in accordance with the present invention. DNA microarrays are miniature, highdensity arrays of nucleic acids positioned on a solid support, such as glass. Each cell or element within the array contains numerous copies of a single nucleic acid species that acts as a target for hybridization with a complementary nucleic acid sequence mRNA). In expression profiling using DNA microarray technology, mRNA is first extracted from a cell or tissue sample and then converted enzymatically to fluorescently labeled cDNA. This material is hybridized to the microarray and unbound cDNA is removed by washing. The expression of discrete genes represented on the array is then visualized by quantitating the amount of labeled cDNA that is specifically bound to each target nucleic acid molecule. In this way, the expression of thousands of genes can be quantitated in a high throughput, parallel manner from a single sample of biological material.
This high throughput expression profiling has a broad range of applications with respect to the IL-lra-R molecules of the invention, including, but not limited to: the identification and validation of IL-lra-R disease-related genes as targets for therapeutics; molecular toxicology of related IL-lra-R molecules and inhibitors thereof; stratification of populations and generation of surrogate markers for clinical trials; and enhancing related IL-lra-R polypeptide small molecule drug discovery by aiding in the identification of selective compounds in high throughput screens.
Chemical Derivatives Chemically modified derivatives of IL-lra-R polypeptides may be prepared by one skilled in the art, given the disclosures described herein. IL-lra- R polypeptide derivatives are modified in a manner that is different either in the type or location of the molecules naturally attached to the polypeptide.
Derivatives may include molecules formed by the deletion of one or more naturally-attached chemical groups. The polypeptide comprising the amino acid sequence of any of SEQ ID NO: 2, SEQ ID NO: 4, SEQ ID NO: 6, or SEQ ID 59 WO 01/42304 PCT/US00/32940 NO: 36, or other IL-lra-R polypeptide, may be modified by the covalent attachment of one or more polymers. For example, the polymer selected is typically water-soluble so that the protein to which it is attached does not precipitate in an aqueous environment, such as a physiological environment.
Included within the scope of suitable polymers is a mixture of polymers.
Preferably, for therapeutic use of the end-product preparation, the polymer will be pharmaceutically acceptable.
The polymers each may be of any molecular weight and may be branched or unbranched. The polymers each typically have an average molecular weight of between about 2 kDa to about 100 kDa (the term "about" indicating that in preparations of a water-soluble polymer, some molecules will weigh more, some less, than the stated molecular weight). The average molecular weight of each polymer is preferably between about 5 kDa and about 50 kDa, more preferably between about 12 kDa and about 40 kDa and most preferably between about kDa and about 35 kDa.
Suitable water-soluble polymers or mixtures thereof include, but are not limited to, N-linked or O-linked carbohydrates, sugars, phosphates, polyethylene glycol (PEG) (including the forms of PEG that have been used to derivatize proteins, including mono-(Ci-Clo), alkoxy-, or aryloxy-polyethylene glycol), monomethoxy-polyethylene glycol, dextran (such as low molecular weight dextran of, for example, about 6 kD), cellulose, or other carbohydrate based polymers, poly-(N-vinyl pyrrolidone) polyethylene glycol, propylene glycol homopolymers, polypropylene oxide/ethylene oxide co-polymers, polyoxyethylated polyols glycerol), and polyvinyl alcohol. Also encompassed by the present invention are bifunctional crosslinking molecules which may be used to prepare covalently attached IL-lra-R polypeptide multimers.
In general, chemical derivatization may be performed under any suitable condition used to react a protein with an activated polymer molecule. Methods for preparing chemical derivatives of polypeptides will generally comprise the steps of: reacting the polypeptide with the activated polymer molecule (such as a WO 01/42304 PCT/US00/32940 reactive ester or aldehyde derivative of the polymer molecule) under conditions whereby the polypcptide comprising the amino acid sequence of any of SEQ ID NO: 2, SEQ ID NO: 4, SEQ ID NO: 6, or SEQ ID NO: 36, or other IL-lra-R polypeptide, becomes attached to one or more polymer molecules,, and (b) obtaining the reaction products. The optimal reaction conditions will be determined based on known parameters and the desired result. For example, the larger the ratio of polymer molecules to protein, the greater the percentage of attached polymer molecule. In one embodiment, the IL-Ira-R polypeptide derivative may have a single polymer molecule moiety at the amino-terminus.
See, U.S. Patent No. 5,234,784.
The pegylation of a polypeptide may be specifically carried out using any of the pegylation reactions known in the art. Such reactions are described, for example, in the following references: Francis et al., 1992, Focus on Growth Factors 3:4-10; European Patent Nos. 0154316 and 0401384; and U.S. Patent No.
4,179,337. For example, pegylation may be carried out via an acylation reaction or an alkylation reaction with a reactive polyethylene glycol molecule (or an analogous reactive water-soluble polymer) as described herein. For the acylation reactions, a selected polymer should have a single reactive ester group. For reductive alkylation, a selected polymer should have a single reactive aldehyde group. A reactive aldehyde is, for example, polyethylene glycol propionaldehyde, which is water stable, or mono Ci-Clo alkoxy or aryloxy derivatives thereof (see U.S. Patent No. 5,252,714).
In another embodiment, IL-Ira-R polypeptides may be chemically coupled to biotin. The biotin/IL-lra-R polypeptide molecules are then allowed to bind to avidin, resulting in tetravalent avidin/biotin/IL-lra-R polypeptide molecules. ILlra-R polypeptides may also be covalently coupled to dinitrophenol (DNP) or trinitrophenol (TNP) and the resulting conjugates precipitated with anti-DNP or anti-TNP-IgM to form decameric conjugates with a valency of Generally, conditions that may be alleviated or modulated by the administration of the present IL-lra-R polypeptide derivatives include those described herein for IL-lra-R polypeptides. However, the IL-lra-R polypeptide 61 WO 01/42304 PCT/US00/32940 derivatives disclosed herein may have additional activities, enhanced or reduced biological activity, or other characteristics, such as increased or decreased halflife, as compared to the non-derivatized molecules.
Genetically Engineered Non-Human Animals Additionally included within the scope of the present invention are nonhuman animals such as mice, rats, or other rodents; rabbits, goats, sheep, or other farm animals, in which the genes encoding native IL-lra-R polypeptide have been disrupted "knocked out") such that the level of expression of IL-lra-R polypeptide is significantly decreased or completely abolished. Such animals may be prepared using techniques and methods such as those described in U.S. Patent No. 5,557,032.
The present invention further includes non-human animals such as mice, rats, or other rodents; rabbits, goats, sheep, or other farm animals, in which either the native form of an IL-lra-R gene for that animal or a heterologous IL-lra-R gene is over-expressed by the animal, thereby creating a "transgenic" animal.
Such transgenic animals may be prepared using well known methods such as those described in U.S. Patent No 5,489,743 and PCT Pub. No. WO 94/28122.
The present invention further includes non-human animals in which the promoter for one or more of the IL-lra-R polypeptides of the present invention is either activated or inactivated by using homologous recombination methods) to alter the level of expression of one or more of the native IL-lra-R polypeptides.
These non-human animals may be used for drug candidate screening. In such screening, the impact of a drug candidate on the animal may be measured.
For example, drug candidates may decrease or increase the expression of the ILlra-R gene. In certain embodiments, the amount of IL-lra-R polypeptide that is produced may be measured after the exposure of the animal to the drug candidate.
Additionally, in certain embodiments, one may detect the actual impact of the drug candidate on the animal. For example, over-expression of a particular gene may result in, or be associated with, a disease or pathological condition. In such cases, one may test a drug candidate's ability to decrease expression of the gene 62 WO 01/42304 PCT/US00/32940 or its ability to prevent or inhibit a pathological condition. In other examples, the production of a particular metabolic product such as a fragment of a polypeptide, may result in, or be associated with, a disease or pathological condition. In such cases, one may test a drug candidate's ability to decrease the production of such a metabolic product or its ability to prevent or inhibit a pathological condition.
Assaying for Other Modulators of IL-lra-R Polypeptide Activity In some situations, it may be desirable to identify molecules that are modulators, agonists or antagonists, of the activity of IL-lra-R polypeptide.
Natural or synthetic molecules that modulate IL-lra-R polypeptide may be identified using one or more screening assays, such as those described herein.
Such molecules may be administered either in an ex vivo manner or in an in vivo manner by injection, or by oral delivery, implantation device, or the like.
"Test molecule" refers to a molecule that is under evaluation for the ability to modulate increase or decrease) the activity of an IL-lra-R polypeptide.
Most commonly, a test molecule will interact directly with an IL-lra-R polypeptide. However, it is also contemplated that a test molecule may also modulate IL-lra-R polypeptide activity indirectly, such as by affecting IL-lra-R gene expression, or by binding to an IL-lra-R polypeptide binding partner receptor or ligand). In one embodiment, a test molecule will bind to an IL-Ira-R polypeptide with an affinity constant of at least about 10 6 M, preferably about 8 M, more preferably about 10" 9 M, and even more preferably about 10 10 M.
Methods for identifying compounds that interact with IL-lra-R polypeptides are encompassed by the present invention. In certain embodiments, an IL-lra-R polypeptide is incubated with a test molecule under conditions that permit the interaction of the test molecule with an IL-lra-R polypeptide, and the extent of the interaction is measured. The test molecule can be screened in a substantially purified form or in a crude mixture.
In certain embodiments, an IL-lra-R polypeptide agonist or antagonist may be a protein, peptide, carbohydrate, lipid, or small molecular weight molecule that interacts with IL-lra-R polypeptide to regulate its activity.
63 WO 01/42304 PCT/US00/32940 Molecules which regulate IL-lra-R polypeptide expression include nucleic acids which are complementary to nucleic acids encoding an IL-lra-R polypeptide, or are complementary to nucleic acids sequences which direct or control the expression of IL-lra-R polypeptide, and which act as anti-sense regulators of expression.
Once a test molecule has been identified as interacting with an IL-lra-R polypeptide, the molecule may be further evaluated for its ability to increase or decrease IL-ira-R polypeptide activity. The measurement of the interaction of a test molecule with IL-lra-R polypeptide may be carried out in several formats, including cell-based binding assays, membrane binding assays, solution-phase assays, and immunoassays. In general, a test molecule is incubated with an ILlra-R polypeptide for a specified period of time, and IL-lra-R polypeptide activity is determined by one or more assays for measuring biological activity.
The interaction of test molecules with IL-lra-R polypeptides may also be assayed directly using polyclonal or monoclonal antibodies in an immunoassay.
Alternatively, modified forms of IL-Ira-R polypeptides containing epitope tags as described herein may be used in solution and immunoassays.
In the event that IL-lra-R polypeptides display biological activity through an interaction with a binding partner a receptor or a ligand), a variety of in vitro assays may be used to measure the binding of an IL-lra-R polypeptide to the corresponding binding partner (such as a selective binding agent, receptor, or ligand). These assays may be used to screen test molecules for their ability to increase or decrease the rate and/or the extent of binding of an IL-lra-R polypeptide to its binding partner. In one assay, an IL-lra-R polypeptide is immobilized in the wells of a microtiter plate. Radiolabeled IL-Ira-R polypeptide binding partner (for example, iodinated IL-Ira-R polypeptide binding partner) and a test molecule can then be added either one at a time (in either order) or simultaneously to the wells. After incubation, the wells can be washed and counted for radioactivity, using a scintillation counter, to determine the extent to which the binding partner bound to the IL-lra-R polypeptide. Typically, a molecule will be tested over a range of concentrations, and a series of control 64 WO 01/42304 PCT/US00/32940 wells lacking one or more elements of the test assays can be used for accuracy in the evaluation of the results. An alternative to this method involves reversing the "positions" of the proteins, immobilizing IL-lra-R polypeptide binding partner to the microtiter plate wells, incubating with the test molecule and radiolabeled IL-lra-R polypeptide, and determining the extent of IL-lra-R polypeptide binding. See, Current Protocols in Molecular Biology, chap. 18 (Ausubel et al., eds., Green Publishers Inc. and Wiley and Sons 1995).
As an alternative to radiolabeling, an IL-lra-R polypeptide or its binding partner may be conjugated to biotin, and the presence of biotinylated protein can then be detected using streptavidin linked to an enzyme, such as horse radish peroxidase (HRP) or alkaline phosphatase which can be detected colorometrically, or by fluorescent tagging of streptavidin. An antibody directed to an IL-lra-R polypeptide or to an IL-lra-R polypeptide binding partner, and which is conjugated to biotin, may also be used for purposes of detection following incubation of the complex with enzyme-linked streptavidin linked to AP or HRP.
A IL-lra-R polypeptide or an IL-lra-R polypeptide binding partner can also be immobilized by attachment to agarose beads, acrylic beads, or other types of such inert solid phase substrates. The substrate-protein complex can be placed in a solution containing the complementary protein and the test compound. After incubation, the beads can be precipitated by centrifugation, and the amount of binding between an IL-lra-R polypeptide and its binding partner can be assessed using the methods described herein. Alternatively, the substrate-protein complex can be immobilized in a column with the test molecule and complementary protein passing through the column. The formation of a complex between an ILlra-R polypeptide and its binding partner can then be assessed using any of the techniques described herein radiolabelling or antibody binding).
Another in vitro assay that is useful for identifying a test molecule which increases or decreases the formation of a complex between an IL-lra-R polypeptide binding protein and an IL-lra-R polypeptide binding partner is a surface plasmon resonance detector system such as the BIAcore assay system WO 01/42304 PCT/US00/32940 (Pharmacia, Piscataway, NJ). The BIAcore system is utilized as specified by the manufacturer. This assay essentially involves the covalent binding of either IL- Ira-R polypeptide or an IL-lra-R polypeptide binding partner to a dextran-coated sensor chip that is located in a detector. The test compound and the other complementary protein can then be injected, either simultaneously or sequentially, into the chamber containing the sensor chip. The amount of complementary protein that binds can be assessed based on the change in molecular mass that is physically associated with the dextran-coated side of the sensor chip, with the change in molecular mass being measured by the detector system.
In some cases, it may be desirable to evaluate two or more test compounds together for their ability to increase or decrease the formation of a complex between an IL-lra-R polypeptide and an IL-lra-R polypeptide binding partner. In these cases, the assays set forth herein can be readily modified by adding such additional test compound(s) either simultaneously with, or subsequent to, the first test compound. The remainder of the steps in the assay are as set forth herein.
In vitro assays such as those described herein may be used advantageously to screen large numbers of compounds for an effect on the formation of a complex between an IL-lra-R polypeptide and IL-lra-R polypeptide binding partner. The assays may be automated to screen compounds generated in phage display, synthetic peptide, and chemical synthesis libraries.
Compounds which increase or decrease the formation of a complex between an IL-lra-R polypeptide and an IL-lra-R polypeptide binding partner may also be screened in cell culture using cells and cell lines expressing either ILlra-R polypeptide or IL-lra-R polypeptide binding partner. Cells and cell lines may be obtained from any mammal, but preferably will be from human or other primate, canine, or rodent sources. The binding of an IL-lra-R polypeptide to cells expressing IL-lra-R polypeptide binding partner at the surface is evaluated in the presence or absence of test molecules, and the extent of binding may be determined by, for example, flow cytometry using a biotinylated antibody to an IL-lra-R polypeptide binding partner. Cell culture assays can be used advantageously to further evaluate compounds that score positive in protein 66 WO 01/42304 PCT/US00/32940 binding assays described herein.
Cell cultures can also be used to screen the impact of a drug candidate.
For example, drug candidates may decrease or increase the expression of the IL- Ira-R gene. In certain embodiments, the amount of IL-lra-R polypeptide or an IL-lra-R polypeptide fragment that is produced may be measured after exposure of the cell culture to the drug candidate. In certain embodiments, one may detect the actual impact of the drug candidate on the cell culture. For example, the overexpression of a particular gene may have a particular impact on the cell culture.
In such cases, one may test a drug candidate's ability to increase or decrease the expression of the gene or its ability to prevent or inhibit a particular impact on the cell culture. In other examples, the production of a particular metabolic product such as a fragment of a polypeptide, may result in, or be associated with, a disease or pathological condition. In such cases, one may test a drug candidate's ability to decrease the production of such a metabolic product in a cell culture.
Internalizing Proteins The tat protein sequence (from HIV) can be used to internalize proteins into a cell. See, Falwell et 1994, Proc. Natl. Acad. Sci. U.S.A. 91:664-68.
For example, an 11 amino acid sequence SEQ ID NO: 18) of the HIV tat protein (termed the "protein transduction domain," or TAT PDT) has been described as mediating delivery across the cytoplasmic membrane and the nuclear membrane of a cell. See Schwarze et al., 1999, Science 285:1569- 72; and Nagahara et al., 1998, Nat. Med. 4:1449-52. In these procedures, FITCconstructs (FITC-labeled G-G-G-G-Y-G-R-K-K-R-R-Q-R-R-R; SEQ ID NO: 19), which penetrate tissues following intraperitoneal administration, are prepared, and the binding of such constructs to cells is detected by fluorescence-activated cell sorting (FACS) analysis. Cells treated with a tat-p-gal fusion protein will demonstrate P-gal activity. Following injection, expression of such a construct can be detected in a number of tissues, including liver, kidney, lung, heart, and brain tissue. It is believed that such constructs undergo some degree of unfolding in order to enter the cell, and as such, may require a refolding following entry into 67 WO 01/42304 PCT/US00/32940 the cell.
It will thus be appreciated that the tat protein sequence may be used to internalize a desired polypeptide into a cell. For example, using the tat protein sequence, an IL-lra-R antagonist (such as an anti-IL-Ira-R selective-binding agent, small molecule, soluble receptor, or antisense oligonucleotide) can be administered intracellularly to inhibit the activity of an IL-lra-R molecule. As used herein, the term "IL-lra-R molecule" refers to both IL-lra-R nucleic acid molecules and IL-lra-R polypeptides as defined herein. Where desired, the IL- Ira-R protein itself may also be internally administered to a cell using these procedures. See also, Straus, 1999, Science 285:1466-67.
Cell Source Identification Using IL-lra-R Polypeptide In accordance with certain embodiments of the invention, it may be useful to be able to determine the source of a certain cell type associated with an IL-Ira- R polypeptide. For example, it may be useful to determine the origin of a disease or pathological condition as an aid in selecting an appropriate therapy. In certain embodiments, nucleic acids encoding an IL-lra-R polypeptide can be used as a probe to identify cells described herein by screening the nucleic acids of the cells with such a probe. In other embodiments, one may use anti-IL-lra-R polypeptide antibodies to test for the presence of IL-lra-R polypeptide in cells, and thus, determine if such cells are of the types described herein.
IL-lra-R Polypeptide Compositions and Administration Therapeutic compositions are within the scope of the present invention.
Such IL-lRA-R polypeptide pharmaceutical compositions may comprise a therapeutically effective amount of an IL-lra-R polypeptide or an IL-lra-R nucleic acid molecule in admixture with a pharmaceutically or physiologically acceptable formulation agent selected for suitability with the mode of administration. Pharmaceutical compositions may comprise a therapeutically effective amount of one or more IL-I ra-R polypeptide selective binding agents in admixture with a pharmaceutically or physiologically acceptable formulation 68 WO 01/42304 PCT/US00/32940 agent selected for suitability with the mode of administration.
Acceptable formulation materials preferably are nontoxic to recipients at the dosages and concentrations employed.
The pharmaceutical composition may contain formulation materials for modifying, maintaining, or preserving, for example, the pH, osmolarity, viscosity, clarity, color, isotonicity, odor, sterility, stability, rate of dissolution or release, adsorption, or penetration of the composition. Suitable formulation materials include, but are not limited to, amino acids (such as glycine, glutamine, asparagine, arginine, or lysine), antimicrobials, antioxidants (such as ascorbic acid, sodium sulfite, or sodium hydrogen-sulfite), buffers (such as borate, bicarbonate, Tris-HCI, citrates, phosphates, or other organic acids), bulking agents (such as mannitol or glycine), chelating agents (such as ethylenediamine tetraacetic acid (EDTA)), complexing agents (such as caffeine, polyvinylpyrrolidone, beta-cyclodextrin, or hydroxypropyl-beta-cyclodextrin), fillers, monosaccharides, disaccharides, and other carbohydrates (such as glucose, mannose, or dextrins), proteins (such as serum albumin, gelatin, or immunoglobulins), coloring, flavoring and diluting agents, emulsifying agents, hydrophilic polymers (such as polyvinylpyrrolidone), low molecular weight polypeptides, salt-forming counterions (such as sodium), preservatives (such as benzalkonium chloride, benzoic acid, salicylic acid, thimerosal, phenethyl alcohol, methylparaben, propylparaben, chlorhexidine, sorbic acid, or hydrogen peroxide), solvents (such as glycerin, propylene glycol, or polyethylene glycol), sugar alcohols (such as mannitol or sorbitol), suspending agents, surfactants or wetting agents (such as pluronics; PEG; sorbitan esters; polysorbates such as polysorbate 20 or polysorbate 80; triton; tromethamine; lecithin; cholesterol or tyloxapal), stability enhancing agents (such as sucrose or sorbitol), tonicity enhancing agents (such as alkali metal halidcs preferably sodium or potassium chloride or mannitol sorbitol), delivery vehicles, diluents, excipients and/or pharmaceutical adjuvants. See Remington's Pharmaceutical Sciences (18th Ed., A.R. Gennaro, ed., Mack Publishing Company 1990.
The optimal pharmaceutical composition will be determined by a skilled 69 WO 01/42304 PCT/US00/32940 artisan depending upon, for example, the intended route of administration, delivery format, and desired dosage. See, Remington's Pharmaceutical Sciences, supra. Such compositions may influence the physical state, stability, rate of in vivo release, and rate of in vivo clearance of the IL-lra-R molecule.
The primary vehicle or carrier in a pharmaceutical composition may be either aqueous or non-aqueous in nature. For example, a suitable vehicle or carrier for injection may be water, physiological saline solution, or artificial cerebrospinal fluid, possibly supplemented with other materials common in compositions for parenteral administration. Neutral buffered saline or saline mixed with serum albumin are further exemplary vehicles. Other exemplary pharmaceutical compositions comprise Tris buffer of about pH 7.0-8.5, or acetate buffer of about pH 4.0-5.5, which may further include sorbitol or a suitable substitute. In one embodiment of the present invention, IL-lra-R polypeptide compositions may be prepared for storage by mixing the selected composition having the desired degree of purity with optional formulation agents (Remington's Pharmaceutical Sciences, supra) in the form of a lyophilized cake or an aqueous solution. Further, the IL-lra-R polypeptide product may be formulated as a lyophilizate using appropriate excipients such as sucrose.
The IL-lra-R polypeptide pharmaceutical compositions can be selected for parenteral delivery. Alternatively, the compositions may be selected for inhalation or for delivery through the digestive tract, such as orally. The preparation of such pharmaceutically acceptable compositions is within the skill of the art.
The formulation components are present in concentrations that are acceptable to the site of administration. For example, buffers are used to maintain the composition at physiological pH or at a slightly lower pH, typically within a pH range of from about 5 to about 8.
When parenteral administration is contemplated, the therapeutic compositions for use in this invention may be in the form of a pyrogen-free, parenterally acceptable, aqueous solution comprising the desired IL-lra-R molecule in a pharmaceutically acceptable vehicle. A particularly suitable vehicle WO 01/42304 PCT/US00/32940 for parenteral injection is sterile distilled water in which an IL-lra-R molecule is formulated as a sterile, isotonic solution, properly preserved. Yet another preparation can involve the formulation of the desired molecule with an agent, such as injectable microspheres, bio-erodible particles, polymeric compounds (such as polylactic acid or polyglycolic acid), beads, or liposomes, that provides for the controlled or sustained release of the product which may then be delivered via a depot injection. Hyaluronic acid may also be used, and this may have the effect of promoting sustained duration in the circulation. Other suitable means for the introduction of the desired molecule include implantable drug delivery devices.
In one embodiment, a pharmaceutical composition may be formulated for inhalation. For example, IL-lra-R polypeptide may be formulated as a dry powder for inhalation. IL-lra-R polypeptide or nucleic acid molecule inhalation solutions may also be formulated with a propellant for aerosol delivery. In yet another embodiment, solutions may be nebulized. Pulmonary administration is further described in PCT Pub. No. WO 94/20069, which describes the pulmonary delivery of chemically modified proteins.
It is also contemplated that certain formulations may be administered orally. In one embodiment of the present invention, IL-Ira-R polypeptides that are administered in this fashion can be formulated with or without those carriers customarily used in the compounding of solid dosage forms such as tablets and capsules. For example, a capsule may be designed to release the active portion of the formulation at the point in the gastrointestinal tract when bioavailability is maximized and pre-systemic degradation is minimized. Additional agents can be included to facilitate absorption of the IL-lra-R polypeptide. Diluents, flavorings, low melting point waxes, vegetable oils, lubricants, suspending agents, tablet disintegrating agents, and binders may also be employed.
Another pharmaceutical composition may involve an effective quantity of IL-lra-R polypeptides in a mixture with non-toxic excipients that are suitable for the manufacture of tablets. By dissolving the tablets in sterile water, or another appropriate vehicle, solutions can be prepared in unit-dose form. Suitable 71 WO 01/42304 PCT/US00/32940 excipients include, but are not limited to, inert diluents, such as calcium carbonate, sodium carbonate or bicarbonate, lactose, or calcium phosphate; or binding agents, such as starch, gelatin, or acacia; or lubricating agents such as magnesium stearate, stearic acid, or talc.
Additional IL-Ira-R polypeptide pharmaceutical compositions will be evident to those skilled in the art, including formulations involving IL-lra-R polypeptides in sustained- or controlled-delivery formulations. Techniques for formulating a variety of other sustained- or controlled-delivery means, such as liposome carriers, bio-erodible microparticles or porous beads and depot injections, are also known to those skilled in the art. See, PCT/US93/00829, which describes the controlled release of porous polymeric microparticles for the delivery of pharmaceutical compositions.
Additional examples of sustained-release preparations include semipermeable polymer matrices in the form of shaped articles, e.g. films, or microcapsules. Sustained release matrices may include polyesters, hydrogels, polylactides Patent No. 3,773,919 and European Patent No. 058481), copolymers of L-glutamic acid and gamma ethyl-L-glutamate (Sidman et al., 1983, Biopolymers 22:547-56), poly(2-hydroxyethyl-methacrylate) (Langer et al., 1981, J. Biomed. Mater. Res. 15:167-277 and Langer, 1982, Chem. Tech. 12:98- 105), ethylene vinyl acetate (Langer et al., supra) or poly-D(-)-3-hydroxybutyric acid (European Patent No. 133988). Sustained-release compositions may also include liposomes, which can be prepared by any of several methods known in the art. See, Eppstein et al., 1985, Proc. Nail. Acad. Sci. USA 82:3688-92; and European Patent Nos. 036676, 088046, and 143949.
The IL-lra-R pharmaceutical composition to be used for in vivo administration typically must be sterile. This may be accomplished by filtration through sterile filtration membranes. Where the composition is lyophilized, sterilization using this method may be conducted either prior to, or following, lyophilization and reconstitution. The composition for parenteral administration may be stored in lyophilized form or in a solution. In addition, parenteral compositions generally are placed into a container having a sterile access port, for 72 WO 01/42304 PCT/US00/32940 example, an intravenous solution bag or vial having a stopper pierceable by a hypodermic injection needle.
Once the pharmaceutical composition has been formulated, it may be stored in sterile vials as a solution, suspension, gel, emulsion, solid, or as a dehydrated or lyophilized powder. Such formulations may be stored either in a ready-to-use form or in a form lyophilized) requiring reconstitution prior to administration.
In a specific embodiment, the present invention is directed to kits for producing a single-dose administration unit. The kits may each contain both a first container having a dried protein and a second container having an aqueous formulation. Also included within the scope of this invention are kits containing single and multi-chambered pre-filled syringes liquid syringes and lyosyringes).
The effective amount of an IL-lra-R pharmaceutical composition to be employed therapeutically will depend, for example, upon the therapeutic context and objectives. One skilled in the art will appreciate that the appropriate dosage levels for treatment will thus vary depending, in part, upon the molecule delivered, the indication for which the IL-lra-R molecule is being used, the route of administration, and the size (body weight, body surface, or organ size) and condition (the age and general health) of the patient. Accordingly, the clinician may titcr the dosage and modify the route of administration to obtain the optimal therapeutic effect. A typical dosage may range from about 0.1 pg/kg to up to about 100 mg/kg or more, depending on the factors mentioned above. In other embodiments, the dosage may range from 0.1 pg/kg up to about 100 mg/kg; or 1 pg/kg up to about 100 mg/kg; or 5 pg/kg up to about 100 mg/kg.
The frequency of dosing will depend upon the pharmacokinetic parameters of the IL-lra-R molecule in the formulation being used. Typically, a clinician will administer the composition until a dosage is reached that achieves the desired effect. The composition may therefore be administered as a single dose, as two or more doses (which may or may not contain the same amount of the desired molecule) over time, or as a continuous infusion via an implantation device or 73 WO 01/42304 PCT/US00/32940 catheter. Further refinement of the appropriate dosage is routinely made by those of ordinary skill in the art and is within the ambit of tasks routinely performed by them. Appropriate dosages may be ascertained through use of appropriate doseresponse data.
The route of administration of the pharmaceutical composition is in accord with known methods, orally; through injection by intravenous, intraperitoneal, intracerebral (intraparenchymal), intracerebroventricular, intramuscular, intraocular, intraarterial, intraportal, or intralesional routes; by sustained release systems; or by implantation devices. Where desired, the compositions may be administered by bolus injection or continuously by infusion, or by implantation device.
Alternatively or additionally, the composition may be administered locally via implantation of a membrane, sponge, or other appropriate material onto which the desired molecule has been absorbed or encapsulated. Where an implantation device is used, the device may be implanted into any suitable tissue or organ, and delivery of the desired molecule may be via diffusion, timed-release bolus, or continuous administration.
In some cases, it may be desirable to use IL-lra-R polypeptide pharmaceutical compositions in an ex vivo manner. In such instances, cells, tissues, or organs that have been removed from the patient are exposed to IL-lra- R polypeptide pharmaceutical compositions after which the cells, tissues, or organs are subsequently implanted back into the patient.
In other cases, an IL-lra-R polypeptide can be delivered by implanting certain cells that have been genetically engineered, using methods such as those described herein, to express and secrete the IL-lra-R polypeptide. Such cells may be animal or human cells, and may be autologous, heterologous, or xenogeneic.
Optionally, the cells may be immortalized. In order to decrease the chance of an immunological response, the cells may be encapsulated to avoid infiltration of surrounding tissues. The encapsulation materials are typically biocompatible, semi-permeable polymeric enclosures or membranes that allow the release of the protein product(s) but prevent the destruction of the cells by the patient's immune 74 WO 01/42304 PCT/US00/32940 system or by other detrimental factors from the surrounding tissues.
As discussed herein, it may be desirable to treat isolated cell populations (such as stem cells, lymphocytes, red blood cells, chondrocytes, neurons, and the like) with one or more IL-lra-R polypeptides. This can be accomplished by exposing the isolated cells to the polypeptide directly, where it is in a form that is permeable to the cell membrane.
Additional embodiments of the present invention relate to cells and methods homologous recombination and/or other recombinant production methods) for both the in vitro production of therapeutic polypeptides and for the production and delivery of therapeutic polypeptides by gene therapy or cell therapy. Homologous and other recombination methods may be used to modify a cell that contains a normally transcriptionally-silent IL-lra-R gene, or an underexpressed gene, and thereby produce a cell which expresses therapeutically efficacious amounts of IL-lra-R polypeptides.
Homologous recombination is a technique originally developed for targeting genes to induce or correct mutations in transcriptionally active genes.
Kucherlapati, 1989, Prog. in Nucl. Acid Res. Mol. Biol. 36:301. The basic technique was developed as a method for introducing specific mutations into specific regions of the mammalian genome (Thomas et al., 1986, Cell 44:419-28; Thomas and Capecchi, 1987, Cell 51:503-12; Doetschman et al., 1988, Proc. Natl.
Acad. Sci. U.S.A. 85:8583-87) or to correct specific mutations within defective genes (Doetschman et al., 1987, Nature 330:576-78). Exemplary homologous recombination techniques are described in U.S. Patent No. 5,272,071; European Patent Nos. 9193051 and 505500; PCT/US90/07642, and PCT Pub No. WO 91/09955).
Through homologous recombination, the DNA sequence to be inserted into the genome can be directed to a specific region of the gene of interest by attaching it to targeting DNA. The targeting DNA is a nucleotide sequence that is complementary (homologous) to a region of the genomic DNA. Small pieces of targeting DNA that are complementary to a specific region of the genome are put in contact with the parental strand during the DNA replication process. It is a WO 01/42304 PCT/USOO/32940 general property of DNA that has been inserted into a cell to hybridize, and therefore, recombine with other pieces of endogenous DNA through shared homologous regions. If this complementary strand is attached to an oligonucleotide that contains a mutation or a different sequence or an additional nucleotide, it too is incorporated into the newly synthesized strand as a result of the recombination. As a result of the proofreading function, it is possible for the new sequence of DNA to serve as the template. Thus, the transferred DNA is incorporated into the genome.
Attached to these pieces of targeting DNA are regions of DNA that may interact with or control the expression of an IL-lra-R polypeptide, flanking sequences. For example, a promoter/enhancer element, a suppressor, or an exogenous transcription modulatory element is inserted in the genome of the intended host cell in proximity and orientation sufficient to influence the transcription of DNA encoding the desired IL-lra-R polypeptide. The control element controls a portion of the DNA present in the host cell genome. Thus, the expression of the desired IL-lra-R polypeptide may be achieved not by transfection of DNA that encodes the IL-lra-R gene itself, but rather by the use of targeting DNA (containing regions of homology with the endogenous gene of interest) coupled with DNA regulatory segments that provide the endogenous gene sequence with recognizable signals for transcription of an IL-1ra-R gene.
In an exemplary method, the expression of a desired targeted gene in a cell a desired endogenous cellular gene) is altered via homologous recombination into the cellular genome at a preselected site, by the introduction of DNA which includes at least a regulatory sequence, an exon, and a splice donor site. These components are introduced into the chromosomal (genomic) DNA in such a manner that this, in effect, results in the production of a new transcription unit (in which the regulatory sequence, the exon, and the splice donor site present in the DNA construct are operatively linked to the endogenous gene). As a result of the introduction of these components into the chromosomal DNA, the expression of the desired endogenous gene is altered.
Altered gene expression, as described herein, encompasses activating (or 76 WO 01/42304 PCT/US00/32940 causing to be expressed) a gene which is normally silent (unexpressed) in the cell as obtained, as well as increasing the expression of a gene which is not expressed at physiologically significant levels in the cell as obtained. The embodiments further encompass changing the pattern of regulation or induction such. that it is different from the pattern of regulation or induction that occurs in the cell as obtained, and reducing (including eliminating) the expression of a gene which is expressed in the cell as obtained.
One method by which homologous recombination can be used to increase, or cause, IL-lra-R polypeptide production from a cell's endogenous IL-lra-R gene involves first using homologous recombination to place a recombination sequence from a site-specific recombination system CrelloxP, FLP/FRT) (Sauer, 1994, Curr. Opin. Biotechnol., 5:521-27; Sauer, 1993, Methods Enzymol., 225:890-900) upstream of 5' to) the cell's endogenous genomic IL-lra-R polypeptide coding region. A plasmid containing a recombination site homologous to the site that was placed just upstream of the genomic IL-lra-R polypeptide coding region is introduced into the modified cell line along with the appropriate recombinase enzyme. This recombinase causes the plasmid to integrate, via the plasmid's recombination site, into the recombination site located just upstream of the genomic IL-lra-R polypeptide coding region in the cell line (Baubonis and Sauer, 1993, Nucleic Acids Res. 21:2025-29; O'Gorman et al., 1991, Science 251:1351-55). Any flanking sequences known to increase transcription enhancer/promoter, intron, translational enhancer), if properly positioned in this plasmid, would integrate in such a manner as to create a new or modified transcriptional unit resulting in de novo or increased IL-lra-R polypeptide production from the cell's endogenous IL-1 ra-R gene.
A further method to use the cell line in which the site specific recombination sequence had been placed just upstream of the cell's endogenous genomic IL-lra-R polypeptide coding region is to use homologous recombination to introduce a second recombination site elsewhere in the cell line's genome. The appropriate recombinase enzyme is then introduced into the two-recombinationsite cell line, causing a recombination event (deletion, inversion, and 77 WO 01/42304 PCT/US00/32940 translocation) (Sauer, 1994, Curr. Opin. Biotechnol., 5:521-27; Sauer, 1993, Methods Enzymol., 225:890-900) that would create a new or modified transcriptional unit resulting in de novo or increased IL-lra-R polypeptide production from the cell's endogenous IL-Ira-R gene.
An additional approach for increasing, or causing, the expression of ILlra-R polypeptide from a cell's endogenous IL-lra-R gene involves increasing, or causing, the expression of a gene or genes transcription factors) and/or decreasing the expression of a gene or genes transcriptional repressors) in a manner which results in de novo or increased IL-lra-R polypeptide production from the cell's endogenous IL-lra-R gene. This method includes the introduction of a non-naturally occurring polypeptide a polypeptide comprising a site specific DNA binding domain fused to a transcriptional factor domain) into the cell such that de novo or increased IL-lra-R polypeptide production from the cell's endogenous IL-lra-R gene results.
The present invention further relates to DNA constructs useful in the method of altering expression of a target gene. In certain embodiments, the exemplary DNA constructs comprise: one or more targeting sequences, a regulatory sequence, an exon, and an unpaired splice-donor site. The targeting sequence in the DNA construct directs the integration of elements into a target gene in a cell such that the elements arc operatively linked to sequences of the endogenous target gene. In another embodiment, the DNA constructs comprise: one or more targeting sequences, a regulatory sequence, an exon, a splice-donor site, an intron, and a spliceacceptor site, wherein the targeting sequence directs the integration of elements (a) such that the elements of(b) are operatively linked to the endogenous gene. The targeting sequence is homologous to the preselected site in the cellular chromosomal DNA with which homologous recombination is to occur. In the construct, the exon is generally 3' of the regulatory sequence and the splice-donor site is 3' of the exon.
If the sequence of a particular gene is known, such as the nucleic acid sequence of IL-lra-R polypeptide presented herein, a piece of DNA that is 78 WO 01/42304 PCT/US00/32940 complementary to a selected region of the gene can be synthesized or otherwise obtained, such as by appropriate restriction of the native DNA at specific recognition sites bounding the region of interest. This piece serves as a targeting sequence upon insertion into the cell and will hybridize to its homologous region within the genome. If this hybridization occurs during DNA replication, this piece of DNA, and any additional sequence attached thereto, will act as an Okazaki fragment and will be incorporated into the newly synthesized daughter strand of DNA. The present invention, therefore, includes nucleotides encoding an IL-lra- R polypeptide, which nucleotides may be used as targeting sequences.
IL- ra-R polypeptide cell therapy, the implantation of cells producing IL-lra-R polypeptides, is also contemplated. This embodiment involves implanting cells capable of synthesizing and secreting a biologically active form of IL-lra-R polypeptide. Such IL-lra-R polypeptide-producing cells can be cells that are natural producers of IL-lra-R polypeptides or may be recombinant cells whose ability to produce IL-lra-R polypeptides has been augmented by transformation with a gene encoding the desired IL-lra-R polypeptide or with a gene augmenting the expression of IL-lra-R polypeptide. Such a modification may be accomplished by means of a vector suitable for delivering the gene as well as promoting its expression and secretion. In order to minimize a potential immunological reaction in patients being administered an IL-lra-R polypeptide, as may occur with the administration of a polypeptide of a foreign species, it is preferred that the natural cells producing IL-lra-R polypeptide be of human origin and produce human IL-lra-R polypeptide. Likewise, it is preferred that the recombinant cells producing IL-lra-R polypeptide be transformed with an expression vector containing a gene encoding a human IL- ra-R polypeptide.
Implanted cells may be encapsulated to avoid the infiltration of surrounding tissue. Human or non-human animal cells may be implanted in patients in biocompatible, semipermeable polymeric enclosures or membranes that allow the release of IL-lra-R polypeptide, but that prevent the destruction of the cells by the patient's immune system or by other detrimental factors from the surrounding tissue. Alternatively, the patient's own cells, transformed to produce 79 WO 01/42304 PCT/US00/32940 IL-lra-R polypeptides ex vivo, may be implanted directly into the patient without such encapsulation.
Techniques for the encapsulation of living cells are known in the art, and the preparation of the encapsulated cells and their implantation in patients may be routinely accomplished. For example, Baetge et al. (PCT Pub. No. WO 95/05452 and PCT/US94/09299) describe membrane capsules containing genetically engineered cells for the effective delivery of biologically active molecules. The capsules are biocompatible and are easily retrievable. The capsules encapsulate cells transfected with recombinant DNA molecules comprising DNA sequences coding for biologically active molecules operatively linked to promoters that are not subject to down-regulation in vivo upon implantation into a mammalian host.
The devices provide for the delivery of the molecules from living cells to specific sites within a recipient. In addition, see U.S. Patent Nos. 4,892,538; 5,011,472; and 5,106,627. A system for encapsulating living cells is described in PCT Pub.
No. WO 91/10425 (Aebischer et See also, PCT Pub. No. WO 91/10470 (Aebischer et Winn et al., 1991, Exper. Neurol. 113:322-29; Aebischer et al., 1991, Exper. Neurol. 111:269-75; and Tresco et al., 1992, ASAIO 38:17-23.
In vivo and in vitro gene therapy delivery of IL- ra-R polypeptides is also envisioned. One example of a gene therapy technique is to use the IL-lra-R gene (either genomic DNA, cDNA, and/or synthetic DNA) encoding an IL-lra-R polypeptide which may be operably linked to a constitutive or inducible promoter to form a "gene therapy DNA construct." The promoter may be homologous or heterologous to the endogenous IL-lra-R gene, provided that it is active in the cell or tissue type into which the construct will be inserted. Other components of the gene therapy DNA construct may optionally include DNA molecules designed for site-specific integration endogenous sequences useful for homologous recombination), tissue-specific promoters, enhancers or silencers, DNA molecules capable of providing a selective advantage over the parent cell, DNA molecules useful as labels to identify transformed cells, negative selection systems, cell specific binding agents (as, for example, for cell targeting), cell-specific internalization factors, transcription factors enhancing expression from a vector, WO 01/42304 PCT/US00/32940 and factors enabling vector production.
A gene therapy DNA construct can then be introduced into cells (either ex vivo or in vivo) using viral or non-viral vectors. One means for introducing the gene therapy DNA construct is by means of viral vectors as described herein.
Certain vectors, such as retroviral vectors, will deliver the DNA construct to the chromosomal DNA of the cells, and the gene can integrate into the chromosomal DNA. Other vectors will function as episomes, and the gene therapy DNA construct will remain in the cytoplasm.
In yet other embodiments, regulatory elements can be included for the controlled expression of the IL-lra-R gene in the target cell. Such elements are turned on in response to an appropriate effector. In this way, a therapeutic polypeptide can be expressed when desired. One conventional control means involves the use of small molecule dimerizers or rapalogs to dimerize chimeric proteins which contain a small molecule-binding domain and a domain capable of initiating a biological process, such as a DNA-binding protein or transcriptional activation protein (see PCT Pub. Nos. WO 96/41865, WO 97/31898, and WO 97/31899). The dimerization of the proteins can be used to initiate transcription of the transgene.
An alternative regulation technology uses a method of storing proteins expressed from the gene of interest inside the cell as an aggregate or cluster. The gene of interest is expressed as a fusion protein that includes a conditional aggregation domain that results in the retention of the aggregated protein in the endoplasmic reticulum. The stored proteins are stable and inactive inside the cell.
The proteins can be released, however, by administering a drug small molecule ligand) that removes the conditional aggregation domain and thereby specifically breaks apart the aggregates or clusters so that the proteins may be secreted from the cell. See Aridor et al., 2000, Science 287:816-17 and Rivcra et al., 2000, Science 287:826-30.
Other suitable control means or gene switches include, but are not limited to, the systems described herein. Mifepristone (RU486) is used as a progesterone antagonist. The binding of a modified progesterone receptor ligand-binding 81 WO 01/42304 PCT/US00/32940 domain to the progesterone antagonist activates transcription by forming a dimer of two transcription factors that then pass into the nucleus to bind DNA. The ligand-binding domain is modified to eliminate the ability of the receptor to bind to the natural ligand. The modified steroid hormone receptor system is further described in U.S. Patent No. 5,364,791 and PCT Pub. Nos. WO 96/40911 and WO 97/10337.
Yet another control system uses ecdysone (a fruit fly steroid hormone) which binds to and activates an ecdysone receptor (cytoplasmic receptor). The receptor then translocates to the nucleus to bind a specific DNA response element (promoter from ecdysone-responsive gene). The ecdysone receptor includes a transactivation domain, DNA-binding domain, and ligand-binding domain to initiate transcription. The ecdysone system is further described in U.S. Patent No.
5,514,578 and PCT Pub. Nos. WO 97/38117, WO 96/37609, and WO 93/03162.
Another control means uses a positive tetracycline-controllable transactivator. This system involves a mutated tet repressor protein DNA-binding domain (mutated tet R-4 amino acid changes which resulted in a reverse tetracycline-regulated transactivator protein, it binds to a tet operator in the presence of tetracycline) linked to a polypeptide which activates transcription.
Such systems are described in U.S. Patent Nos. 5,464,758, 5,650,298, and 5,654,168.
Additional expression control systems and nucleic acid constructs are described in U.S. Patent Nos. 5,741,679 and 5,834,186, to Innovir Laboratories Inc.
In vivo gene therapy may be accomplished by introducing the gene encoding IL-lra-R polypeptide into cells via local injection of an IL-lra-R nucleic acid molecule or by other appropriate viral or non-viral delivery vectors. Hefti, 1994, Neurobiology 25:1418-35. For example, a nucleic acid molecule encoding an IL-lra-R polypeptide may be contained in an adeno-associated virus (AAV) vector for delivery to the targeted cells (see, Johnson, PCT Pub. No. WO 95/34670; PCT App. No. PCT/US95/07178). The recombinant AAV genome typically contains AAV inverted terminal repeats flanking a DNA sequence 82 WO 01/42304 PCT/US00/32940 encoding an IL-lra-R polypeptide operably linked to functional promoter and polyadenylation sequences.
Alternative suitable viral vectors include, but are not limited to, retrovirus, adenovirus, herpes simplex virus, lentivirus, hepatitis virus, parvovirus, papovavirus, poxvirus, alphavirus, coronavirus, rhabdovirus, paramyxovirus, and papilloma virus vectors. U.S. Patent No. 5,672,344 describes an in vivo viralmediated gene transfer system involving a recombinant neurotrophic HSV-1 vector. U.S. Patent No. 5,399,346 provides examples of a process for providing a patient with a therapeutic protein by the delivery of human cells which have been treated in vitro to insert a DNA segment encoding a therapeutic protein.
Additional methods and materials for the practice of gene therapy techniques are described in U.S. Patent Nos. 5,631,236 (involving adenoviral vectors), 5,672,510 (involving retroviral vectors), 5,635,399 (involving retroviral vectors expressing cytokines).
Nonviral delivery methods include, but are not limited to, liposomemediated transfer, naked DNA delivery (direct injection), receptor-mediated transfer (ligand-DNA complex), electroporation, calcium phosphate precipitation, and microparticle bombardment gene gun). Gene therapy materials and methods may also include inducible promoters, tissue-specific enhancerpromoters, DNA sequences designed for site-specific integration, DNA sequences capable of providing a selective advantage over the parent cell, labels to identify transformed cells, negative selection systems and expression control systems (safety measures), cell-specific binding agents (for cell targeting), cell-specific internalization factors, and transcription factors to enhance expression by a vector as well as methods of vector manufacture. Such additional methods and materials for the practice of gene therapy techniques are described in U.S. Patent Nos.
4,970,154 (involving electroporation techniques), 5,679,559 (describing a lipoprotein-containing system for gene delivery), 5,676,954 (involving liposome carriers), 5,593,875 (describing methods for calcium phosphate transfection), and 4,945,050 (describing a process wherein biologically active particles are propelled at cells at a speed whereby the particles penetrate the surface of the cells and 83 WO 01/42304 PCT/US00/32940 become incorporated into the interior of the cells), and PCT Pub. No. WO 96/40958 (involving nuclear ligands).
It is also contemplated that IL-lra-R gene therapy or cell therapy can further include the delivery of one or more additional polypeptide(s) in the same or a different cell(s). Such cells may be separately introduced into the patient, or the cells may be contained in a single implantable device, such as the encapsulating membrane described above, or the cells may be separately modified by means of viral vectors.
A means to increase endogenous IL-Ira-R polypeptide expression in a cell via gene therapy is to insert one or more enhancer elements into the IL-lra-R polypeptide promoter, where the enhancer elements can serve to increase transcriptional activity of the IL-lra-R gene. The enhancer elements used will be selected based on the tissue in which one desires to activate the gene enhancer elements known to confer promoter activation in that tissue will be selected. For example, if a gene encoding an IL-lra-R polypeptide is to be "turned on" in Tcells, the Ick promoter enhancer element may be used. Here, the functional portion of the transcriptional element to be added may be inserted into a fragment of DNA containing the IL-lra-R polypeptide promoter (and optionally, inserted into a vector and/or 5' and/or 3' flanking sequences) using standard cloning techniques. This construct, known as a "homologous recombination construct," can then be introduced into the desired cells either ex vivo or in vivo.
Gene therapy also can be used to decrease IL-lra-R polypeptide expression by modifying the nucleotide sequence of the endogenous promoter.
Such modification is typically accomplished via homologous recombination methods. For example, a DNA molecule containing all or a portion of the promoter of the IL-Ira-R gene selected for inactivation can be engineered to remove and/or replace pieces of the promoter that regulate transcription. For example, the TATA box and/or the binding site of a transcriptional activator of the promoter may be deleted using standard molecular biology techniques; such deletion can inhibit promoter activity thereby repressing the transcription of the corresponding IL-Ira-R gene. The deletion of the TATA box or the transcription WO 01/42304 PCT/US00/32940 activator binding site in the promoter may be accomplished by generating a DNA construct comprising all or the relevant portion of the IL-lra-R polypeptide promoter (from the same or a related species as the IL-lra-R gene to be regulated) in which one or more of the TATA box and/or transcriptional activator binding site nucleotides are mutated via substitution, deletion and/or insertion of one or more nucleotides. As a result, the TATA box and/or activator binding site has decreased activity or is rendered completely inactive. This construct, which also will typically contain at least about 500 bases of DNA that correspond to the native (endogenous) 5' and 3' DNA sequences adjacent to the promoter segment that has been modified, may be introduced into the appropriate cells (either ex vivo or in vivo) either directly or via a viral vector as described herein. Typically, the integration of the construct into the genomic DNA of the cells will be via homologous recombination, where the 5' and 3' DNA sequences in the promoter construct can serve to help integrate the modified promoter region via hybridization to the endogenous chromosomal DNA.
Therapeutic Uses IL-lra-R nucleic acid molecules, polypeptides, and agonists and antagonists thereof can be used to treat, diagnose, ameliorate, or prevent a number of diseases, disorders, or conditions, including those recited herein.
IL-lra-R polypeptide agonists and antagonists include those molecules which regulate IL-lra-R polypeptide activity and either increase or decrease at least one activity of the mature form of the IL-lra-R polypeptide. Agonists or antagonists may be co-factors, such as a protein, peptide, carbohydrate, lipid, or small molecular weight molecule, which interact with IL-lra-R polypeptide and thereby regulate its activity. Potential polypeptide agonists or antagonists include antibodies that react with either soluble or membrane-bound forms of IL-lra-R polypeptides that comprise part or all of the extracellular domains of the said proteins. Molecules that regulate IL-lra-R polypeptide expression typically include nucleic acids encoding IL-lra-R polypeptide that can act as anti-sense regulators of expression.
WO 01/42304 PCT/US00/32940 For example, the IL-lra-R nucleic acid molecules, polypeptides, and agonists and antagonists of the invention can be used to treat, diagnose, ameliorate, or prevent diseases, disorders, or conditions involving immune system dysfunction. Examples of such diseases include, but are not limited to, rheumatoid arthritis, psioriatic arthritis, inflammatory arthritis, osteoarthritis, inflammatory joint disease, autoimmune disease (including autoimmune vasculitis), multiple sclerosis, lupus, diabetes insulin diabetes), inflammatory bowel disease, transplant rejection, graft versus host disease, and inflammatory conditions resulting from strain, sprain, cartilage damage, trauma, orthopedic surgery, infection or other disease processes. Other diseases influenced by the dysfunction of the immune system are encompassed within the scope of the invention.
The IL-lra-R nucleic acid molecules, polypeptides, and agonists and antagonists of the invention can also be used to treat, diagnose, ameliorate, or prevent diseases, disorders, or conditions involving infection. Examples of such diseases include, but are not limited to, leprosy, viral infections (such as hepatitis or HIV), bacterial infection (such as clostridium-associated illnesses, including clostridium-associatcd diarrhea), pulmonary tuberculosis, acute febrile illness, fever, acute phase response of the liver, septicemia, or septic shock. Other diseases involving infection are encompassed within the scope of the invention.
The IL-lra-R nucleic acid molecules, polypeptides, and agonists and antagonists of the invention can also be used to treat, diagnose, ameliorate, or prevent diseases, disorders, or conditions involving weight disorders. Examples of such diseases include, but are not limited to obesity, anorexia, cachexia (including AIDS-induced cachexia), myopathies muscle protein metabolism, such as in sepsis), and hypoglycemia. Other diseases involving weight disorders are encompassed within the scope of the invention.
The IL-lra-R nucleic acid molecules, polypeptides, and agonists and antagonists of the invention can also be used to treat, diagnose, ameliorate, or prevent diseases, disorders, or conditions involving neuronal dysfunction.
Examples of such diseases include, but are not limited to, Alzheimer's disease, 86 WO 01/42304 PCT/US00/32940 Parkinson's disease, neurotoxicity as induced by HIV), ALS, brain injury, stress, depression, nociception and other pain (including cancer-related pain), hyperalgesia, epilepsy, learning impairment and memory disorders, sleep disturbance, and peripheral and central neuropathies. Other neurological disorders are encompassed within the scope of the invention.
The IL-lra-R nucleic acid molecules, polypeptides, and agonists and antagonists of the invention can also be used to treat, diagnose, ameliorate, or prevent diseases, disorders, or conditions involving the lung. Examples of such diseases include, but are not limited to, acute or chronic lung injury (including interstitial lung disease), acute respiratory disease syndrome, pulmonary hypertension, emphysema, cystic fibrosis, pulmonary fibrosis, and asthma. Other diseases of the lung are encompassed within the scope of the invention.
The IL-lra-R nucleic acid molecules, polypeptides, and agonists and antagonists of the invention can also be used to treat, diagnose, ameliorate, or prevent diseases, disorders, or conditions involving the skin. Examples of such diseases include, but are not limited to, psoriasis, eczema, and wound healing.
Other diseases of the skin are encompassed within the scope of the invention.
The IL-lra-R nucleic acid molecules, polypeptides, and agonists and antagonists of the invention can also be used to treat, diagnose, ameliorate, or prevent diseases, disorders, or conditions involving the kidney. Examples of such diseases include, but are not limited to, acute and chronic glomerulonephritis.
Other diseases of the kidney are encompassed within the scope of the invention.
The IL-lra-R nucleic acid molecules, polypeptides, and agonists and antagonists of the invention can also be used to treat, diagnose, ameliorate, or prevent diseases, disorders, or conditions involving the bone. Examples of such diseases include, but are not limited to, osteoporosis, osteopetrosis, osteogenesis imperfecta, Paget's disease, periodontal disease, temporal mandibular joint disease, and hypercalcemia. Other diseases of the bone are encompassed within the scope of the invention.
The IL-lra-R nucleic acid molecules, polypeptides, and agonists and antagonists of the invention can also be used to treat, diagnose, ameliorate, or 87 WO 01/42304 PCT/US00/32940 prevent diseases, disorders, or conditions involving the vascular system.
Examples of such diseases include, but are not limited to, hemorrhage or stroke, hemorrhagic shock, ischemia (including cardiac ischemia and cerebral ischemia, brain injury as a result of trauma, epilepsy, hemorrhage or stroke, each of which may lead to neurodegeneration), atherosclerosis, congestive heart failure, restenosis, reperfusion injury, and angiogenesis. Other diseases of the vascular system are encompassed within the scope of the invention.
The IL-lra-R nucleic acid molecules, polypeptides, and agonists and antagonists of the invention can also be used to treat, diagnose, ameliorate, or prevent diseases, disorders, or conditions involving tumor cells. Examples of such diseases include, but are not limited to, lymphomas, bone sarcoma, chronic and acute myelogenous leukemia (CML and AML) and other leukemias, multiple myeloma, lung cancer, breast cancer, tumor metastasis, and side effects from radiation therapy. Other diseases involving tumor cells are encompassed within the scope of the invention.
The IL-lra-R nucleic acid molecules, polypeptides, and agonists and antagonists of the invention can also be used to treat, diagnose, ameliorate, or prevent diseases, disorders, or conditions involving the reproductive system.
Examples of such diseases include, but are not limited to, infertility, miscarriage, pre-term labor and delivery, and endometriosis. Other diseases involving the reproductive system are encompassed within the scope of the invention.
The IL-lra-R nucleic acid molecules, polypeptides, and agonists and antagonists of the invention can also be used to treat, diagnose, ameliorate, or prevent diseases, disorders, or conditions involving the eye. Examples of such diseases include, but are not limited to, inflammatory eye disease (as may be associated with, for example, corneal transplant), retinal degeneration, blindness, macular degeneration, glaucoma, uveitis, and retinal neuropathy. Other diseases of the eye are encompassed within the scope of the invention.
The IL-lra-R nucleic acid molecules, polypeptides, and agonists and antagonists of the invention can also be used to treat diseases such as acute pancreatitis, chronic fatigue syndrome, fibromyalgia, and Kawasaki's disease 88 WO 01/42304 PCT/US00/32940
(MLNS).
IL-1 inhibitors include any protein capable of specifically preventing activation of cellular receptors to IL-1, which may result from any number of mechanisms. Such mechanisms include down-regulating IL-1 production, binding free IL-1, interfering with IL-1 binding to its receptor, interfering with the formation of the IL-1 receptor complex association of IL-I receptor with IL- I receptor accessory protein), and interfering with the modulation of IL-1 signaling after binding to its receptor. Such interleukin-1 inhibitors include: interleukin-1 receptor antagonists such as IL-lra-R, as described herein, anti-IL-1 receptor monoclonal antibodies European Patent No. 623674), IL-I binding proteins such as soluble IL-I receptors U.S. Patent Nos. 5,492,888, 5,488,032, 5,464,937, 5,319,071, and 5,180,812), anti-IL-1 monoclonal antibodies PCT Pub. Nos. WO 95/01997, WO 94/02627, WO 90/06371; U.S. Patent No. 4,935,343; and European Patent Nos. 364778, 267611 and 220063), IL-1 receptor accessory proteins and antibodies thereto PCT Pub. No. WO 96/23067); inhibitors of interleukin-lp converting enzyme (ICE) or caspase I, which can be used to inhibit IL-l production and secretion, interleukin-1p protease inhibitors, and other compounds and proteins which block in vivo synthesis or extracellular release of IL-1.
Exemplary IL-1 inhibitors are disclosed in US Patent Nos. 5,747,444, 5,359,032, 5,608,035, 5,843,905, 5,359,032, 5,866,576, 5,869,660, 5,869,315, 5,872,095, 5,955,480; PCT Pub. Nos. WO 98/21957, WO 96/09323, WO 91/17184, WO 96/40907, WO 98/32733, WO 98/42325, WO 98/44940, WO 98/47892, WO 98/56377, WO 99/03837, WO 99/06426, WO 99/06042, WO 91/17249, WO 98/32733, WO 98/17661, WO 97/08174, WO 95/34326, WO 99/36426, and WO 99/36415; European Patent Nos. 534978 and 894795; and French Patent Application FR 2762514.
Interleukin-1 receptor antagonist (IL-Ira) is a human protein that acts as a natural inhibitor of interleukin-1. Preferred receptor antagonists (including IL-Ira and variants and derivatives thereof), as well as methods of making and using thereof, are described in U.S. Patent No. 5,075,222; PCT Pub. Nos. WO 89 WO 01/42304 PCT/US00/32940 91/08285, WO 91/17184, WO92/16221, WO 93/21946, WO 94/06457, WO 94/21275, WO 94/21235, WO 94/20517, WO 96/22793, WO 97/28828, and WO 99/36541; Austrian Patent No. AU 9173636; French Patent No. FR 2706772; and German Patent No. DE 4219626. Such proteins include glycosylated as well as non-glycosylated IL-I receptor antagonists.
Specifically, three exemplary forms of IL-Ira and variants thereof are disclosed and described in the 5,075,222 patent. The first of these, called "IL-li," is characterized as a 22-23 kD molecule on SDS-PAGE with an approximate isoelectric point of 4.8, eluting from a MonoQ FPLC column at around 52 mM NaCI in Tris buffer, pH 7.6. The second, IL-rap, is characterized as a 22-23 kD protein, eluting from a MonoQ column at 48 mM NaCI. Both IL-lra and IL- Irap are glycosylated. The third, IL-lraX, is characterized as a 20 kD protein, eluting from a MonoQ column at 48 mM NaCI, and is non-glycosylated. U.S.
Patent No. 5,075,222 also discloses methods for isolating the genes responsible for coding the inhibitors, cloning the gene in suitable vectors and cell types, and expressing the gene to produce the inhibitors.
Those skilled in the art will recognize that many combinations of deletions, insertions, and substitutions (individually or collectively "variant(s)" herein) can be made within the amino acid sequences of IL-lra-R, provided that the resulting molecule is biologically active possesses the ability to affect one or more of the diseases and disorders such as those recited herein.) As contemplated by the present invention, an IL-lra-R polypeptide may be administered as an adjunct to other therapy and also with other pharmaceutical compositions suitable for the indication being treated. An IL-lra-R polypeptide and any of one or more additional therapies or pharmaceutical formulations may be administered separately, sequentially, or simultaneously.
In a specific embodiment, the present invention is directed to the use of an IL-lra-R polypeptide in combination (pre-treatment, post-treatment, or concurrent treatment) with any of one or more TNF inhibitors for the treatment or prevention of the diseases and disorders recited herein.
Such TNF inhibitors include compounds and proteins that block in vivo WO 01/42304 PCT/US00/32940 synthesis or extracellular release of TNF. In a specific embodiment, the present invention is directed to the use of an IL-Ira-R polypeptide in combination (pretreatment, post-treatment, or concurrent treatment) with any of one or more of the following TNF inhibitors: TNF binding proteins (soluble TNF receptor type-1 and soluble TNF receptor type-II ("sTNFRs"), as defined herein), anti-TNF antibodies, granulocyte colony stimulating factor, thalidomide, BN 50730, tenidap, E 5531, tiapafant PCA 4248, nimesulide, panavir, rolipram, RP 73401, peptide T, MDL 201,449A, R,3S)-Cis-l-[9-(2,6-diaminopurinyl)]-3-hydroxy-4cyclopentene hydrochloride, (1R,3R)-trans- -(9-(2,6-diamino)purinc]-3acetoxycyclopentane, (1 R,3R)-trans- 1 -[9-adenyl)-3-azidocyclopentane hydrochloride and (1R,3R)-trans-1 -(6-hydroxy-purin-9-yl)-3-azidocyclo-pentane.
TNF binding proteins are disclosed in the art Patent No. 5,136,021; European Patent Nos. 308378, 422339, 393438, 398327, 412486, 418014, 433900, 464533, 512528, 526905, 568928, 417563; PCT Pub. Nos.
WO90/13575, WO 91/03553, WO 92/01002, W092/13095, WO 92/16221, WO93/07863, WO 93/21946, WO 93/19777, WO 94/06476, PCT App. No.
PCT/US97/12244; English Patent Nos. GB 2218101 and 2246569; and Japanese Patent App. No. JP 127,800/1991).
For example, European Patent Nos. 393438 and 422339 teach the amino acid and nucleic acid sequences of a soluble TNF receptor type I (also known as "sTNFR-I" or "30kDa TNF inhibitor") and a soluble TNF receptor type II (also known as "sTNFR-II" or "40kDa TNF inhibitor"), collectively termed "sTNFRs," as well as modified forms thereof fragments, functional derivatives, and variants). European Patent Nos. 393438 and 422339 also disclose methods for isolating the genes responsible for coding the inhibitors, cloning the gene in suitable vectors and cell types, and expressing the gene to produce the inhibitors.
Additionally, polyvalent forms molecules comprising more than one active moiety) of sTNFR-I and sTNFR-II have also been disclosed. In one embodiment, the polyvalent form may be constructed by chemically coupling at least one TNF inhibitor and another moiety with any clinically acceptable linker, for example polyethylene glycol (PCT Pub. Nos. WO 92/16221 and WO 95/34326), by a 91 WO 01/42304 PCT/US00/32940 peptide linker (Neve et al., 1996, Cytokine, 8:365-70), by chemically coupling to biotin and then binding to avidin (PCT Pub. No. WO 91/03553) and, finally, by combining chimeric antibody molecules Patent No. 5,116,964; PCT Pub.
Nos. WO 89/09622 and WO 91/16437; and European Patent No. 315062).
Anti-TNF antibodies include MAK 195F Fab antibody (Holler et al., 1993, 1st International Symposium on Cytokines in Bone Marrow Transplantation 147), CDP 571 anti-TNF monoclonal antibody (Rankin et al., 1995, Br. J. Rheumatol., 34:334-42), BAY X 1351 murine anti-tumor necrosis factor monoclonal antibody (Kieft et al., 1995, 7th European Congress of Clinical Microbiology and Infectious Diseases CenTNF cA2 anti-TNF monoclonal antibody (Elliott et al., 1994, Lancet, 344:1125-27; Elliott et al., 1994, Lancet, 344:1105-10).
In a specific embodiment, the present invention is directed to the use of an IL-lra-R polypeptide in combination (pretreatment, post-treatment, or concurrent treatment) with secreted or soluble human fas antigen or recombinant versions thereof (PCT Pub. No. WO 96/20206; Mountz et al., 1995, J. Immunol., 155:4829-37; and European Patent No. 510691). PCT Pub. No. WO 96/20206 discloses secreted human fas antigen (native and recombinant, including an Ig fusion protein), methods for isolating the genes responsible for coding the soluble recombinant human fas antigen, methods for cloning the gene in suitable vectors and cell types, and methods for expressing the gene to produce the inhibitors.
European Patent No. 510691 teaches nucleic acids coding for human fas antigen, including soluble fas antigen, vectors expressing for said nucleic acids, and transformants transfected with the vector. When administered parenterally, doses of a secreted or soluble fas antigen fusion protein each are generally from about 1 pg/kg to about 100 pg /kg.
Current treatment of the diseases and disorders recited herein, including acute and chronic inflammation such as rheumatic diseases, commonly includes the use of first line drugs for control of pain and inflammation; these drugs are classified as non-steroidal, anti-inflammatory drugs (NSAIDs). Secondary treatments include corticosteroids, slow acting antirheumatic drugs (SAARDs), or disease modifying (DM) drugs. Information regarding the following compounds 92 WO 01/42304 PCT/US00/32940 can be found in The Merck Manual of Diagnosis and Therapy (16th ed. 1992) and in Pharmaprojects (PJB Publications Ltd).
In a specific embodiment, the present invention is directed to the use of an IL- ra-R polypeptide and any of one or more NSAIDs for the treatment of the diseases and disorders recited herein, including acute and chronic inflammation such as rheumatic diseases, and graft versus host disease. NSAIDs owe their antiinflammatory action, at least in part, to the inhibition of prostaglandin synthesis (Goodman and Gilman, The Pharmacological Basis of Therapeutics (7th ed.
1985)). NSAIDs can be characterized into at least nine groups: salicylic acid derivatives, propionic acid derivatives, acetic acid derivatives, fenamic acid derivatives, carboxylic acid derivatives, butyric acid derivatives, (7) oxicams, pyrazoles, and pyrazolones.
In another specific embodiment, the present invention is directed to the use of an IL-lra-R polypeptide in combination (pretreatment, post-treatment, or concurrent treatment) with any of one or more salicylic acid derivatives, prodrug esters, or pharmaceutically acceptable salts thereof. Such salicylic acid derivatives, prodrug esters, and pharmaceutically acceptable salts thereof comprise: acetaminosalol, aloxiprin, aspirin, benorylate, bromosaligenin, calcium acetylsalicylate, choline magnesium trisalicylate, magnesium salicylate, choline salicylate, diflusinal, etersalate, fendosal, gentisic acid, glycol salicylate, imidazole salicylate, lysine acetylsalicylate, mesalamine, morpholine salicylate, 1naphthyl salicylate, olsalazine, parsalmide, phenyl acetylsalicylate, phenyl salicylate, salacetamide, salicylamide O-acetic acid, salsalate, sodium salicylate and sulfasalazine. Structurally related salicylic acid derivatives having similar analgesic and anti-inflammatory properties are also intended to be encompassed by this group.
In an additional specific embodiment, the present invention is directed to the use of an IL-lra-R polypeptide in combination (pretreatment, post-treatment, or concurrent treatment) with any of one or more propionic acid derivatives, 3 0 prodrug esters, or pharmaceutically acceptable salts thereof. The propionic acid derivatives, prodrug esters, and pharmaceutically acceptable salts thereof 93 WO 01/42304 PCT/US00/32940 comprise: alminoprofen, benoxaprofen, bucloxic acid, carprofen, dexindoprofen, fenoprofen, flunoxaprofen, fluprofen, flurbiprofen, furcloprofen, ibuprofen, ibuprofen aluminum, ibuproxam, indoprofen, isoprofen, ketoprofen, loxoprofen, miroprofen, naproxen, naproxen sodium, oxaprozin, piketoprofen, pimeprofen, pirprofen, pranoprofen, protizinic acid, pyridoxiprofen, suprofen, tiaprofenic acid and tioxaprofen. Structurally related propionic acid derivatives having similar analgesic and anti-inflammatory properties are also intended to be encompassed by this group.
In yet another specific embodiment, the present invention is directed to the use of an IL-lra-R polypeptide in combination (pretreatment, post-treatment, or concurrent treatment) with any of one or more acetic acid derivatives, prodrug esters, or pharmaceutically acceptable salts thereof. The acetic acid derivatives, prodrug esters, and pharmaceutically acceptable salts thereof comprise: acemetacin, alclofenac, amfenac, bufexamac, cinmetacin, clopirac, delmetacin, diclofenac potassium, diclofenac sodium, etodolac, felbinac, fenclofenac, fenclorac, fenclozic acid, fentiazac, furofenac, glucametacin, ibufenac, indomethacin, isofezolac, isoxepac, lonazolac, metiazinic acid, oxametacin, oxpinac, pimetacin, proglumetacin, sulindac, talmetacin, tiaramide, tiopinac, tolmetin, tolmetin sodium, zidomctacin and zomepirac. Structurally related acetic acid derivatives having similar analgesic and anti-inflammatory properties are also intended to be encompassed by this group.
In another specific embodiment, the present invention is directed to the use of an IL-Ira-R polypeptide in combination (pretreatment, post-treatment, or concurrent treatment) with any of one or more fenamic acid derivatives, prodrug esters, or pharmaceutically acceptable salts thereof. The fenamic acid derivatives, prodrug esters, and pharmaceutically acceptable salts thereof comprise: enfenamic acid, etofenamate, flufenamic acid, isonixin, meclofenamic acid, meclofenamate sodium, medofenamic acid, mefenamic acid, niflumic acid, talniflumate, terofenamate, tolfenamic acid and ufenamate. Structurally related fenamic acid derivatives having similar analgesic and anti-inflammatory properties are also intended to be encompassed by this group.
94 WO 01/42304 PCT/US00/32940 In an additional specific embodiment, the present invention is directed to the use of an IL-lra-R polypeptide in combination (pretreatment, post-treatment, or concurrent treatment) with any of one or more carboxylic acid derivatives, prodrug esters, or pharmaceutically acceptable salts thereof. The carboxylic acid derivatives, prodrug esters, and pharmaceutically acceptable salts thereof which can be used comprise: clidanac, diflunisal, flufenisal, inoridine, ketorolac and tinoridine. Structurally related carboxylic acid derivatives having similar analgesic and anti-inflammatory properties are also intended to be encompassed by this group.
In yet another specific embodiment, the present invention is directed to the use of an IL-lra-R polypeptide in combination (pretreatment, post-treatment, or concurrent treatment) with any of one or more butyric acid derivatives, prodrug esters, or pharmaceutically acceptable salts thereof. The butyric acid derivatives, prodrug esters, and pharmaceutically acceptable salts thereof comprise: bumadizon, butibufen, fenbufen and xenbucin. Structurally related butyric acid derivatives having similar analgesic and anti-inflammatory properties are also intended to be encompassed by this group.
In another specific embodiment, the present invention is directed to the use of an IL-lra-R polypeptide in combination (pretreatment, post-treatment, or concurrent treatment) with any of one or more oxicams, prodrug esters, or pharmaceutically acceptable salts thereof. The oxicams, prodrug esters, and pharmaceutically acceptable salts thereof comprise: droxicam, enolicam, isoxicam, piroxicam, sudoxicam, tenoxicam and 4-hydroxyl-l,2-benzothiazine 1,1-dioxide 4-(N-phenyl)-carboxamide. Structurally related oxicams having similar analgesic and anti-inflammatory properties are also intended to be encompassed by this group.
In still another specific embodiment, the present invention is directed to the use of an IL-lra-R polypeptide in combination (pretreatment, post-treatment, or concurrent treatment) with any of one or more pyrazoles, prodrug esters, or pharmaceutically acceptable salts thereof. The pyrazoles, prodrug esters, and pharmaceutically acceptable salts thereof which may be used comprise: WO 01/42304 WO 0142304PCTIUSOO/32940 difenamizole and epirizole. Structurally related pyrazoles having similar analgesic and anti -inflammatory properties are also intended to be encompassed by this group.
In an additional specific embodiment, the present invention is directed to the use of an IL-lra-R polypeptide in combination (pretreatment, post-treatment or, concurrent treatment) with any of one or more pyrazolones, prodrug esters, or pharmaceutically acceptable salts thereof. The pyrazolones, prodrug esters, and pharmaceutically acceptable salts thereof which may be used comprise: apazone, azapropazone, benzpipcrylon, feprazone, mofebutazone, morazone, oxyphenbutazone, phenylbutazone, pipebuzone, propylphenazone, ramifenazone, suxibuzone and thiazolinobutazone. Structurally related pyrazalones having similar analgesic and anti-inflammatory properties are also intended to be encompassed by this group.
In another specific embodiment, the present invention is directed to the use of an IL-I ra-R polypeptide in combination (pretreatment, post-treatment, or concurrent treatment) with any of one or more of the following: NSAIDs: Cacetamidocaproic acid, S-adenosylmethionine, 3-wmino-4-hydroxybutyric acid, amixetrine, anitrazafen, antrafenine, bendazac, bendazac lysinate, benzydamine, beprozin, broperamole, bucolome, bufezolac, ciproquazone, cloximate, dazidamine, deboxamet, detomidine, difenpiramide, difenpyramide, difisalamine, ditazol, emorfazone, fanetizole mesylate, fenflumizole, floctafenine, flumizole, flunixin, fluproquazone, fopirtoline, fosfosal, guaimesal, guaiazolene, isonixirn, lefetam-ine HCI, leflunomide, lofemizole, lotifazole, lysiri clonixinate, meseclazone, nabumnetone, nictindole, nimesulide, orgotein, orpanoxin, oxaceprol, oxapadol, paranyline, perisoxal, perisoxal citrate, pifoxime, piproxen, pirazolac, pirfenidone, proquazone, proxazole, thielavin B, tiflamnizole, timegadine, tolectin, tolpadol, tryptamid and those designated by company code number such as 480156S, AA861, AD1590, AFP8O2, AFP860, A17713, APSO4, AU8001, BPPC, BW54OC, CH[NOIN 127, CNlOO, EB382, EL508, F1044, FK-506, GV3658, ITF182, KCNTE16090, KME4, LA2851, MR714, MR897, MY309, 0N03144, PR823, PVIO2, PVlO8, R830, RS2131, SCRIS2, SH440, SIR133, 96 WO 01/42304 PTUO/24 PCTIUSOO/32940 SQ27239, ST281, SY600I, TA6Q, TAI-901 (4-benzoyl-l-indancarboxylic acid), TVX27O6, U60257, UR2301 and WY41770. Structurally related NSAIDs having similar analgesic and anti-inflammatory properties to the NSAIDs are also intended to be encompassed by this group.
In still another specific embodiment, the present invention is directed to the use of an IL-I ra-R polypeptide in combination (pretreatment, post-treatment or concurrent treatment) with any of one or more corticosteroids, prodrug esters, or pharmaceutically acceptable salts thereof for the treatment of the diseases and disorders recited herein, including acute and chronic inflammation such as rheumatic diseases, graft versus host disease, and multiple sclerosis.
Corticosteroids, prodrug esters, and pharmaceutically acceptable salts thereof include hydrocortisone and compounds which are derived from hydrocortisone, such as 21 -acetoxypregnenolone, alclomerasone, algestone, amcinonide, beclomethasone, betamnethasone, betamethasone valerate, budesonide, chloroprednisone, clobetasol, clobetasol propionate, clobetasone, clobetasone butyrate, clocortolone, cloprednol, corticosterone, cortisone, cortivazol, deflazacon, desonide, desoximerasone, dexamethasone, diflorasone, diflucortolone, difluprednate, enoxolone, fluazacort, flucloronide, flumethasone, flumnethasone pivalate, flucinolone acetonide, flunisolide, fluocinonide, fluorocinolone acetonide, fluo~cortin butyl, fluocortolone, fluocortolone hexanoate, diflucortolone valerate, fluorometholone, fluperolone acetate, fluprednidene acetate, fluprednisolone, flurandenolide, formocortal, halcinonide, halometasone, halopredone acetate, hydrocortamate, hydrocortisone, hydrocortisone acetate, hydrocortisone butyrate, hydrocortisone phosphate, hydrocortisone 21-sodium succinate, hydrocortisone tebutate, mazipredone, medrysone, meprednisone, methylprednisolone, mometasone furoate, paramethasone, prednicarbate, prednisolone, prednisolone 2 1 -diedryamninoacetate, prednisolone sodium phosphate, prednisolone sodium succmnate, prednisolone sodium 21-rnsulfobenzoate, prednisolone sodium 21 -stearoglycolate, prednisolone tebutate, prednisolone 2 1 -trimethylacetate, prednisone, prednival, prednylidene, prednylidene 2 1-diethylaminoacetate, tixocortol, triamcinolone, triamcinolone WO 01/42304 PCT/US0/32940 acetonide, triamcinolone benetonide and triamcinolone hexacetonide. Structurally related corticosteroids having similar analgesic and anti-inflammatory properties are also intended to be encompassed by this group.
In another specific embodiment, the present invention is directed to. the use of an IL-lra-R polypeptide in combination (pretreatment, post-treatment, or concurrent treatment) with any of one or more slow-acting antirheumatic drugs (SAARDs) or disease modifying antirheumatic drugs (DMARDS), prodrug esters, or pharmaceutically acceptable salts thereof for the treatment of the diseases and disorders recited herein, including acute and chronic inflammation such as rheumatic diseases, graft versus host disease, and multiple sclerosis. SAARDs or DMARDS, prodrug esters, and pharmaceutically acceptable salts thereof comprise: allocupreide sodium, auranofin, aurothioglucose, aurothioglycanide, azathioprine, brequinar sodium, bucillamine, calcium 3-aurothio-2-propanol-1sulfonate, chlorambucil, chloroquine, clobuzarit, cuproxoline, cyclophosphamide, cyclosporin, dapsone, 15-deoxyspergualin, diacerein, glucosamine, gold salts cycloquine gold salt, gold sodium thiomalate, gold sodium thiosulfate), hydroxychloroquine, hydroxychloroquine sulfate, hydroxyurea, kebuzone, levamisole, lobenzarit, melittin, 6-mercaptopurine, methotrexate, mizoribine, mycophenolate mofetil, myoral, nitrogen mustard, D-penicillamine, pyridinol imidazoles such as SKNF86002 and SB203580, rapamycin, thiols, thymopoietin and vincristine. Structurally related SAARDs or DMARDs having similar analgesic and anti-inflammatory properties are also intended to be encompassed by this group.
In another specific embodiment, the present invention is directed to the use of an IL-lra-R polypeptide in combination (pretreatment, post-treatment, or concurrent treatment) with any of one or more COX2 inhibitors, prodrug esters, or pharmaceutically acceptable salts thereof for the treatment of the diseases and disorders recited herein, including acute and chronic inflammation. Examples of COX2 inhibitors, prodrug esters, or pharmaceutically acceptable salts thereof include, for example, celecoxib. Structurally related COX2 inhibitors having similar analgesic and anti-inflammatory properties are also intended to be 98 WO 01/42304 WO 0142304PCTfUSOOI3 2940 encompassed by this group.
In still another specific embodiment, the present invention is directed to the use of an IL-i ra-R polypeptide in combination (pretreatment, post-treatment, or concurrent treatment) with any of one or more antimicrobials, prodrug esters, or pharmaceutically acceptable salts thereof for the treatment of the diseases and disorders recited herein, including acute and chronic inflammation.
Antimicrobials include, for example, the broad classes of penicillins, cephalosponins and other beta-lactams, aminoglycosides, azoles, quinolones, macrolides, ri famycins, tetracyclines, sulfoniamides, lincosamides and polymyxins. The penicillins include, but are not limited to, penicillin G, penicillin V, methicillin, nafillin, oxacillin, cloxacillin, dicloxacillin, floxacillin, ampicillin, ampicillin/sulbactam, amoxi cillin, amoxicillin/clavulanate, hetacillin, cyclacillin, bacampicillin, carbenicillin, carbenicillin indanyl, ticarcillin, ticarcillinlclavulanate, azlocillin, meziocillin, peperacillin, and mecillinam. The cephalosporins and other beta-lactams include, but are not limited to, cephalothin, cephapirin, cephalexin, cephradine, cefazolin, cefadroxil, cefaclor, cefamandole, cefotetan, cefoxitin, ceruroxime, cefonicid, ceforadine, cefixime, cefotaxime, moxalactam, ceftizoxime, cetriaxone, ccphoperazone, ceftazidime, imipenem and aztreonam. The aminoglycosides include, but are not limited to, streptomycin, gentamnicin, tobramycin, amikacin, netilmicin, kanamycin and neomycin. The azoles; include, but are not limited to, fluconazole. The quinolones include, but are not limited to, nalidixic acid, norfioxacin, enoxacin, ciprofloxacin, ofloxacin, sparfioxacin and temnafloxacin. The macrolides include, but are not limited to, erythomycin, spiramycin and azithromycin. The rifamycins include, but are not limited to, rifampin. The tetracyclines include, but are not limited to, spicycline, chlortetracycline, clomocycline, demeclocycline, deoxycycline, guamecycline, lymccycline, meclocycline, methacycline, minocycline, oxytetracycline, penimepicycline, pipacycline, rolitetracycline, sancycline, senociclin and tetracycline. The sulfonamides include, but are not limited to, sulfanilamide, sulfamethoxazole, sulfacetamide, sulfadiazine, sulfisoxazole and co-trimoxazole (trimethopri-l/sulfamnethoxazole). The lincosamides include, but are not limited 99 WO 01/42304 PCT/US00/32940 to, clindamycin and lincomycin. The polymyxins (polypeptides) include, but are not limited to, polymyxin B and colistin.
Agonists or antagonists of IL-IRA-R polypeptide function may be used (simultaneously or sequentially) in combination with one or more cytokines, growth factors, antibiotics, anti-inflammatories, and/or chemotherapeutic agents as is appropriate for the condition being treated.
Other diseases caused by or mediated by undesirable levels of one or more of IL-1, IL-lra, or IL-lra-R polypeptide are encompassed within the scope of the invention. Undesirable levels include excessive levels of IL-1, IL-Ira, or IL-lra-R polypeptide and sub-normal levels of IL-1, IL-Ira, or IL-lra-R polypeptide.
Uses of IL-lra-R Nucleic Acids and Polypeptides Nucleic acid molecules of the invention (including those that do not themselves encode biologically active polypeptides) may be used to map the locations of the IL-lra-R gene and related genes on chromosomes. Mapping may be done by techniques known in the art, such as PCR amplification and in situ hybridization.
IL-Ira-R nucleic acid molecules (including those that do not themselves encode biologically active polypeptides), may be useful as hybridization probes in diagnostic assays to test, either qualitatively or quantitatively, for the presence of an IL-lra-R nucleic acid molecule in mammalian tissue or bodily fluid samples.
Other methods may also be employed where it is desirable to inhibit the activity of one or more IL-lra-R polypeptides. Such inhibition may be effected by nucleic acid molecules that are complementary to and hybridize to expression control sequences (triple helix formation) or to IL-lra-R mRNA. For example, antisense DNA or RNA molecules, which have a sequence that is complementary to at least a portion of an IL-lra-R gene can be introduced into the cell. Antisense probes may be designed by available techniques using the sequence of the IL-lra-R gene disclosed herein. Typically, each such antisense molecule will be complementary to the start site end) of each selected IL-Ira-R gene. When the antisense molecule then hybridizes to the corresponding IL-lra-R mRNA, 100 WO 01/42304 PCT/US00/32940 translation of this mRNA is prevented or reduced. Anti-sense inhibitors provide information relating to the decrease or absence of an IL-lra-R polypeptide in a cell or organism.
Alternatively, gene therapy may be employed to create a dominantnegative inhibitor of one or more IL-lra-R polypeptides. In this situation, the DNA encoding a mutant polypeptide of each selected IL-Ira-R polypeptide can be prepared and introduced into the cells of a patient using either viral or non-viral methods as described herein. Each such mutant is typically designed to compete with endogenous polypeptide in its biological role.
In addition, an IL-lra-R polypeptide, whether biologically active or not, may be used as an immunogen, that is, the polypeptide contains at least one epitope to which antibodies may be raised. Selective binding agents that bind to an IL-lra-R polypeptide (as described herein) may be used for in vivo and in vitro diagnostic purposes, including, but not limited to, use in labeled form to detect the presence of IL-lra-R polypeptide in a body fluid or cell sample. The antibodies may also be used to prevent, treat, or diagnose a number of diseases and disorders, including those recited herein. The antibodies may bind to an IL-lra-R polypeptide so as to diminish or block at least one activity characteristic of an IL- Ira-R polypeptide, or may bind to a polypeptide to increase at least one activity characteristic of an IL-lra-R polypeptide (including by increasing the pharmacokinetics of the IL-lra-R polypeptide).
The IL-lra-R polypeptides of the present invention can be used to clone IL-lra-R polypeptide receptors, using an expression cloning strategy.
Radiolabeled ('2lodine) IL-lra-R polypeptide or affinity/activity-tagged IL-lra-R polypeptide (such as an Fc fusion or an alkaline phosphatase fusion) can be used in binding assays to identify a cell type or cell line or tissue that expresses IL-lra- R polypeptide receptors. RNA isolated from such cells or tissues can be converted to cDNA, cloned into a mammalian expression vector, and transfected into mammalian cells (such as COS or 293 cells) to create an expression library.
A radiolabeled or tagged IL-lra-R polypeptide can then be used as an affinity ligand to identify and isolate from this library the subset of cells that express the 101 WO 01/42304 PCT/US00/32940 IL-lra-R polypeptide receptors on their surface. DNA can then be isolated from these cells and transfected into mammalian cells to create a secondary expression library in which the fraction of cells expressing IL-lra-R polypeptide receptors is many-fold higher than in the original library. This enrichment process can be repeated iteratively until a single recombinant clone containing an IL-lra-R polypeptide receptor is isolated. Isolation of the IL-lra-R polypeptide receptors is useful for identifying or developing novel agonists and antagonists of the IL-ira- R polypeptide signaling pathway. Such agonists and antagonists include soluble IL-lra-R polypeptide receptors, anti-IL-lra-R polypeptide receptor antibodies, small molecules, or antisense oligonucleotides, and they may be used for treating, preventing, or diagnosing one or more of the diseases or disorders described herein.
A deposit of cDNA encoding human IL-lra-R polypeptide, having Accession No. PTA-1423, was made with the American Type Culture Collection (ATCC), 10801 University Boulevard, Manassas, VA 20110-2209 on February 29, 2000.
The following examples are intended for illustration purposes only, and should not be construed as limiting the scope of the invention in any way.
Example 1: Cloning of the Human IL-Ira-R Polypeptide Gene Generally, materials and methods as described in Sambrook et al. supra were used to clone and analyze the gene encoding human IL-lra-R polypeptide.
To isolate cDNA sequences encoding human IL-lra-R polypeptide, a search of a proprietary database (Amgen, Thousand Oaks, CA) was performed using clone shosl-00003-dl as a query sequence. A 423 bp sequence identified in this manner was found to share 41% identity to the query sequence at the amino acid level. The region of highest homology within this sequence was used to design gene specific oligonucleotides for the identification of cDNA sources and the generation of cDNA clones, using various PCR strategies.
A number of cDNA libraries were analyzed in amplification reactions containing 10ng of cDNA library template DNA, 10 pmol each of the amplimers 102 WO 01/42304 PCT/US00/32940 2349-98 SEQ ID NO: 20) and 2349-99 SEQ ID NO: 21), and Ready-To-Go PCR beads (Amersham-Pharmacia, Piscataway, NJ) (Pharmacia, Piscataway, NJ), in a total reaction volume..of 25 Il.
Reactions were performed at 95C for 5 minutes for one cycle; 95 0 C for seconds, 63 0 C for 15 seconds, and 72C for 1 minute for 30 cycles; and 72 0 C for minutes for one cycle. A PCR product of the expected size (153 bp) was identified in a number of cDNA libraries, including human fetal scalp (oligo-dT primed) and human placenta (oligo-dT primed and random primed).
The fetal scalp and placenta cDNA libraries were prepared as follows.
Total RNA was extracted from human fetal scalp and from human placenta using standard RNA extraction procedures and poly-A RNA was selected from this total RNA using standard procedures. Random primed or oligo-dT primed cDNA was synthesized from this poly-A RNA using the Superscript Plasmid System for cDNA Synthesis and Plasmid Cloning kit (Gibco-BRL), according to the manufacturer's suggested protocols, or other suitable procedure. The resulting cDNA was digested with appropriate restriction enzymes and was then ligated into pSPORT-1, or other suitable cloning vector. Ligation products were transformed into E. coli using standard techniques, and bacterial transformants were selected on culture plates containing either ampicillin, tetracycline, kanamycin, or chloramphenicol. The cDNA library consisted of all, or a subset, of these transformants.
Both 5'RACE and 3'RACE reactions were performed in order to generate the full-length cDNA sequence for IL-lra-R polypeptide. To isolate cDNA sequences corresponding to the 5' end of the cDNA sequence for IL-lra-R polypeptide, 5'RACE was performed using 10 ng of an oligo-dT-primed human fetal scalp cDNA library in pSPORTI and the primers 1572-36 SEQ ID NO: 22) and 2349-99.
Reactions were performed at 94 0 C for 1 minute for one cycle; 94 0 C for 5 seconds and 72 0 C for 5 minutes for 5 cycles; 94 0 C for 5 seconds, 70 0 C for 10 seconds, and 72 0 C for 3 minutes for 5 cycles; 94 0 C for 5 seconds, 68 0 C for 10 seconds, and 103 WO 01/42304 PCT/US00/32940 72 0 C for 3 minutes for 25 cycles; and 72 0 C for 7 minutes for one cycle. Nested PCR was performed using a portion of the 5'RACE amplification product and the primers 2328-91 SEQ ID NO: 23) and 2351-47 SEQ ID NO: 24). Nested PCR was performed at 94 0 C for 1 minute for one cycle; 94 0 C for 5 seconds and 72 0 C for 5 minutes for 5 cycles; 94 0 C for 5 seconds, 0 C for 10 seconds, and 72 0 C for 3 minutes for 5 cycles; 94 0 C for 5 seconds, 68°C for 10 seconds, and 72 0 C for 3 minutes for 25 cycles; and 72 0 C for 7 minutes for one cycle.
To isolate cDNA sequences corresponding to the 3' end of the cDNA sequence for IL-lra-R polypeptide, 3'RACE was performed using 10 ng of an oligo-dT-primed human fetal scalp cDNA library in pSPORTI and the primers 2351-48 SEQ ID NO: and 2329-93 SEQ ID NO: 26). Reactions were performed at 94 0 C for 1 minute for one cycle; 94C for 5 seconds and 72 0 C for 5 minutes for 5 cycles; 94 0 C for 5 seconds, 70°C for seconds, and 72 0 C for 3 minutes for 5 cycles; 94 0 C for 5 seconds, 68 0 C for seconds, and 72 0 C for 3 minutes for 25 cycles; and 72 0 C for 7 minutes for one cycle. Nested PCR was performed using a portion of the 3'RACE amplification product and the primers 2363-04 SEQ ID NO: 27) and 2329-94 SEQ ID NO: 28). Nested PCR was performed at 95 0 C for 1 minute for one cycle; 95°C for 30 seconds and 68 0 C for 1 minute for 30 cycles; and 68 0 C for 7 minutes for one cycle. The full-length cDNA sequence for IL-Ira- R polypeptide was assembled from the resulting collection of 5'RACE and 3'RACE clones.
The 5' portion of the cDNA sequence for IL-lra-R polypeptide was confirmed by independently isolating cDNA sequences from a human placenta MarathonTM cDNA library (Clontech). To isolate cDNA sequences corresponding to the 5' end of the cDNA sequence for IL-lra-R polypeptide, 5'RACE was performed using 1 ng of human placenta cDNA and the primers AP-1 104 WO 01/42304 PCT/US00/32940 SEQ ID NO: 29; Clontech) and 2353-87 SEQ ID NO: 30). Reactions were performed at 94C for 2 minutes for one cycle; 94C for 5 seconds and 72 0 C for 1.5 minutes for 5 cycles; 94 0 C for seconds and 70°C for 1.5 minutes for 5 cycles; 94 0 C for 5 seconds and 68 0 C for minutes for 25 cycles; and 86 0 C for 7 minutes for one cycle. Nested PCR was performed using a portion of the 5'RACE amplification product and the primers AP-1 and 2349-52
SEQ
ID NO: 31). Nested PCR was performed at 94 0 C for 2 minutes for one cycle; 94 0 C for 10 seconds and 68 0 C for 1.5 minutes for 30 cycles; and 68 0 C for 7 minutes for one cycle.
Sequence analysis of the predicted cDNA sequence for human IL-lra-R polypeptide indicated that the gene comprises a 456 bp open reading frame encoding a protein of 152 amino acids (Figures IA-IB). A sequence variant of the human IL-lra-R polypeptide was also identified. Sequence analysis of this variant indicated that the gene for this variant also comprises a 456 bp open reading frame encoding a protein of 152 amino acids (Figures 2A-2B). The nucleotide sequence of this variant differs from that of the human IL-lra-R gene at two nucleotide positions.
Figures 4A-4B illustrate the amino acid sequence alignment of human ILla (IL-l_alpha; SEQ ID NO: human IL-1p (IL-l_beta; SEQ ID NO: 8), human IL-I receptor antagonist (IL-IRA; SEQ ID NO: human IL-16 (ILl_delta; SEQ ID NO: 10), human IL-lra-R polypeptide (IL-lra-R; SEQ ID NO: human Tango-77 (Tango-77; SEQ ID NO: 11), human Zilla4 (Zilla4; SEQ ID NO: 12), human IL-1l (IL-l_zeta; SEQ ID NO: 13), human IL-I receptor antagonist p (IL-IRA_beta; SEQ ID NO: 14), human SPOIL II (Spoil_II; SEQ ID NO: 15), human IL-le (IL-l_epsilon; SEQ ID NO: 16), and human IL-ll (ILl_eta; SEQ ID NO: 17). Figure 5 schematically illustrates the phylogenetic relationship of the IL-Ira gene family.
Example 2: Cloning of the Human IL-lra-R Polypeptide Gene Splice Variant 105 WO 01/42304 PCT/US00/32940 Generally, materials and methods as described in Sambrook et al. supra were used to clone and analyze the gene encoding a human IL-lra-R polypeptide splice variant.
Full-length cDNA sequences encoding a splice variant of human IL-lra-R polypeptide, were isolated from a human placenta cDNA library, which was prepared as follows. Total RNA was extracted from human placenta using standard RNA extraction procedures and poly-A RNA was selected from this total RNA using standard procedures. Oligo-dT primed cDNA was synthesized from this poly-A' RNA using the Superscript Plasmid System for cDNA Synthesis and Plasmid Cloning kit (Gibco-BRL), according to the manufacturer's suggested protocols. The resulting cDNA was digested with Not I and then fractionated by electrophoresis on a 0.8% agarose gel. Fragments of 0.8 to 1.6 kb were isolated, purified, and then ligated into pSPORT-1. Ligation products were transformed into E. coli using standard techniques, and bacterial transformants were selected on culture plates containing ampicillin. The resulting transformants were pooled to generate the cDNA library and plasmid DNA was prepared from 12 pools (each containing approximately 80,000 colonies) of this cDNA library.
Each of the cDNA library pools was analyzed in amplification reactions containing lOng of template DNA, the amplimers 2349-98 and 2349-99 at a concentration of 0.4 M each, and Ready-To-Go PCR beads, in a total reaction volume of 25 pl. Reactions were performed at 94 0 C for 5 minutes for one cycle; 94 0 C for 30 seconds, 65 0 C for 30 seconds, and 72°C for 1 minute for 30 cycles; and 72 0 C for 10 minutes for one cycle. The cDNA library pools were also analyzed in amplification reactions containing 10ng of template DNA, the amplimers 2349-51 SEQ ID NO: 32) and 2349-52 at a concentration of 0.4 pM each, and Ready- To-Go PCR beads, in a total reaction volume of 25 pl. Reactions were performed at 94 0 C for 5 minutes for one cycle; 94°C for 30 seconds, 65 0 C for 30 seconds, and 72 0 C for 1 minute for 30 cycles; and 72 0 C for 10 minutes for one cycle. The PCR products obtained in these reactions were then analyzed on agarose gels.
Plasmid DNA from three pools yielded PCR products having the expected 106 WO 01/42304 PCT/US00/32940 size, and 3 x 10 s clones from each of these positive pools were screened by PCR using the amplimers 2349-98 and 2349-99. One of the PCR products (zhvt- 000329) generated by PCR was labeled with 3 P-dCTP and used as a probe to rescreen the positive cDNA library pools. Bacteria colonies were plated (at approximately 50,000 per 150 mm plate) and then lifted onto nitrocellulose filters.
Filters were prehybridized in ExpressHyb hybridization solution (Clontech) for minutes at 68 0 C, and then hybridized in the same solution with the addition of labeled probe at 68 0 C overnight. Following hybridization, the filters were washed twice for 10 minutes at room temperature in 2X SSC and 0.05% SDS, and then twice for 30 minutes at 68 OC in 0.IX SSC and 0.1% SDS. Filters were then exposed to autoradiography with intensifying screens at -80 0 C for 2 hours. A positive clone (RDS#199918503) containing an insert of approximately 1.2 kb was identified from one of the cDNA library pools. A second positive clone (RDS#199918501) containing insert size about 0.8 kb was identified from a separate cDNA library pool. Plasmid DNA was prepared from each of these clones and the sequence was analyzed.
Sequence analysis of the predicted cDNA sequence for one of these clones indicated that the cDNA encoded a variant of human IL-lra-R polypeptide (see Example The gene encoding this variant comprises a 513 bp open reading frame encoding a protein of 171 amino acids (Figure 3).
Sequence analysis of the human genomic sequence containing the IL-lra- R gene (Genbank Accession No. AC016724, contig. fragment 76649-96342) indicated that the first exon of the IL-lra-R gene (see Example 1) lies 4.1 kb downstream of the last exon of IL-I Omega (Genbank Accession No. Z300050).
The close proximity of these two genes in the genome suggests that the present polypeptide might arise from splicing (or a fusion) of the first two exons of IL-I Omega onto the second exon of the IL-lra-R gene, or a variant thereof (see Example This variant of the IL-lra-R polypeptide may therefore be an IL-Ira- R splice variant, fusion protein, or the like. Juxtaposition of the second exon in IL-1 Omega and the second exon of the IL-lra-R gene (or a variant thereof) results in a sequence that encodes a protein with an N-terminus that appears to 107 WO 01/42304 PCT/US00/32940 function as a signal peptide. Figure 6 schematically illustrates the relationship between human IL-lra-R polypeptide (Mature CS329), the sequence variant of human IL-lra-R polypeptide (Mature CS329 Variant protein), and the splice variant of human IL-I ra-R polypeptide (Omega 329 protein).
Example 3: Cloning of the Murine IL-lra-R Polvpeptide Gene Generally, materials and methods as described in Sambrook et al. supra were used to clone and analyze the gene encoding murine IL-lra-R polypeptide.
To isolate cDNA sequences encoding murine IL-lra-R polypeptide, PCR was performed using a day 7 mouse embryo cDNA library template and the amplimers 2557-95 SEQ ID NO: 33) and 2557-96 SEQ ID NO: 34) and standard techniques.
Sequence analysis of the predicted cDNA sequence for murine IL-lra-R polypeptide indicated that the gene comprises a 456 bp open reading frame encoding a protein of 152 amino acids (Figure Figure 8 illustrates the amino acid sequence alignment of human IL-lra-R polypeptide (hulL-lra-R; SEQ ID NO: 2) and murine IL-lra-R polypeptide (mulL-lra-R; SEQ ID NO: 36). Figures 9A-91 illustrate the genomic nucleotide sequence for the murine IL-lra-R gene (SEQ ID NO: 37). The locations of the coding portions of exons 1-4 are indicated (underline).
Example 4: IL-lra-R mRNA Expression Expression of IL-lra-R mRNA was examined by PCR in amplification reactions containing 10ng of cDNA library template DNA, 10 pmol of the amplimers 2349-98 and 2349-99, and Ready-To-Go PCR beads, in a total reaction volume of 25 pl. Reactions were performed at 95 0 C for 5 minutes for one cycle; 0 C for 15 seconds, 63 0 C for 15 seconds, and 72°C for 1 minute for 30 cycles; and 72 0 C for 10 minutes for one cycle. A PCR product of the expected size (153 bp) was identified in a number of cDNA libraries, including human fetal scalp, human fetal eye, human gall bladder, and human placenta. IL-lra-R mRNA 108 WO 01/42304 PCT/US00/32940 expression was also detected in fetal eye and fetal spleen by RT-PCR using the amplimers 2349-51 and 2349-52 and the Titan system (Boehringer). These reactions were performed at 55 0 C for 30 minutes for one cycle; 94 0 C for seconds, 64°C for 15 seconds, and 68 0 C for 50 seconds (with an increase of 2 seconds per cycle) for 30 cycles; and 68 0 C for 7 minutes for one cycle.
Expression of IL-lra-R mRNA was examined by Northern blot analysis using human and mouse RNA blots (Clontech). Blots were first prehybridized for minutes at 65 0 C in Pre-Hyb Solution (Amersham) and then probed overnight at 65°C in the same solution containing 25 ng of a 3 P-labeled probe corresponding to nucleotides 1-474 of the cDNA sequence encoding IL-lra-R polypeptide. The probe was labeled using a Redi Prime II kit (Pharmacia). Following hybridization, blots were washed twice for 1 hour in 6X SSC and 0.1% SDS, twice for 1 hour in 2X SSC and 0.1% SDS, twice for 30 minutes in 0.2X SSC and 0.1% SDS, and then were rinsed in 2X SSC. Following overnight exposure of the Northern blots in a phosphoimager, a transcript of approximately 1.6 kb was detected in mouse skeletal muscle (Figure 10A) and a transcript of approximately 2.9 kb was detected in human pancreas and peripheral blood leukocytes (Figure and 10C). In an analysis of dot blots (Clontech) using the same probe, IL- Ira-R mRNA was detected in murine skeletal muscle, submaxillar gland, and epididymis.
The expression of IL-Ira-R mRNA is localized by in situ hybridization. A panel of normal embryonic and adult mouse tissues is fixed in 4% paraformaldehyde, embedded in paraffin, and sectioned at 5 Pm. Sectioned tissues are permeabilized in 0.2 M HC1, digested with Proteinase K, and acetylated with triethanolamine and acetic anhydride. Sections are prehybridized for 1 hour at 60 0 C in hybridization solution (300 mM NaCI, 20 mM Tris-HCI, pH 5 mM EDTA, IX Denhardt's solution, 0.2% SDS, 10 mM DTT, 0.25 mg/ml tRNA, 25 pg/ml polyA, 25 pg/ml polyC and 50% formamide) and then hybridized overnight at 60 0 C in the same solution containing 10% dextran and 2 x 10 4 cpm/Pl 109 WO 01/42304 PCT/US0O/32940 of a "P-labeled antisense riboprobe complementary to the human IL-lra-R gene.
The riboprobe is obtained by in vitro transcription of a clone containing human IL-lra-R cDNA sequences using standard techniques.
Following hybridization, sections are rinsed in hybridization solution, treated with RNaseA to digest unhybridized probe, and then washed in 0.1X SSC at 55°C for 30 minutes. Sections are then immersed in NTB-2 emulsion (Kodak, Rochester, NY), exposed for 3 weeks at 4 developed, and counterstained with hematoxylin and eosin. Tissue morphology and hybridization signal are simultaneously analyzed by darkfield and standard illumination for brain (one sagittal and two coronal sections), gastrointestinal tract (esophagus, stomach, duodenum, jejunum, ileum, proximal colon, and distal colon), pituitary, liver, lung, heart, spleen, thymus, lymph nodes, kidney, adrenal, bladder, pancreas, salivary gland, male and female reproductive organs (ovary, oviduct, and uterus in the female; and testis, epididymus, prostate, seminal vesicle, and vas deferens in the male), BAT and WAT (subcutaneous, peri-renal), bone (femur), skin, breast, and skeletal muscle.
Example 5: Production of IL-lra-R Polvpeptides A. Expression of IL-lra-R Polypeptides in Bacteria PCR is used to amplify template DNA sequences encoding an IL-lra-R polypeptide using primers corresponding to the 5' and 3' ends of the sequence.
The amplified DNA products may be modified to contain restriction enzyme sites to allow for insertion into expression vectors. PCR products are gel purified and inserted into expression vectors using standard recombinant DNA methodology.
An exemplary vector, such as pAMG21 (ATCC no. 98113) containing the lux promoter and a gene encoding kanamycin resistance is digested with Bam HI and Nde I for directional cloning of inserted DNA. The ligated mixture is transformed into an E. coli host strain by electroporation and transformants are selected for kanamycin resistance. Plasmid DNA from selected colonies is isolated and subjected to DNA sequencing to confirm the presence of the insert.
Transformed host cells are incubated in 2xYT medium containing 110 WO 01/42304 PCT/US00/32940 pg/mL kanamycin at 30 0 C prior to induction. Gene expression is induced by the addition of N-(3-oxohexanoyl)-dl-homoserine lactone to a final concentration of ng/mL followed by incubation at either 30 0 C or 37°C for six hours. The expression of IL-lra-R polypeptide is evaluated by centrifugation of the- culture, resuspension and lysis of the bacterial pellets, and analysis of host cell proteins by SDS-polyacrylamide gel electrophoresis.
Inclusion bodies containing IL-ra-R polypeptide are purified as follows.
Bacterial cells are pelleted by centrifugation and resuspended in water. The cell suspension is lysed by sonication and pelleted by centrifugation at 195,000 xg for 5 to 10 minutes. The supernatant is discarded, and the pellet is washed and transferred to a homogenizer. The pellet is homogenized in 5 mL of a Percoll solution (75% liquid Percoll and 0.15 M NaCI) until uniformly suspended and then diluted and centrifuged at 21,600 xg for 30 minutes. Gradient fractions containing the inclusion bodies are recovered and pooled. The isolated inclusion bodies are analyzed by SDS-PAGE.
A single band on an SDS polyacrylamide gel corresponding to E. coliproduced IL-lra-R polypeptide is excised from the gel, and the N-terminal amino acid sequence is determined essentially as described by Matsudaira et al., 1987, J.
Biol. Chem. 262:10-35.
B. Expression of IL-lra-R Polypeptide in Mammalian Cells PCR is used to amplify template DNA sequences encoding an IL-lra-R polypeptide using primers corresponding to the 5' and 3' ends of the sequence.
The amplified DNA products may be modified to contain restriction enzyme sites to allow for insertion into expression vectors. PCR products are gel purified and inserted into expression vectors using standard recombinant DNA methodology.
An exemplary expression vector, pCEP4 (Invitrogen, Carlsbad, CA), that contains an Epstein-Barr virus origin of replication, may be used for the expression of IL- Ira-R polypeptides in 293-EBNA-1 cells. Amplified and gel purified PCR products are ligated into pCEP4 vector and introduced into 293-EBNA cells by lipofection. The transfected cells are selected in 100 ug/mL hygromycin and the 111 WO 01/42304 PCT/USOO/32940 resulting drug-resistant cultures are grown to confluence. The cells are then cultured in serum-free media for 72 hours. The conditioned media is removed and IL-Ira-R polypeptide expression is analyzed by SDS-PAGE.
IL-lra-R polypeptide expression may be detected by silver .staining.
Alternatively, IL-lra-R polypeptide is produced as a fusion protein with an epitope tag, such as an IgG constant domain or a FLAG epitope, which may be detected by Western blot analysis using antibodies to the peptide tag.
IL-lra-R polypeptides may be excised from an SDS-polyacrylamide gel, or IL-lra-R fusion proteins are purified by affinity chromatography to the epitope tag, and subjected to N-terminal amino acid sequence analysis as described herein.
C. Expression and Purification of IL-lra-R Polypeptide in Mammalian Cells IL-lra-R polypeptide expression constructs are introduced into 293 EBNA or CHO cells using either a lipofection or calcium phosphate protocol.
To conduct functional studies on the IL-lra-R polypeptides that are produced, large quantities of conditioned media are generated from a pool of hygromycin selected 293 EBNA clones. The cells are cultured in 500 cm Nunc Triple Flasks to 80% confluence before switching to serum free media a week prior to harvesting the media. Conditioned media is harvested and frozen at -20 0 C until purification.
Conditioned media is purified by affinity chromatography as described below. The media is thawed and then passed through a 0.2 pm filter. A Protein G column is equilibrated with PBS at pH 7.0, and then loaded with the filtered media. The column is washed with PBS until the absorbance at An reaches a baseline. IL-lra-R polypeptide is eluted from the column with 0.1 M Glycine- HCI at pH 2.7 and immediately neutralized with 1 M Tris-HCI at pH Fractions containing IL-lra-R polypeptide are pooled, dialyzed in PBS, and stored at -70 0
C.
For Factor Xa cleavage of the human IL-lra-R polypeptide-Fc fusion polypeptide, affinity chromatography-purified protein is dialyzed in 50 mM Tris- HCI, 100 mM NaCI, 2 mM CaCI 2 at pH 8.0. The restriction protease Factor Xa is 112 WO 01/42304 PCT/US00/32940 added to the dialyzed protein at 1/100 and the sample digested overnight at room temperature.
Example 6: Production of Anti-IL- ra-R Polypeptide Antibodies Antibodies to IL-lra-R polypeptides may be obtained by immunization with purified protein or with IL-lra-R peptides produced by biological or chemical synthesis. Suitable procedures for generating antibodies include those described in Hudson and Bay, Practical Immunology (2nd ed., Blackwell Scientific Publications).
In one procedure for the production of antibodies, animals (typically mice or rabbits) are injected with an IL-lra-R antigen (such as an IL-lra-R polypeptide), and those with sufficient serum titer levels as determined by ELISA are selected for hybridoma production. Spleens of immunized animals are collected and prepared as single cell suspensions from which splenocytes are recovered. The splenocytes are fused to mouse myeloma cells (such as Agl4 cells), are first incubated in DMEM with 200 U/mL penicillin, 200 1 Ig/mL streptomycin sulfate, and 4 mM glutamine, and are then incubated in HAT selection medium (hypoxanthine, aminopterin, and thymidine). After selection, the tissue culture supernatants are taken from each fusion well and tested for anti- IL-lra-R antibody production by ELISA.
Alternative procedures for obtaining anti-IL-lra-R antibodies may also be employed, such as the immunization of transgenic mice harboring human Ig loci for production of human antibodies, and the screening of synthetic antibody libraries, such as those generated by mutagenesis of an antibody variable domain.
Antibodies to IL-lra-R polypeptides were obtained by immunizing New Zealand White rabbits with full-length IL-lra-R polypeptide produced in and isolated from E. coli. Crude polyclonal immune serum was collected and used in immunoprecipitation analysis using recombinant IL-Ira-R polypeptide (Figures 1 A- 1B, lanes 1, 4, and recombinant IL-lra-R polypeptide variant (Figures 11A-11B, lanes 2, 5, and and recombinant IL-lra (Figures 11A-11B; lanes 3, 113 WO 01/42304 PCT/US00/32940 6, and Gels were prepared by loading either 10 ng (Figure 11A, lanes 4-6) or 0.6 pg (Figure 1 IB, lanes 7-9) of recombinant polypeptide directly onto an 18% Tris-glycine gel or by immunoprecipitating 1 pg of recombinant polypeptide with 0.2 pl of crude antiserum (Figure 11A, lanes 1-3) prior to loading the sample on the gel. Following SDS-PAGE separation, gels were blotted onto PVDF membranes and developed using a 1:1000 dilution of crude anti-IL-lra-R serum followed by HRP-conjugated Protein A and ECL detection. The gel shown in Figure 11B was stained with Gelcode Blue (Pierce) following blotting.
Antibodies to IL-lra-R polypeptide were shown to detect both membraneimmobilized IL-lra-R polypeptide and IL-lra-R polypeptide variant, but not IL- Ira. In solution, IL-lra-R polypeptide was more efficiently recognized than the IL-I ra-R polypeptide variant.
Example 7: Expression of IL-lra-R Polvpeptide in Transgenic Mice To assess the biological activity of IL-lra-R polypeptide, a construct encoding an IL-lra-R polypeptide/Fc fusion protein under the control of a liver specific ApoE promoter is prepared. The delivery of this construct is expected to cause pathological changes that are informative as to the function of IL-lra-R polypeptide. Similarly, a construct containing the full-length IL-lra-R polypeptide under the control of the beta actin promoter is prepared. The delivery of this construct is expected to result in ubiquitous expression.
To generate these constructs, PCR is used to amplify template DNA sequences encoding an IL-lra-R polypeptide using primers that correspond to the and 3' ends of the desired sequence and which incorporate restriction enzyme sites to permit insertion of the amplified product into an expression vector.
Following amplification, PCR products are gel purified, digested with the appropriate restriction enzymes, and ligated into an expression vector using standard recombinant DNA techniques. For example, amplified IL-lra-R polypeptide sequences can be cloned into an expression vector under the control of the human p-actin promoter as described by Graham et al., 1997, Nature Genetics, 17:272-74 and Ray et al., 1991, Genes Dev. 5:2265-73.
114 WO 01/42304 PCT/US00/32940 Following ligation, reaction mixtures are used to transform an E. coli host strain by electroporation and transformants are selected for drug resistance.
Plasmid DNA from selected colonies is isolated and subjected to DNA sequencing to confirm the presence of an appropriate insert and absence of mutation. The IL- Ira-R polypcptide expression vector is purified through two rounds of CsCI density gradient centrifugation, cleaved with a suitable restriction enzyme, and the linearized fragment containing the IL-Ira-R polypeptide transgene is purified by gel electrophoresis. The purified fragment is resuspended in 5 mM Tris, pH 7.4, and 0.2 mM EDTA at a concentration of 2 mg/mL.
Single-cell embryos from BDF1 x BDF1 bred mice are injected as described (PCT Pub. No. WO 97/23614). Embryos are cultured overnight in a
CO
2 incubator and 15-20 two-cell embryos are transferred to the oviducts of a pseudopregnant CDI female mice. Offspring obtained from the implantation of microinjected embryos are screened by PCR amplification of the integrated transgene in genomic DNA samples as follows. Ear pieces are digested in 20 mL ear buffer (20 mM Tris, pH 8.0, 10 mM EDTA, 0.5% SDS, and 500 mg/mL proteinase K) at 55 0 C overnight. The sample is then diluted with 200 mL of TE, and 2 mL of the ear sample is used in a PCR reaction using appropriate primers.
At 8 weeks of age, transgenic founder animals and control animals are sacrificed for necropsy and pathological analysis. Portions of spleen are removed and total cellular RNA isolated from the spleens using the Total RNA Extraction Kit (Qiagen) and transgene expression determined by RT-PCR. RNA recovered from spleens is converted to cDNA using the SuperScript M Preamplification System (Gibco-BRL) as follows. A suitable primer, located in the expression vector sequence and 3' to the IL-lra-R polypeptide transgene, is used to prime cDNA synthesis from the transgene transcripts. Ten mg of total spleen RNA from transgenic founders and controls is incubated with 1 mM of primer for 10 minutes at 70 0 C and placed on ice. The reaction is then supplemented with 10 mM Tris- HCI, pH 8.3, 50 mM KCI, 2.5 mM MgCI 2 10 mM of each dNTP, 0.1 mM DTT, and 200 U of SuperScript II reverse transcriptase. Following incubation for minutes at 42 0 C, the reaction is stopped by heating for 15 minutes at 72 0 C and 115 WO 01/42304 PCT/US00/32940 digested with 2U of RNase H for 20 minutes at 37 0 C. Samples are then amplified by PCR using primers specific for IL-lra-R polypeptide.
Example 8: Biological Activity of IL-lra-R Polypeptide in Transgenic Mice Prior to euthanasia, transgenic animals are weighed, anesthetized by isofluorane and blood drawn by cardiac puncture. The samples are subjected to hematology and serum chemistry analysis. Radiography is performed after terminal exsanguination. Upon gross dissection, major visceral organs are subject to weight analysis.
Following gross dissection, tissues liver, spleen, pancreas, stomach, the entire gastrointestinal tract, kidney, reproductive organs, skin and mammary glands, bone, brain, heart, lung, thymus, trachea, esophagus, thyroid, adrenals, urinary bladder, lymph nodes and skeletal muscle) are removed and fixed in buffered Zn-Formalin for histological examination. After fixation, the tissues are processed into paraffin blocks, and 3 mm sections are obtained. All sections are stained with hematoxylin and exosin, and are then subjected to histological analysis.
The spleen, lymph node, and Peyer's patches of both the transgenic and the control mice are subjected to immunohistology analysis with B cell and T cell specific antibodies as follows. The formalin fixed paraffin embedded sections are deparaffinized and hydrated in deionized water. The sections are quenched with 3% hydrogen peroxide, blocked with Protein Block (Lipshaw, Pittsburgh, PA), and incubated in rat monoclonal anti-mouse B220 and CD3 (Harlan, Indianapolis, IN). Antibody binding is detected by biotinylated rabbit anti-rat immunoglobulins and peroxidase conjugated streptavidin (BioGenex, San Ramon, CA) with DAB as a chromagen (BioTek, Santa Barbara, CA). Sections are counterstained with hematoxylin.
After necropsy, MLN and sections of spleen and thymus from transgenic animals and control littermates are removed. Single cell suspensions are prepared by gently grinding the tissues with the flat end of a syringe against the bottom of a 100 mm nylon cell strainer (Becton Dickinson, Franklin Lakes, NJ). Cells are 116 WO 01/42304 PCT/US00/32940 washed twice, counted, and approximately 1 x 106 cells from each tissue are then incubated for 10 minutes with 0.5 pg CD16/32(FcylllIII/I) Fc block in a 20 pL volume. Samples are then stained for 30 minutes at 2-8 0 C in a 100 uL volume of PBS (lacking Ca and Mg), 0.1% bovine serum albumin, and 0.01% sodium azide with 0.5 pg antibody of FITC or PE-conjugated monoclonal antibodies against CD90.2 (Thy-1.2), CD45R (B220), CDI lb(Mac-l), Gr-1, CD4, or CD8 (PharMingen, San Diego, CA). Following antibody binding, the cells are washed and then analyzed by flow cytometry on a FACScan (Becton Dickinson).
Example 9: Functionation of IL-lra-R Polypeptide To evaluate whether IL-Ira-R polypeptide affects cellular responses to IL- I, IL-18, or other cytokines, the following experiments were performed. First, murine bone marrow cells were transduced with the IL-lra-R gene by means of retroviral infection as previously described (Yan et.al.,1999, Exp. Hematol.
27:1409-17). Transduced bone marrow was then used to transplant lethally irradiated recipient mice. Upon hematopoietic recovery, bone marrow and spleen cells from I-lra-R-transduced and control mice mice transduced with empty retroviral vector) were subjected to a series of analyses, including FACS analysis (Figure 12), colony assays (Figures 13A-13B), and y-interferon production in response to either IL-12 (Figure 14) or IL-12 plus TL-18 (Figure The data suggest that IL-lra-R gene expression affects the relative abundance and/or cytokine response of specific subsets of hematopoietic progenitors in the spleen (most prominently G-CSF induced GM-CFC; SCF/Epo induced BFU-E/CFU-Mix; and CSF-I induced M-CFC) and bone marrow (IL-3 and G-CSF induced GM-CFC; SCF/Epo induced BFU-E/CFU-Mix and IL-3/Epo induced GM-CFC/CFU-Mix).
Expression of IL-lra-R polypeptide also correlates with decreased surface expression of CD4 and NK1.1 on splenocytes and with a decreased ability of splenocytes to produce y-interferon in response to IL-12 alone or IL-12 plus IL- 18.
While the present invention has been described in terms of the preferred embodiments, it is understood that variations and modifications will occur to those skilled in the art. Therefore, it is intended that the appended claims cover all such equivalent variations that come within the scope of the invention as claimed.
Throughout the specification, unless the context requires otherwise, the word "comprise" or variations such as "comprises" or "comprising", will be understood to imply the inclusion of a stated integer or group of integers but not the exclusion of any other integer or group of integers.
oo *oo• *0*0* O *D *i 118 EDITORIAL NOTE APPLICATION NUMBER 19450/01 The following Sequence Listing pages 1 to 27 are part of the description. The claims pages follow on pages 119 to 130.
WO 01/42304 WO 0142304PCTIUSOO/32940 SEQUENCE LISTIN!G <110> Saris, Christiaan M.
Giles, Jennifer Mu, Sharon X.
Xia, Min Bass, Michael B.
Craveiro, Roger <120> Interieukin-l Receptor Antagonist-Related Molecules and Uses Thereof <130> 00-1213 <140> <141> <150> 60/170,191 <151> 1999-12-10 <150> 60/188,053 <151> 2000-03-09 <150> 60/194,521 <151> 2000-04-04 <150> 60/195,910 <151> 2000-04-10 <160> 37 <170> Patentln Ver. <210> <211> <212> <213> <220> <22 1> <222> 1 1020
DNA
Homo sapiens
CDS
(64) (522) <400> 1 cagggatcag ggttccagga actcaggatc tgcagtgagg gga atg tgt icc ctc ccc atg gca aga tac tac Met Cys Ser Leu Pro Met Ala Arg Tyr Tyr accagacacc actgattgca ata att aaa tat gca 101 Ile Ile Lys Tyr Ala gac cag aag gct cta tac aca aga gat ggc Asp Gin Lys Ala Leu Tyr Thr Arg Asp Gly 25 cag ctg ctg gtg Gin Leu Leu Val gga gat Gly Asp cct gtt gca Pro Val Ala aga ggc ttg Arg Gly Leu so gac aac tgc tgt Asp Asn Cys Cys gac cgc acc aag Asp Arg Thr Lys gca gag aag atc tgc aca Ala Glu Lys Ile Cys Thr ctt cct aac Leu Pro Asn atc cag gga Ile Gin Gly ccc att tic ctg Pro Ile Phe Leu 1/27 WO 01/42304 WO 0142304PCTJUSOO/32940 ggg agc cgc tgc ctg gca tgt gig gag Gly Ser Arg Cys Leu Ala Cys Val Glu 70 aca gaa gag ggg cct tcc cta Thr Glu Glu Gly Pro Ser Leu cag ctg gag gat gtg aac att gag gaa ctg tac aaa ggt ggt gaa gag Gin Leu Glu Asp Val Asn Ilie Glu Glu Leu Tyr Lys Gly Gly Glu Glu a5 90 gcc aca cgc tic acc ttc tic cag agc agc tca ggc tcc gcc tic agg Ala Thr Arg Phe Thr Phe Phe Gin Ser Ser Ser Gly Ser Ala Phe Arg 100 105 110 cit gag gct gct. gcc tgg cci ggc tgg tic ctg tgt ggc ccg gca gag Leu Glu Ala Ala Ala Ti-p Pro Gly Ti-p Phe Leu Cys Gly Pro Ala Glu 115 120 125 ccc cag cag cca gta cag ctc acc aag gag agt gag ccc tca gcc cgt Pro Gin Gin Pro Val Gin Leu Thr Lys Glu Ser Glu Pro Ser Ala Arg 130 135 140 acc aag ttt tac ttt gaa cag agc tgg tag ggagacagga aacigcgttt Thr Lys Phe Tyr Phe Glu Gin Ser Ti-p 145 150 tagccttgtg cccccaaacc aagctcatcc tgctcagggt ctatggtagg cagaataatg tcccccgaaa tatgtccaca tcctaaiccc aagaictgtg cataigttac catacaigtc caaagaggtt ttgcaaatgt gattatgtta. aggatcttga aatgaggaga caatcctggg ttatccttgt gggctcagtt taatcacaag aaggaggcag gaagggagag tcagagagag aatggaagat accatgcttc taattttgaa gatggagtga ggggccitga gccaacaaat.
gcaggtgttt itagaaggtg gaaaagccaa gggaacggat tctcctctag agtctccgga aggaacacag cicttgacac atggatca gctcagtgac acccatttca gacttctgac ctccacaact ataaaataat aaacttgtgt tattgtaaac ctctaaaaaa aaaaaaaa.
<210> 2 <211> 152 <212> PRT <213> Homno sapiens <400> 2 Met Cys Ser Leu Pro Met Ala Arg Tyr Tyr Ile Ile Lys Tyr Ala Asp 1 5 10 Gin Lys Ala Leu Tyr Thr Arg Asp Gly Gin Leu Leu Val Gly Asp Pro 25 Val Ala Asp Asn Cys Cys Ala Glu Lys Ile Cys Thi- Leu Pro Asn Arg 40 Gly Leu Asp Arg Thr Lys Val Pro Ie Phe Leu Gly Ile Gin Gly Gly 55 Ser Arg Cys Leu Ala Cys Val Glu Thr Glu Glu Gly Pro Ser Leu Gin 70 75 2/27 348 396 444 492 542 602 662 722 782 842 902 962 1020 WO 01/42304 WO 0142304PCTIUSOO/32940 Leu Giu Asp Val Asn Ile Giu Glu Leu Tyr Lys 90 Thr Arg Phe Thr Phe Phe Gin Ser Ser Ser Gly 100 105 Giu Ala Ala Ala Trp Pro Gly Trp Phe Leu Cys 115 120 Gin Gin Pro Val Gin Leu Thr Lys Glu Ser Giu 130 135 Lys Phe Tyr Phe Giu Gin Ser Trp 145 150 <210> 3 <211> 1020 <212> DNA <213> Homo sapiens <220> <221> CDS <222> (64)..(522) <400> 3 cagggatcag ggttccagga actcaggatc tgcagtgagg Gly Gly Giu Ser Ala Phe 110 Gly Pro Ala 125 Pro Ser Ala 140 Giu Ala Arg Leu Giu Pro Arg Thr accagacacc actgattgca gga atg tgt tcc etc ccc atg gca aga tac tac ata att aaa tat Met Cys Ser Leu Pro Met Ala Arg Tyr Tyr Ile Ile Lys Tyr gac Asp ect Pro aga Arg ggg Gly cag Gin gcc Ala ctt Leu aag get Lys Ala gca gac Ala Asp ttg gcc Leu Ala cgc tgc Arg Cys gag gat Giu Asp cgc ttc Arg Phe gct gct Ala Ala cta tac aca.
Leu Tyr Thr aac tgc tgt Asn Cys Cys cgc acc aag Arg Thr Lys cig gca tgt Leu Ala Cys gtg aac at Val Asn Ile 85 acc ttc ttc Thr Phe Phe 100 gcc tgg ccL Ala Trp Pro aga Arg gca Ala gtc Val gtg Val gag Giu cag Gin Gly gat ggc cag Asp Gly Gin 25 gag aag ate Giu Lys Ile 40 CCC att ttc Pro Ile Phe gag aca gaa Giu Thr Giu gaa ctg tac Giu Leu Tyr agc agc tca Ser Ser Ser 105 tgg ttC ctg Trp Phe Leu 120 ctg Leu tgc cys Ctg Leu gag Giu aaa.
Lys ggc Gly tgt Cys gtg Val ctt Leu ate Ile cct Pro ggt Gly 5CC Ala CCg Pro 125 gga Gly ct Pro eag Gin Lee Ser gaa.
Giu ttc Phe 110 gca.
Ala 3/27 WO 01/42304 WO 0142304PCT/USOO/32940 ccc cag cag cca gta cag ctc acc aag gag agt gag ccc tca gcc cgt 492 Pro Gin Gin Pro Val Gin Leu Thr Lys Giu Ser Glu Pro Ser Ala Arg 130 135 140 acc aag ttt tac ttt gaa cag agc tgg tag ggagacagga aactgcgttt 542 Thr Lys Phe Tyr Phe Giu Gin Ser Trp 145 150 tagccttgtg cccccaaacc aagctcatcc tgctcagggt ctatggtagg cagaataatg 602 tcccccgaaa tatgtccaca tcctaatccc aagatctgtg catatgttac catacatqtc 662 caaagaggtt ttgcaaatgt gattatgtta aggatcttga aatgaggaga caatcctggg 722 ttatccttgt gggctcagtt taatcacaag aaggaggcag gaagggagag tcagagagag 782 aatggaagat accatgcttc taattttgaa gatggagtga ggggccttga gccaacaaat 842 gcaggtgttt ttagaaggtg gaaaagccaa gggaacggat tctcctctag agtctccgga 902 aggaacacag ctcttgacac atggatttca gctcagtgac acccatttca gacttctgac 962 ctccacaact ataaaataat aaacttgtgt tattgtaaac ctctaaaaaa aaaaaaaa 1020 <210> 4 <211> 152 <212> PRT <213> Homo sapiens <400> 4 1 Gin Cys Ser Leu Pro Met Ala Arg Tyr Lys Ala Leu Tyr Thr Arg Asp Tyr Ile Ile Lys 10 Gin Leu Leu Val Ile Cys Ile Leu Tyr Ala Asp is Gly Asp Pro Pro Asn Arg Val Ala Asp Asn Cys Cys Ala Gly Leu Ser Arg Ala Arg Thr Lys Val Ile Phe Leu Gly Ile Gin Gly Gly Cys Leu Ala Leu Cys Val Giu Thr Giu Giu Gly Pro Ser 70 75 Ile Glu Giu Leu Tyr Lys Gly Gly Giu Giu Asp Val Leu Gin Giu Ala Arg Leu Thr Arg Phe Glu Ala Ala 115 Gin Gin Pro Phe Phe Gin Ser Ser Ser Gly Ser Ala Phe 110 Ala Giu Pro Trp, Pro Gly Phe Leu Cys Gly Val Gin Leu 130 Lys Phe 145 Thr Lys 135 Ser Trp Glu Ser Giu Ser Ala Arg Thr Tyr Phe Giu 4/27 WO 01/42304 WO 0142304PCTIUSOO/32940 <210> <211> 744 <212> DNA <213> H-omo sapiens <220> <221> CDS <222> (58)..(573) <400> gctcccgcca ggagaaagga acattctgag gggagtctac accctgtgga gctcaag atg gtc ctg agt Met Val Leu Ser 1 att gca gga atg Ile Ala Gly met tat gca gac cag Tyr Ala Asp Gin gga gat cct gtt Gly Asp Pro Val cct aac aga ggc Pro Asn Arg Gly cag gga. ggg agc Gin Gly Gly Ser tcc cta. cag ctg Ser Leu Gin Leu 100 gaa gag gcc aca Giu Giu Ala Thr 115 ttc agg ctt gag Phe Arg Leu Giu 130 gca gag ccc cag Ala Giu Pro Gin 145 gcc cgt acc aag Ala Arq Thr Lvs gcg ctg tgc ttc cgt Aia Leu Cys Phe Arg gag gac Glu Asp ccc Pro tac Tyr 40 tgc Cys acc Thr gca Ala aac Asn ttc Phe 120 tgg Trp cag Gin gaa Giu cag aca cca ctg Gin Thr Pro Leu tac ata att aaa Tyr Ile Ile Lys cag ctg ctg gtg Gin Leu Leu Val atc tgc ata. ctt Ile CysIle Leu ttc ctig ggg atc Phe Leu Giy Ile s0 gaa gag ggg cct Giu Giu Gly Pro tac aaa. ggt ggt Tyr Lys Gly Giy 110 tca ggc tcc gcc Ser Gly Ser Aia 125 ctg tgt ggc ccg Leu Cys Gly Pro agt gag ccc tca Ser Giu Pro Ser 160 ggagacagga.
Phe Tyr Phe 165 Gin Ser Trp 170 aactgcgttt tagccttgtg cccccaaacc aagctcatcc cagaataatg tcccccgaaa tatgtccaca tcctaatccc 5/27 tgctcagggt ctatggtagg 643 aagatctgtg catatgttac 703 WO 01/42304 catacatgtc caaagaggtt ttgcaaatgt gattatgtta a <210> 6 <211> 171 <212> PRT <213> Homto sapiens PCTUSOOI32940 <400> 6 Met Val 1 Ile Ala Tyr Ala Gly Asp Pro Asn Gin Gly Ser Leu Giu Glu Phe Arg 130 Ala Glu 145 Ala Arg Ser Met Gin Val Gly Ser Leu 100 Thr Glu Gin Lys Cys Phe Pro Met Tyr Thr Cys Cys Thr Lys Ala Cys Asn Ile 105 Phe Phe i2 0 Trp Pro Gin Leu Glu Gin Leu Lys Val Leu Ile Pro Gly Aia Pro Ser 160 7 <211> 269 <212> PRT <213> Homo sapiens <400> 7 Met Ala Giu Val Pro Lys Leu Ala Ser Glu Met Met Ala Tyr Tyr Ser 1 5 10 is Gly Asn Giu Asp Asp Leu Phe Phe Glu Ala Asp Gly Pro Lys Gin Met 25 Lys Cys Ser Phe Gin Asp Leu Asp Leu Cys Pro LeU Asp Gly Giy Ilie 40 Gin Leu Arg Ilie Ser Asp His His Tyr Ser Lys Gly Phe Arg Gin Ala 55 6/27 WO 01/42304 WO 0142304PCTIUSOO/32940 Ala Ser Val Val Val Ala Met Asp Lys Leu 70 Lys Met Leu Val Pro Thr Glu 1.00 Asp Lys Gin Gin Glu 180 Leu Glu Phe Giu Thr 260 Gin Pro Pro Leu Gin 150 Giu Asn Leu Arg Ser 230 Met Phe Thr Trp Cys Tyr 140 Val Ile Val Lys Ile 220 Trp Gly Ser <210> 8 <211> 153 <212> PRT <213> Homo sapiens <400> 8 Ala Pro Val Arg Ser Leu Asn Cys Thr Leu Arg Asp Ser Gin Gin Lys 1 S 10 Ser Leu Val Met Ser Gly Pro Tyr Giu Leu Lys Ala Leu His Leu Gin 25 Gly Gin Asp Met Glu Gin Gin Val Val Phe Ser Met Ser Phe Vai Gin 40 Giy Giu Giu Ser Asn Asp Lys Ile Pro Vai Ala Leu Gly Leu Lys Glu 55 Lys Asn Leu Tyr Leu Ser Cys Val Leu Lys Asp Asp Lys Pro Thr Leu 70 75 7/27 WO 01/42304 PCT/US00/32940 Gin Leu Glu Ser Val Asp Pro Lys Asn Tyr Pro Lys Lys Lys Met Glu 90 Lys Arg Phe Val Phe Asn Lys Ile Glu Ile Asn Asn Lys Leu Glu Phe 100 105 110 Glu Ser Ala Gin Phe Pro Asn Trp Tyr Ile Ser Thr Ser Gin Ala Glu 115 120 125 Asn Met Pro Val Phe Leu Gly Gly Thr Lys Gly Gly Gin Asp Ile Thr 130 135 140 Asp Phe Thr Met Gin Phe Val Ser Ser 145 150 <210> 9 <211> 177 <212> PRT <213> Homo sapiens <400> 9 Met Glu Ile Cys Arg Gly Leu Arg Ser His Leu Ile Thr Leu Leu Leu 1 Phe Ser Tyr Val Leu Ser Leu Asp Phe 145 Met Glu Thr Ile Arg Ile Leu Val Ile Asp Gly Gly Leu Gin Gin Asp 120 Ser Phe 135 Glu Ala Met Val <210> <211> 155 8/27 WO 01/42304 PCTIUSOO/32940 <212> PRT <213> HOMO sapiens <400> Met Val Leu Ser Gly Ala Leu Cys Phe Arg Met Lys Asp Ser Ala Leu 1. 5 10 Lys Val Leu Tyr Leu His Asn Asn Gin Leu Leu Ala Gly Gly Leu His 25 Ala Gly Lys Val Ile Lys Gly Giu Giu Ile Ser Val Val Pro Asn Arg 40 Trp Leu Asp Ala 3cr Leu Ser Pro Val Ile Leu Gly Val Gin Gly Gly so 55 Ser Gin Cys Leu Ser Cys Gly Val Giy Gin Glu Pro Thr Leu Thr Leu 70 75 s0 Giu Pro Val Asn Ile Met Giu Leu Tyr Leu Gly Ala Lys Glu Ser Lys 90 Ser Phe Thr Phe Tyr Arg Arg Asp Met Gly Leu Thr Ser Ser Phe Giu 100 105 110 Ser Ala Ala Tyr Pro Gly Trp Phe Leu Cys Thr Val Pro Glu Ala Asp 115 120 125 Gin Pro Val Arg Leu Thr Gin Leu Pro Giu Asn Gly Gly Trp Asn Ala 130 135 140 Pro Ile Thr Asp Phe Tyr Phe Gin Gin Cys Asp 145 150 155 <210> 11 <211> 178 <212> PRT <213> Homo sapiens <400> 11 Met Ser Phe V61 Gly Glu Asn Ser Gly Val Lys Met Gly Ser Giu Asp 1 5 10 Trp Glu Lys Asp Glu Pro Gin Cys Cys Leu Giu Asp Pro Ala Gly Ser 25 Pro Leu Giu Pro Gly Pro Ser Leu Pro Thr Met Asn Phe Val His Thr 40 Lys Ile Phe Phe Ala Leu Ala Ser Ser Leu Ser Ser Ala Ser Ala Glu 55 Lys Gly Ser Pro Ile Leu Leu Gly Vai Ser Lys Gly Glu Phe Cys Leu 70 75 Tyr Cys Asp Lys Asp Lys Gly Gin Ser His Pro Ser Leu Gin Leu Lys 90 Lys Giu Lye Leu Met Lys Leu Ala Ala Gin Lys Glu Scr Ala Arg Arg 100 105 110 9/27 WO 01/42304 WO 0142304PCTIUSOOI3 2940 Pro Phe Ser Ala 130 Glu Pro 145 Phe Ser Ser Asp Ile Phe Tyr Arg Ala Gin Val Gly Ser Trp Asn Met 115 120 125 Ala His Pro Gly Trp Phe Ile Cys Thr Ser Cys Asn 135 140 Val Gly Val Thr Asp Lys Phe Glu Asn Arg Lys His 150 155 Phe Gin Pro Val Cys Lys Ala Glu Met Ser Pro Ser 165 170 Leu Glu Cys Asn Ile Glu 160 Glu Val 175 <210> 12 <211> 218 <212> PRT <213> Homo sapiens <400> 12 met Ser Phe Val Gly Glu Asn Ser Gly Val Lys Met Gly Ser Giu Asp 1 Trp Pro Ser Gin Pro Ser Val Ser Ala 145 Val Ile Glu Leu Pro Asp Asp Leu Ser His 130 Gin Gly Cys Gin Ser Leu Val Arg Ala Cys Leu 135 Arg Leu Cys Cys Leu Asn Leu Pro Glu Leu 120 Lys Arg Glu Asn Pro Phe Ser Leu Ala Ile Asp 125 Met Tyr Pro Val Ser Thr Asp Val Scr Gly Gin Ala Gin 160 Phe Lys Phe Glu Asn Arg Lys His Ile Glu Phe Ser Phe Gin Pro Val Cys Lys 195 200 205 10/27 WO 01/42304 WO 0142304PCTIUSOO/32940 Ala Glu Met Ser Pro Ser Glu Val Ser Asp 210 215 <210> 13 <211> 192 <212> PRT <213> Homo sapiens <400> 13 <210> 14 <211> 169 <212> PRT <213> Homo sapiens <400> 14 Met Arg Gly Thr Pro Gly Asp Ala Asp Gly Gly Gly Arg Ala Val Tyr 1 5 10 Gin Ser Met Cys Lys Pro Ile Thr Gly Thr Ile Asn Asp Leu Asn Gin 11/27 WO 01/42304 WO 0142304PCTIUSOO/32940 Gin Val Asp Ser Glu Ala Asn Pro Leu Gin Pro Val Thr Leu 130 Arg Asp 145 Thr Ala <210> <211> 208 <212> PRT <213> Homo sapiens <400> Met Arg Gly Thr Pro 1 5 Gin Ser Ser Glu Ser Pro Ser Ala Leu Thr Pro Leu Cys Thr LeU Thr Ile Asn Asp Leu Leu Val Ala Val Pro Val Ile Thr Cys Lys 100 Pro Ilie Tyr Leu Gly 115 Lys Val Gly Glu Gin iLeu kVal )Pro t Lys -Asp Ala Trp *Glu Gly Met *Glu Val *Trp *Val LLeu Glu iLeu 12/27 WO 01/42304 PCTUSOO/32940 130 135 140 Asp Leu Tyr Gly Gin Pro Giu Pro Val Lys Pro Phe Leu Phe Tyr Arg 145 150 155 160 Ala Lys Thr Gly Arg Thr Ser Thr Leu Giu Ser Val Ala Phe Pro Asp 165 170 175 Trp Phe Ile Ala Ser Ser Lys Arg Asp Gin Pro Ile Ile Leu Thr Set- .180 185 190 Glu Leu Gly Lys Set- Tyr Asn Thr Ala Phe Glu Leu Asn Ile Asn Asp 195 200 205 <210> 16 <211> 158 <212> PRT <213> Homo sapiens <400> 16 Met Giu Lys Ala Leu Lys Ile Asp Thr Pro Gin Gin Gly Set- Ile Gin 1 5 10 Asp Ile Asn His Arg Val Trp, Val Leu Gin Asp Gin Thr Leu Ile Ala 25 Vai Pro Arg Lys Asp Arg Met Set- Pro Vai Thr Ile Ala Leu Ile Ser 40 Cys Arg His Val Giu Thr Leu Giu Lys Asp Arg Gly Asn Pro Ile Tyr 55 Leu Giy Leu Asn Gly Leu Asn Leu Cys Leu Met Cys Ala Lys Val Gly 70 75 Asp Gin Pro Thr Leu Gin Leu Lys Glu Lys Asp Ile Met Asp Leu Tyr 90 Asn Gin Pro Giu Pro Vai Lys Ser Phe Leu Phe Tyr His Set- Gin Ser 100 105 110 Gly Arg Asn Set- Thr Phe Giu Set- Val Ala Phe Pro Gly Trp, Phe Ile 115 120 125 Ala Val Set- Set- Giu Gly Gly Cys Pro Leu Ile Leu Thr Gin Giu Leu 130 135 140 Gly Lys Ala Asn Thr Thr Asp Phe Gly Leu Thr Met Leu Phe 145 150 155 <210> 17 <211> 157 <212> PRT <213> Homo sapiens <400> 17 13/27 WO 01/42304 WO 0142304PCTIUSOO/32940 Met Asn Pro Gin Arg Giu Ala Ala Pro Lys Ser Tyr Ala Ile Arg Asp 1 S 10 Ser Arg Gin Met Val Trp, Val Leu Ser Gly Asn Ser Leu Ile Ala Ala 25 Pro Leu Ser Arg Ser Ile Lys Pro Val Thr Leu His Leu Ile Ala Cys 40 Arg Asp Thr Giu Phe Ser Asp Lys Giu Lys Giy Asn Met Val Tyr Leu 55 Gly Ile Lys Gly Lys Asp Leu Cys Leu Phe Cys Ala Giu Ile Gin Gly 70 75 Lys Pro Thr Leu Gin Leu Lys Giu Lys Asn Ile Met Asp Leu Tyr Val 90 Glu Lys Lys Ala Gin Lys Pro Phe Leu Phe Phe His Asn Lys Giu Gly 100 105 110 Ser Thr Ser Val Phe Gin Ser Val Ser Tyr Pro Gly Trp Phe Ile Ala 115 120 125 Thr Ser Thr Thr Ser Gly Gin Pro Ile Phe Leu Thr Lys Giu Arg Gly 130 135 140 Ile Thr Asn Asn Thr Asn Phe Tyr Leu Asp Ser Val Giu 145 150 155 <210> 18 <211> 11 <212> PRT <213> Human immunodeficiency virus type 1 <400> 18 Tyr Gly Arg Lys Lys Arg Arg Gin Arg Arg Arg 1 5 <210> 19 <211> <212> PRT <213> Artificial Sequ~ence <220> <223> Description of Artificial Sequence: internalizing domain derived from HIV tat protein <400> 19 Gly Gly Gly Gly Tyr Gly Arg Lys Lys Arg Arg Gin Arg Arg Arg 1 5 10 <210> <211> 23 <212> DNA <213> Artificial Sequence <220> 14/27 WO 01/42304 PCT/US00/32940 <223> Description of Artificial Sequence: Oligonucleotide 2349-98 <400> cacacgcttc accttctttc cag 23 <210> 21 <211> 23 <212> DNA <213> Artificial Sequence <220> <223> Description of Artificial Sequence: Oligonucleotide 2349-99 <400> 21 taaaacttgg tacgggctga ggg 23 <210> 22 <211> 23 <212> DNA <213> Artificial Sequence <220> <223> Description of Artificial Sequence: Oligonucleotide 1572-36 <400> 22 gtgtggaatt gtgagcggat aac 23 <210> 23 <211> 23 <212> DNA <213> Artificial Sequence <220> <223> Description of Artificial Sequence: Oligonucleotide 2328-91 <400> 23 ctatgaccat gattacgcca age 23 <210> 24 <211> <212> DNA <213> Artificial Sequence <220> <223> Description of Artificial Sequence: Oligonucleotide 2351-47 <400> 24 gctgtactgg ctgctggggc <210> <211> 21 15/27 WO 01/42304 PCT/US00/32940 <212> DNA <213> Artificial Sequence <220> <223> Description of Artificial Sequence: Oligonucleotide 2351-48 <400> ccttcaggct tgaggctgct g 21 <210> 26 <211> 21 <212> DNA <213> Artificial Sequence <220> <223> Description of Artificial Sequence: Oligonucleotide 2329-93 <400> 26 cgggcctctt cgctattacg c 21 <210> 27 <211> 21 <212> DNA <213> Artificial Sequence <220> <223> Description of Artificial Sequence: Oligonucleotide 2363-04 <400> 27 cctggctggt tcctgtgtgg c 21 <210> 28 <211> 21 <212> DNA <213> Artificial Sequence <220> <223> Description of Artificial Sequence: Oligonucleotide 2329-94 <400> 28 tggcgaaagg gggatgtgct g 21 <210> 29 <211> 27 <212> DNA <213> Artificial Sequence <220> <223> Description of Artificial Sequence: Oligonucleotide AP-1 <400> 29 ccatcctaat acgactcact atagggc 27 16/27 WO 01/42304 PCT/US00/32940 <210> <211> 22 <212> DNA <213> Artificial Sequence <220> <223> Description of Artificial Sequence: Oligonucleotide 2353-87 <400> ccttggtgag ctgtactggc tg 22 <210> 31 <211> 19 <212> DNA <213> Artificial Sequence <220> <223> Description of Artificial Sequence: Oligonucleotide 2349-52 <400> 31 ccgggccaca caggaacca 19 <210> 32 <211> 24 <212> DNA <213> Artificial Sequence <220> <223> Description of Artificial Sequence: Oligonucleotide 2349-51 <400> 32 aagaggccac acgcttcacc ttct 24 <210> 33 <211> 26 <212> DNA <213> Artificial Sequence <220> <223> Description of Artificial Sequence: Oligonucleotide 2557-95 <400> 33 aagccttttt cttctttgcc tcagtg 26 <210> 34 <211> 26 <212> DNA <213> Artificial Sequence <220> <223> Description of Artificial Sequence: 17/27 WO 01/42304 PCT/USOO/32940 oligonucleotide 2557-96 <400> 34 tgccatttaa tgtaacaegg tcacag 26 <210> <211> 459 <212> DNA <213> Mus musculus <220> <221> CDS <222> <400> atg tgc tee ctt ccc atg gca aga tac tac ata atc aag gat gca cat 48 Met Cys Ser Leu Pro Met Ala Arg Tyr Tyr Ie Ile Lys Asp Ala His 1 5 10 caa aag get ttg tac aca cgg aat ggc cag etc ctg ctg gga gac cet 96 Gin Lys Ala Leu Tyr Thr Arg Asn Gly Gin Leu Leu Leu Gly Asp Pro 25 gat tca gac aat tat agt cca gag aag gte tgt ate ctt ect aae cga 144 Asp 5cr Asp Asn Tyr Ser Pro Giu Lys Val Cys Ile Leu Pro Asn Arg 40 ggc eta gac cgc tec aag gte ccc atc ttc ctg ggg atg cag gga gga 192 Giy Led Asp Arg 5cr Lys Val Pro Ile Phe Leu Gly Met Gin Gly Gly 55 agt tgc tgc ctg gcg tgt gta aag aca aga gag gga cct ctc ctg cag 240 Cys Cys Leu Ala Cys Val Lys Thr Arg Giu Gly Pro Leu Leu Gin 70 75 ctg gag gat gtg aac atc gag gac cta tac aag gga ggt gaa caa ace 288 Leu Giu Asp Val Asn Ile Glu Asp Leu Tyr Lys Gly Giy Giu Gin Thr 90 ace egt ttc ace ttt tte eag aga age ttg gga tet gee ttc agg ctt 336 Thr Arg Phe Thr Phe Phe Gin Arg 5cr Leu Giy Ser Aia Phe Arg Leu 100 105 110 gag gct get gee tgc cct ggc tgg ttt etc tgt gge eea get gag ccc 384 Glu Ala Ala Ala Cys Pro Giy Trp Phe Leu Cys Gly Pro Ala Giu Pro 115 120 125 eag cag cca gtg cag ctc ace aaa gag agt gaa ccc tee ace cat act 432 Gin Gin Pro Vai Gin Leu Thr Lys Giu Ser Giu Pro Ser Thr His Thr 130 135 140 gaa ttc tac ttt gag atg agt egg taa 459 Giu Phe Tyr Phe Glu Met Ser Arg 145 150 <210> 36 <211> 152 <212> PRT <213> MuS museulus 18/27 WO 01/42304 WO 0142304PCT[USOO/32940 <400> 36 Met Cys Ser Leu Pro met Ala Arg Tyr Tyr Ile Ile Lys Asp Ala His 1 5 10 Gin Lys Ala Leu Tyr Thr Arg Asn Gly Gin Leu Leu Leu Gly Asp Pro 25 Asp Ser Asp Asn Tyr Ser Pro Glu Lys Val Cys Ile Leu Pro Asn Arg 40 Gly Leu Asp Arg Ser Lys Val Pro Ile Phe Leu Gly Met Gin ly Gly 55 Ser Cys Cys Leu Ala Cys Val Lys Thr Arg Glu Gly Pro Leu Leu Gin 70 75 Leu Giu Asp Val Asn Ie Giu Asp Leu Tyr Lys Gly Gly Glu Gin Thr 90 Tbr Arg Phe Thr Phe Phe Gin Arg Ser Leu Gly Ser Ala Phe Arg Leu 100 105 110 Glu Ala Ala Ala Cys Pro Gly Trp Phe Leu Cys Gly Pro Ala Glu Pro 115 120 125 Gin Gin Pro Val Gin Leu Thr Lys Giu Ser Glu Pro Ser Thr His Thr 130 135 140 Glu Phe Tyr Phe Glu Met Ser Arg 145 150 <210> 37 <211> 13017 <212> DNA <213> Mus nmsculus <220> <221> exon <222> (8025)..(8054) <220> <221> exon <222> (9548)..(9631) <220> <221> exon <222> (9967)..(10092) <220> <221> exon <222> (10358)..(10570) <400> 37 actagtctcc catagacaac agctgaatgt acgaggtcag aagcaaggcc tgccccagaa ccattgcaag ccaggtgctg tcttgattgt agcctcataa aaaactgatg cagaattgcc 120 ccaccaacat gctccagatt cctgctccac agaaaccctg tgaactaacc atgttgcttt 180 19/27 WO 01/42304 tagattctgc gtagttactt attgacacca gttaactgtt cctgcacacc caaaagaggg cagcttgaca gcaacctctg tctaccacag ccaggcaggg caacactact aggtcacctc tattctgtgc taaggt tgga agaacatggt gtttgaatcc cagaatttaa gaaatagaaa tggtggtggt cacagtaaag actacctact gggcctatat gaaatgtttc aaaaagcaaa tacattttgc caaatattaa cctggtctta cataccatac tgctctgttt agttccttgg PCTIUSOO/32940 agtaagttga tattatgatc gcctttcact acctgttcc aggatgtttc ggtccatatc ggtttacagt cttgtcctag acctctcaga tttctgcaca ttgggacact ctataggaag ttatctccct gctgcaccaa ttgtacagaa caactcattc aaatacttta cacacttgtg ggtgatgqtg tgaggcatca actgatttta tttcagagag ataatcaggt atccaaccca tttttattat ccataaaaat gagttgttgg atttggccac tctcatatgc caacagagat taatctgcag aaaactttat aacccaaata aaaaggtaca tcatatttct agaaccccag ttgtagaagg aaccttczagt gttcctgagg cccccttcac gtacccatcc tatttgaaaa gtaaaaagtt tatgttttta tgtactttcc ttgaaggcca t cccagggaa gtggcggtgg gtggtggtgg tggcctgaga gagttctata gaaatgttca catcagagaa cctcatgctt tataaattac aaaaaattcc ggcaaaacct ttggagagag ccttggctta ggtacgctat taaataacat ttctccactc gcctatttaa attccctttc acctaaaaca gtatttagct accagaaaga ccatatacat atgcagaccc actgcctgac cctcgaccta gatgagagtc ggcttggggt agcacccggc tccactcata ccttttgtag gctcacactt tggtggtgat tggtggtggt gagtcaggca attttagtag cagcaggtca aaggcaaagg aatqcattca ctggagaata atgtgtgctt gacctttgaa tctaatgtca cagctaacta gtgtctttga tcgatgaaag tttccatttt atgctgatca gaccatagct ttggggacta gtaaaactca aggtagccaa ctaagctccc caggacactg tgggagt tag cagaaaccat atggtcat tt ctctggcatg ataatgct tc caaacccttg ggtgacagaa ctaaatccag ggtggtcgtg ggtcgtggtg tcacagctat cagccacagg actgcagaca agctgatgga aagg tc tgcg tttttgtctg ctcccagggg gtagttact t catctaaagg tggctccagc cacagcagaa agaaacatgt ccttctcaga tacttctctt gcatctccca caagtgaaag cttgtcaggc gacagaagag cagcaccatt acctcagttt tctcatggtg tcacttttca gctatgataa catctgacct gcaaaatttc taaaagagta t ttaaaaata aatgaaagaa gtggtggtgg gtgtaatgat tcaagtgaaa cctggggcct gtgaagatca ctttatcctg ggcagaagaa aattatctcc ctataaagcc ttgaagatgc gttactctga taaactataa taaatgctta 240 300 360 420 480 540 600 660 720 780 840 900 960 1020 1080 1140 1200 1260 1320 1380 1440 1500 1560 1620 1680 1740 1800 1860 1920 1980 gtgaacatta ctgattgcct gacaggacac ctcacacttt ggtactttca acagagggat 2040 20/27 WO 01/42304 WOOI/2304PCTIUSOOI32940 gtaaacttat ccaattctag ctttacatcc cctccccttc ttaagtctct aaaaagcata gatccacatq cctgtgtatg tgactctgtt atccttctcc atctccccag aaaaaaattt acataaactg aaatagcttt tgttagttta cattttttct cctcatcaag agagaacgtt atggataaat atgacttact tgcaaagtca ctagagtcca ctacagctga aaaggagttc tcattgtggt ttccaacctc cattgtttat agcaggtcag atactcatta gaagaacaat ctctgaaatc caattgctgt tcctctgaga gcagggctag tcc cacagac aaggctgagc ttctttggtt ggtcttcctg ttctcacttc ggcaacacat aacaaggaat ga t tctaa ta cactaccttg aaaccacaac catttcttgc acctttgtgg c tg tccaat t ggactgatgt caacagatga actggagaac gggcccatgg gtttccttac Cttctgatct tctctggatg ttcaggtctc gtagctgcaa tgtctggcct atctccccac gaagaatgaa atttttaggt cctccctcca gaatggagaa acacttcccc agacaacagc tgcacatctg ggtggttcag tggacaccct cataagagtg ggaaatgaaa aagaatcttg gaatgcaatt cacaaaatct aataacaaca tctgtgatcg aaatttgcaa ctttatccct acagataaat gtagaaggat tgagtctctt ctcctgtgcc atatgtgtct tcccaaacag catctggctt agacagaacc aaccactaat ctgtcaaggc ctccttatta tattggcaat agtgggaagt agttccccac ccctcctgga cag tgagg cc ttttgggata ctacatatga actctgagag gtccccttcc tgaggagtct gattgtgaaa tttctctgaa ggattgtaat cttggcatgt aaatagcaaa tctgggtctt atgtcccaaa agtgatggat gaaggaatat gagaaataga gaccatgcac atccccatgc cagtaagttt agccacactc ccatcagcat ccatggagca aacacagtca tttatgagtg tttggactgt aaaagcaaaa ctttttgttt caccaccaca tattccccca agtcagggca gccccgttcc atigaggaggc ccccaagggt agcccacaat ttaaaaacat agtaatttaa aatgcttaag gaaaacctat gtgttgttgg aattgggtct aagctgacac aaggagaat t gagtaaagga gtacatggt t tggacagctg tgtccagggt aaatctaagg gtatcaacta gtgatgaagt aatct t tcta tcaaagaggt atgacagtag actctctccc gttgaagata attggttaac tgtttattca gtcctttttc tcatgaaaca gcccagctag agttgtttag ctaggtccag ccaggtcagt ccttccccta gaagcatttt agaaaaagaa agtgtggaaa caaagttatg caaattttct cag cc tcat t ctcaccaatt acaataagtc tgtataagag aggtggatag gactgaggca ctcatattcc ttaatacgtt attaaatctg cagccctgc t ttcttgatc ttgaccccag ctacagagac cttcccgcaa ttatgaaatc 21.00 2160 2220 2280 2340 2400 2460 2520 2580 2640 2700 2760 2820 2880 2940 3000 3060 3120 3180 3240 3300 3360 3420 3480 3540 3600 3660 3720 3780 tctgggctct tcttcccgga tctagagcca attacagatt ctgtaggttt gacccaccct 3840 21/27 WO 01/42304 gaccag acat ttagaatcca cttacacaca catacacata ggctctaggg aatacttgat tggaaccaca aaaatgtttt tagacagtag tccaaatggt ataatcttgt aaaaacaaaa taagttattt gatcttaagg acatcatgct tgcacactaa gaagggaaga tacatag tt t actggcctgt taagcccaag gtgatgaaga gagagaatta gccagatggt agagatgtac aatggtgact gaggtatatt aaagagaag t acacccaaag taaacaaaat cttaggctat caatgtgcct PCTIUSOO/32940 ta taaacaca ggcaccagga cacacataca caacacatac aagaagaaat agaatttaat gagttttgaa gtttgtgaca ggatgccacc ttctttgaaa ccagcctctc caaacaaaat ttagaatcca tgctgtgtct tcaagatttc taaacaatat attttgttat gacatagaaa gtttttaaga tatagtgaag ggggtggatt gcatgagtcc gtcaagatag tttatgtagg aaaaacaagc ttcagtgccg ggattgtgga atgaaggtgt tgc tcatcac tctcttacaa gtgtctgttc gtgctggtgc actacaggtc cacacacaac acacacacaa gtcttttaga gagaag tata agtcatctca tatggaagct aaggtgctaa acaatctcct atttcaatgg aaaaacacca gcct ttcagt gtcagttgta aggatcttat agcaaagttt gatagtaaag gaaccttggc tctcattctc ccatgtccag ttgagcagga ttgagagaaa gtggccatcg ttaggtttga acaaggttat gaaagaggaa ggagcagatg gctgtggact agctgtacaa ctctcagcct ttacttcctc cctgaagaaa ag tggtgaca acaacataca cacttt t ctg gaatgcggtg gattaggtca aaagaaatta cccatggaga cgtcttcatc tgtgaaactt gaatagattg aaaaaaaaac caaagcttga tagttggata aacttttata gacaggagtt gggaaaatca aaccacataa agctgttatg tgatcttaat ataccaaaag aatgagatgc tggactttag ttcagaaacc actttaaaga tattacaaaa ctcaccacgc gctgaagctc gataticcat tatgaatgac tgccacaagc 22/27 acagttggag gtcggtctct cacacataca taatgtctcc tgatgttcta aagcaagggt tttaggccag cattctgtga accccatcat aaagtagcct aaggcctaag ccataaaatg ttcatgcata gaggtacaga aagcaaataa cagagtac tg aattttgagt tctaatgcat taactgaaca aggagtgaca caatgctgac tat tgcacaa aaccaggcag aggagggtta aataatctct gtgagaggag ccttacactc agcctgtggc agcatataaa ccggaaggaa aaaagagcct actccaggca ctgtgtatct acacacaaca aaaattctca taagtctagg actacatatt agatgttcaa tt ct cat caa ctatcataca tgaaaatata gaccaaaaca aatgagtagc tctgtgttct tgagctatat tttgtcttaa ttagagaagt catggaatca gagcccaaga gacaagatac ggaatggttg tgtgcccttg gcaacctagg gaatgtgatc gcatgtt tac gaaaagaagg tagatttgag acttgaactg tgggaagcag aataaaagtg aagaactcta tgggaat tgg 3900 3960 4020 4080 4140 4200 4260 4320 4380 4440 4500 4560 4620 4680 4740 4800 4860 4920 4980 5040 5100 5160 5220 5280 5340 5400 5460 5520 5580 5640 5700 WO 01/42304 WO 0142304PCT/USOO/3 2940 ctcagaggga acgtcatcaa acaggctggc cttgaggctg ggctgttatt 9gatagagga ttaagacctg acactgtttt gggtaaatgt tgccgtagag gaatctctag ccaatacctg ctcacacata tggcctgaca aataaggata tttaaacaaa caggaaaatg tataacaagc attgggataa tgtttgtgcc cctttcagcc attgaactat aacaacacat tcgtgcagaa gtgactttta gtgtggqtt cctataccct tcccccttac ccaactaggc tagttcatat tctagctcct ctgtatttgc tggacccccg g tc tctgcaa attcgctatt gttgcatgtt catgaggata catgcaagca cacatctgta cagctacggg aagttgtccc caaacaaata tatcagttcc ggcgagagaa tgtttcatat ttaagaacta accagccgtc taatgtgcac acaaatgtca gac tggacat tttataacta gcaatcatgg cctcactctc aaggcaacag t tttatagat cccccaccct tggggcatat cattttctgc tgttgttcta ccattgggag caggcactgg ggtggggcag cttcttccat cttttataat tgaaatgcag caagattaag ggaggatctg atcacagtag ctggccagcc attaccttca cacacgtgca taagcctggc cagttaccga t tt taaataa tcaacaacca cttgtccaca attctttacg tctttaagga tgacagcatt agcatttggg gaaagccagg atctgtgccc aatcatatag attttcttca gctcccctac aaagtttgct tacatatgca cctatggggt ccctgtgttc catatgaaat tctctggatg ggatatttta ccaagtgtgg tctcagattt aaatatgcat attgactccc gcgtatgatg tcccatgcac catatacacc cacatacaaa tcattgcttg atqgttcaga taacacttag ttgactatcc tgtgtgtgtg gcctgcagtt gcatatcctt tccaactttt caatcaat ta atgctgctgt tgatatcctt ccaataatga tttacatttc ccactcactc agaccaaggg gctagagaca tgcagacccc catcctatag agtatctgca gtccatcctt gtccctaatc 2 3/27 cctggggacc catcccagac tagagagtaa caggacccac aaatgggagg agcactaaat acggcatgtg act cagagat taacactaca agtggggcca attacaaaat tgtcggccac tctacacagc tttacttcat atttcctgga caaccttgta cctctgcctg ctgatcccta cactctcaag aagctacttg aaatgctatc ccacttcttg gcctctcttc ccagttctgg ttcagctctt atgactgtga tttggtggct tcat ctt agc tagggagaaa cgtctacctg agcagcatta ctaaagagta ttggctaaat acag ttc cca tgaatcaaga aaggcaagga tgtacttgta taaggacaat agcagtatta tgcctgtgac aaatttacta aaatgagtgt tatgaattta gtatttgaaa tttatcattc aaaataacta gaatgggggc gccctggcat tcttttccca gtctacagtt ccaaaagtcc gccctggctt ccaatgatgg ggttactggt gagtactttc gcatccactt gattatggga tccaaac t tt tgaagtatcc 5760 5820 5880 5940 6000 6060 6120 6180 6240 6300 6360 6420 6480 6540 6600 6660 6720 6780 6840 6900 6960 7020 7080 7140 7200 7260 7320 7380 7440 7500 WO 01/42304 WO 0142304PCTIUSOO/32940 acaagttgat cttccttctt gattttctta tgttttagaa gttgtatctt ggatattcta ggtttctggg ctaatatcca cttatcagtg agtacatatc aagtgaattc ttttgtgatt aggttacctc actcaagatg atattctcca ctatgttcat agcagcccta tttatagtag ccagaagctg gaaagaaccc agtccctcaa cagaggaatg gatacagaaa atgtggcaca tttatgcaat ggagtaccac tcagatatta aaaacaacga atttatgaaa ttctcgggca aaaccctatc taaagaccag gaataaggaa aagatggact gcctgcctgc agctgggaga gctggggaga cctttgtgga ttctgtaata cttaggggta cggaacagct tgtggctgga taattcigag ctccagcatg tctgcccccc aaaaaacatt ctgtttttct gaaagccttt ttcttctttg cctcagtgaa gaccagacac tcccaactgc agga atg tgc tcc ctt ccc atg gca aga tac tac atgtaagtaa tcttaacgat cgctcaatca aggggcctgg agatcacatg agaagggaaa aggctgagtc aaagggacaa agctccctct agccacagaa atctcaaaca ctgaataatt gatcttcatc tttgtcaatc acaacagccc tctttcctgg tgacagaatg gaacaactgt aagagtggta ttgcttagtc cattttacag acccggaaac tcaacctcca cgaggttata caattttcct catgtcatgc aattacccaa aagcagagag tgggatcgga ctctctgttc tctaaactga tgtagctagt tcttagaaag ctcaaacaat cttgagtccc aaggacagca cctttatggt cacctggatt gatacctata tcaaaaaaaa aaaaaggtct cactagatag ccctggctac cctgaaactc tcactgtgta catttaggtg accacgaact cacagagatc tgccttccaa gtgctgggat taaagtatgt accaccacac ctgcatcttt gacaataact gagtggtatc taaattcttc cagtggctaa acagttaagt cccagttccc aaagtctgag aaaaatgcca ggtggtgaaa tctgtacaga cctttgttct taatgtacaa gtgagcctgc tttaaaaaca atacgcaagc tgtttttgct attgctaagt gttgcagaga cagaaaaggc tcccagaagt ggtaactttg gtccagaggt tctgttctca aactcattgt gagctctgaa agcaactgat gggcagctct gaaatcagct gggcaattag gctaataaca ggcataattt taatgtttca cacgcatgac agttcctccc cagctgccct agtacatact taccctccta ggcacgtcat tagacccata ggtataacca gtgactaatc aggccctggt ctaattctaa gttggcctcc tatataagtg ccactcagag tgtacctcat catggctgta gtgggcccag aqtctaggga catagacttt tctattgtCC aatttctgat ttgtgaattt tctacaaaaa gaattttttt taattttaca aatcaaatca cagttactac atcttcagtt ccttcattaa ttagtgttac tatttaaaaa aataaaataa atcaagctca gaaacatcat ggatagggtt cattgtatct ccagggtacc tgagcttcaa agcaactcct cagacagcca tgaaaacatc ctcaattacc tcatgagaag acactattgt 24/27 7560 7620 7680 7740 7800 7860 7920 7980 8036 8084 8144 8204 8264 8324 8384 8444 8504 8564 8624 8684 8744 8804 8864 8924 8984 9044 9104 9164 9224 9284 9344 WO 01/42304 WO 0142304PCTIUSOO/32940 catttctgga gcctctgata atcctgagcc taggcagctt tgggatgaaa caatttctac 9404 ccttattgga acagtgtccc tctcctgtct ggaaaeaatt caccaaaggc tccatgtggt 9464 tgtccagtaa ggtggtatgg ggacagaaat ggacaatgat ecctgagggc agtgatceat 9524 taaccttgcc ctcctatttc aga atc aag gat gca cat caa aag gct ttg tac 9577 aca cgg aat ggc cag ctc ctg etg gga gac cct gat tea gac aat tat 9625 agt eca ggtgatcttc cggtggtggg ggtgggggag tggaggggag ggtgtggggg 9681 gggctctctt ccagaagttg cttagtgtce atctgceaea aggccttgat tCtttCettC 9741 aattgtgtct ctagagacat gagaatattg teaeagtgat aaggagaaga. ggtaggggca gtttcttect gtaaaaaatg aattccattt aceetgeagt etccatacag aaacaggcca gaggggggca gacccagtaa cttctagctg agcetacet tgcttaaaac ctgccatctg tggtcccctc actgtctgaa ttgcattctg tcttacctcc cagag aag gtc tgt ate ett cct aac ega gge eta gac cgc tee aag gtc ccc ate ttc etg ggg atg cag gga gga agt tge tgc etg geg tgt gta aag aca aga gag gga cct etc ctg cag etg gag gtgagacace cctccteatt gcagtcagta ctgceactgg aaeatagtga catctttgaa eecacatgte ccetctettg tttcecatct atctetcttt gcctecaget gagggaetct ageetttggg gatgtaeaga aagaacatgg etteggaaaa etcttceta ttgagtcctt etttggeeaa gcctetgagg cactaagggc tgacgtceea accaaacaet catttcatct eaeagetgtc tcecttteec cacag gat gtg aae ate gag gac eta tae aag gga ggt gaa caa ace ace cgt tte ace ttt ttc cag aga age ttg gga tet gee ttc agg ett gag get get gee tgc cet ggc tgg ttt etc tgt ggc eca get gag ce eag cag eca gtg eag etc ace aaa gag agt gaa ccc tee aec eat act gaa ttc tac ttt gag atg agt egg taa agagaeataa ggctggggec tcgtetagtg eeeeeagtct gagatettet tgeteagcat ctetggaaag eagaataagg aagataccaa agatgtttgg gtcttaatee ecagaatetg tgaeegtgtt acattaaatg geaaagggat ttttttttte ettcatggte catttgggee cattggaate atetgaggce tcatgaggag aaggaagagg tcagagggag actggggeaa actttggtac taaaagtaaz aatggagaca ggqaeeataa getgatgggt aacagtggtt tetagaaace ggaaatgatg agagctetcc tgacaeaggt tctggatttt tetggactga agaatggtga aataatacag ctecattatt ttaagecact gagtttgaga tcattcaatg aagctgtcat aataaaacct gtgctteaea 25/27 9801 9861 9921 9978 10026 10074 10122 10182 10242 10302 10360 10408 10456 10504 10552 10600 10660 10720 10780 10840 10900 10960 11020 WO 01/42304 tacaattcaa tggatgctga catcaggctc tttctatttc c tgat ttag t atggagcaca atatgatata ctaagcccag ggcagagaac ggcatggaga acgtctgtgg' gaatgacttg tgcaagcagg agctctgcat tatgggtagc tggtagagt t qctttaccaa aaacaagggg agtagtttct atc taag t tt aagaactcaa ttgcctccta aacataataa atttccatta aaggacttac gagccaaaci gctaagaaaa atgattggac tttgggtcat aagaaaaact gggatttgaa PCT/USOO/32940 tattggtagg ttccagggtc gactcttcta ataggaaatc gaggggtgag aaagatcttt agcagagcta cttcccaaag ataggtcaag gctccaggca ttaaggagtt caaaggagtt ggccctgtga cctcactcac ctgaccctgt tacttgagtg gggggcactg aagaaaaatt aattacttga cattttctta ga cccagaat t tgagatctg tgattgctgc qtgctggagg aatgtaatag gaggc tgagg acacattctg tttcaacttc ggcacatccc catatcctat actcaaaaat caccccggtg cagtggagtc aaagtccgtc tgtgtccctg ctgctggcac actgagaaga cagcacaaga ggacagctaa cattctaggt tgtctgggct gctgaaaatt tagcccataa tcattgctgg atccaggtac ttcatcttct attggtgaat attggagaat tgattgctta gtctctaatt ggctatgacc agcctctgtt tt cccatgga acagacaaaa gaaggctaca gagccctgac gcttagatta gggttagtta tggaacc cat ctaccatcac agacctctgg ggaagaagct atttcttgga cctgggttga gttgcacccc cccctgctaa ttttttgtgt tggccatgaa tgagccaatg ccctggactc agaatcagca atggtgtgtc aaaataggaa gtggggagct accagcctag cttcagaggt tgtataactt caggcagcac tggaatgagg aagtggaaag agaggttagt attctctgag aattatttta ccacccaggc tatttttttt acgac ca tga attataaagt gtggtaggtc gatgttttag gtggtggaag aagaactcac tagaattagc acttgtgacc 26/27 aagacatcta agagatttca ttgcctgaga agcagggagc caccagtgtc gctctggcta aggaaagcca aaatgaatag attcagaaag attcttgtgg aatgggtaga cagaggagtc gtgctacaga caatttctgt aggcacataa caaactacaa gtggttagaa tcccaactta tggcagtttc tcgcatgtta gcaatgatca acatcaggcc cagtatctgg aggcatggcc gggtcacctt actttccaga ttcatttgat agagaaccaa caaaataaat agaacgctgc gttcaagact gggattctcc caacccagaa gcattagcaa ctggaccgtc ttaagcagtg gacaccaaga ttcagggagg gggttttcct gtgtgagaga caagaatcca gtctaattgt atctaaggat gcctaccttc gctctggttc gcttagggac gttgttcagg tgcattcaga aatgttagtc tggttagtta gcaatgcagt ctcatttctg tcctagtacc tatttgctac cctigccttct gtttcaaact catgttcagt aagaagccca cttctgacca tagaaaaatc tgtggcact t ccagggaggc 11080 11140 11200 11260 11320 11380 11440 11500 11560 11620 11680 11740 11800 11860 11920 11980 12040 12100 12160 12220 12280 12340 12400 12460 12520 12580 12640 12700 12760 12820 12880 WO 01/42304 PTU0124 PCT/USOO/32940 tcctctgaca catcccacqa ctcaggctta aattccttct tctccctaga aggccacgcc 12940 atcttctcaa ccaggccaca gatgctataa ttatgtaaat gtgtgggaga ggcacacttt 13000 agatcttatc cactagt 13017 27/27

Claims (53)

1. An isolated nucleic acid molecule comprising a nucleotide sequence: as set forth in any of SEQ ID NO: 1, SEQ ID NO: 3, SEQ ID NO: 5, or SEQ ID NO: of the DNA insert in ATCC Deposit No. PTA-1423; encoding the polypeptide as set forth in any of SEQ ID NO: 2, SEQ ID NO: 4, SEQ ID NO: 6, or SEQ ID NO: 36; that hybridizes under highly stringent conditions to the complement of any or that is complementary to the nucleotide sequence of any of
2. An isolated nucleic acid molecule comprising: a nucleotide sequence encoding a polypeptide that is at least about percent identical to the polypeptide as set forth in any of SEQ ID NO: 2, SEQ ID NO: 4, SEQ ID NO: 6, or SEQ ID NO: 36, wherein the encoded polypeptide has an activity of 15 the polypeptide set forth in any of SEQ ID NO: 2, SEQ ID NO: 4, SEQ ID NO: 6, or SEQ ID NO: 36; a nucleotide sequence encoding an allelic variant or splice variant of the nucleotide sequence as set forth in any of SEQ ID NO: 1, SEQ ID NO: 3, SEQ ID NO: or SEQ ID NO: 35, the DNA insert in ATCC Deposit No. PTA-1423, or 20 a region of the nucleotide sequence as set forth in any of SEQ ID NO: 1, SEQ ID NO: 3, SEQ ID NO: 5, or SEQ ID NO: 35, the DNA insert in ATCC Deposit No. PTA-1423, or encoding a polypeptide fragment of at least about 25 amino S* acid residues, wherein the polypeptide fragment has an activity of the polypeptide as set forth in any of SEQ ID NO: 2, SEQ ID NO: 4, SEQ ID NO: 6, or SEQ ID NO: 36; i. DEC. 2004 14:59 WRAY AND ASSOCIATES NO. 970 P. 4 1 a nucleotide sequence that hybridizes under highly stringent conditions to the complement of any of a nucleotide sequence that is complementary to the nucleotide sequence of any of
3. An isolated nucleic acid molecule comprising: a nucleotide sequence encoding a polypeptide as set forth in any of SEQ ID NO: 2, SEQ ID NO: 4, SEQ ID NO: 6, or SEQ ID NO: 36 with at least one conservative amino acid substitution, wherein the encoded polypeptide has an activity of the polypeptide set forth in any of SEQ ID NO: 2, SEQ ID NO: 4, SEQ ID NO: 6, or SEQ ID NO: 36; a nucleotide sequence encoding a polypeptide as set forth in any of SEQ ID NO: 2, SEQ ID NO: 4, SEQ ID NO: 6; or SEQ ID NO: 36 which has a C- and/or N- terminal truncation, wherein the encoded polypeptide has an activity of the polypeptide set forth in any of SEQ ID NO: 2, SEQ ID NO: 4, SEQ ID NO: 6, or 15 SEQ ID NO: 36; S" a nucleotide sequence encoding a polypeptide as set forth in any of SEQ ID NO: 2, SEQ ID NO: 4, SEQ ID NO: 6. or SEQ ID NO: 36 with at least one modification selected from the group consisting of conservative amino acid substitutions, C-terminal truncation, and N-terminal truncation, wherein the 20 encoded polypeptide has an activity of the polypeptide set forth in any of SEQ ID NO: 2, SEQ ID NO: 4, SEQ ID NO: 6, or SEQ ID NO: 36; a nucleotide sequence that hybridizes under highly stringent conditions to the complement of any of or a nucleotide sequence that is complementary to the nucleotide sequence of any of
4. A vector comprising the nucleic acid molecule of any of claims 1, 2, or 3. A host cell comprising the vector of claim 4.
6. The host cell of claim 5 that is a eukaryotic cell.
7. The host cell of claim 5 that is a prokaryotic cell. 120 COMS ID No: SBMI-01021273 Received by IP Australia: Time 17:51 Date 2004-12-01 i DEC. 2004 14:59 WRAY AND ASSOCATES NO. 970 P.
8. A process of producing an IL-lra-R polypeptide comprising culturing the host cell of claim 5 under suitable conditions to express the polypeptide. and optionally isolating the polypeptide from the culture.
9. A polypeptide produced by the process of claim 8.
10. The process of Claim 8, wherein the nucleic acid molecule comprises promoter DNA other than the native IL-lra-R promoter DNA operatively linked to a nucleic acid molecule encoding an I L-1 ra-R polypeptide.
11. The isolated nucleic acid molecule according to claim 2, wherein the percent identity is determined using a computer program selected from the group consisting of GAP, BLASTIN, FASTAL. BLASTA, BLASTX, BestFit, and the Smith-Waterman algorithm.
12. A process for determining whether a compound inhibits IL-lra-R polypeptide activity of IL-lra-R polypeptide production comprising exposing a cell according to any of claims 5, 6, or 7 to the compound and measuring iL-i ra-R polypeptide activity or IL- I ra-R polypeptide production in said cell. 15 13. An isolated polypeptide comprising: .5121 COMS ID No: SBMI-01021273 Received by IP Australia: Time 17:51 Date 2004-12-01 the amino acid sequence as set forth in any of SEQ ID NO: 2, SEQ ID NO: 4, SEQ ID NO: 6, or SEQ ID NO: 36; or the amino acid sequence encoded by the DNA insert in ATCC Deposit No. PTA-1423.
14. An isolated polypeptide comprising: an amino acid sequence of an ortholog of any of SEQ ID NO: 2, SEQ ID NO: 4, SEQ ID NO: 6, or SEQ ID NO: 36; an amino acid sequence that is at least about 70 percent identical to the amino acid sequence of any of SEQ ID NO: 2, SEQ ID NO: 4, SEQ ID NO: 6, or SEQ ID NO: 36, wherein the polypeptide has an activity of the polypeptide set forth in any of SEQ ID NO: 2, SEQ ID NO: 4, SEQ ID NO: 6, or SEQ ID NO: 36; a fragment of the amino acid sequence set forth in any of SEQ ID NO: 2, SEQ ID NO: 4, SEQ ID NO: 6, or SEQ ID NO: 36 comprising at least about 25 amino acid residues, wherein the fragment has an activity of the polypeptide set forth in any of SEQ ID NO: 2, SEQ ID NO: 4, SEQ ID NO: 6, or SEQ ID NO: 36, or an amino acid sequence for an allelic variant or splice variant of the amino acid sequence as set forth in any of SEQ ID NO: 2, SEQ ID NO: 4, SEQ ID NO: 6, or SEQ ID NO: 36, the amino acid sequence encoded by the DNA insert in ATCC Deposit No. PTA-1423, or
15. An isolated polypeptide comprising: the amino acid sequence as set forth in any of SEQ ID NO: 2, SEQ ID NO: 4, SEQ ID NO: 6, or SEQ ID NO: 36 with at least one conservative amino acid substitution, wherein the polypeptide has an activity of the polypeptide set forth in any of C SEQ ID NO: 2, SEQ ID NO: 4, SEQ ID NO: 6, or SEQ ID NO: 36; 0* 0 the amino acid sequence as set forth in any of SEQ ID NO: 2, SEQ ID NO: 4, SEQ ID NO: 6, or SEQ ID NO: 36 which has a C- and/or N- terminal truncation, wherein the polypeptide has an activity of the polypeptide set forth in any of SEQ ID NO: 2, SEQ ID NO: 4, SEQ ID NO: 6, or SEQ ID NO: 36; or the amino acid sequence as set forth in any of SEQ ID NO: 2, SEQ ID NO: 4, SEQ ID NO: 6, or SEQ ID NO: 36 with at least one modification selected from the group consisting of conservative amino acid substitutions, C-terminal truncation, and N-terminal truncation, wherein the polypeptide has an activity of the polypeptide set forth in any of SEQ ID NO: 2, SEQ ID NO: 4, SEQ ID NO: 6, or SEQ ID NO: 36.
16. An isolated polypeptide encoded by the nucleic acid molecule of any of claims 1, 2, or 3, wherein the polypeptide has an activity of the polypeptides set forth in any of SEQ ID NO:2, SEQ ID NO: 4, SEQ ID NO:6, or SEQ ID NO: 36.
17. The isolated polypeptide according to claim 14, wherein the percent identity is determined using a computer program selected from the group consisting of GAP, BLASTP, FASTA, BLASTA, BLASTX, BestFit, and the Smith-Waterman algorithm.
18. An antibody or fragment thereof which specifically binds the polypeptide of any of Claims 13, 14, 15, 53 or 54.
19. The antibody or fragment thereof of claim 18 that specifically binds the 20 polypeptide comprising the amino acid sequence as set forth in any of SEQ ID NO: 2, SEQ ID NO: 4, SEQ ID NO: 6, or SEQ ID NO: 36, or a fragment thereof. The antibody or fragment thereof of claim 18 that is a humanized antibody. U
21. The antibody or fragment thereof of claim 18 that is a human antibody or fragment thereof.
22. The antibody or fragment thereof of claim 18 that is a polyclonal antibody or fragment thereof.
23. The antibody or fragment thereof of claim 18 that is a monoclonal antibody or fragment thereof.
24. The antibody or fragment thereof of claim 18 that is a chimeric antibody or fragment thereof. The antibody or fragment thereof of claim 18 that is a CDR-grafted antibody or fragment thereof.
26. The antibody or fragment thereof of claim 18 that is an antiidiotypic antibody or fragment thereof.
27. The antibody or fragment thereof of claim 18 that is a variable region fragment.
28. The variable region fragment of claim 27 that is a Fab or a Fab' fragment.
29. The antibody or fragment thereof of claim 18 that is bound to a detectable label.
30. The antibody or fragment thereof of claim 18 that antagonizes IL-lra-R polypeptide biological activity.
31. A method for treating, preventing, or ameliorating an IL-lra-R polypeptide-related disease, condition, or disorder comprising administering to a patient 20 an effective amount of a selective binding agent according to claim 18.
32. A selective binding agent produced by immunizing an animal with a polypeptide comprising an amino acid sequence of any of SEQ ID NO: 2, SEQ ID NO: 4, SEQ ID NO:6, or SEQ ID NO: 36. *o 124
33. A hybridoma which produces a selective binding agent which is capable of binding a polypeptide according to any of Claims 13, 14, 15, 53 or 54.
34. A method of detecting or quantitating the amount of IL- ra-R polypeptide using the anti-lL-lra-R antibody or fragment of claim 18.
35. A composition comprising the polypeptide of any of Claims 13, 14, 15, 53 or 54 and a pharmaceutically acceptable formulation agent.
36. The composition of claim 35, wherein the pharmaceutically acceptable formulation agent is a carrier, adjuvant, solubilizer, stabilizer, or anti-oxidant.
37. A polypeptide comprising a derivative of the polypeptide of any of Claims 13, 14, 15, 53 or 54.
38. The polypeptide of claim 37 that is covalently modified with a water- soluble polymer.
39. The polypeptide of claim 38, wherein the water-soluble polymer is selected from the group consisting of polyethylene glycol, monomethoxy-polyethylene glycol, dextran, cellulose, poly-(N-vinyl pyrrolidone) polyethylene glycol, propylene glycol homopolymers, polyproylene oxide/ethylene oxide co-polymers, polyoxyethylated polyols, and polyvinyl alcohol. 0 40. A composition comprising a nucleic acid molecule of any of claims 1, 2, or 3 and a pharmaceutically acceptable formulation agent.
41. The composition of claim 40, wherein said nucleic acid molecule is 0 contained in a viral vector. S42. A viral vector comprising a nucleic acid molecule of any of claims 1, 2, or *3. o
43. A fusion polypeptide comprising the polypeptide of any of Claims 13, 14, 53 or 54 fused to a heterologous amino acid sequence.
44. The fusion polypeptide of claim 45, wherein the heterologous amino acid sequence is an IgG constant domain or fragment thereof.
45. A method for treating, preventing, or ameliorating a medical condition comprising administering to a patient the polypeptide of any of Claims 13, 14, 15, 53 or 54, or the polypeptide encoded by the nucleic acid of any of Claims 1, 2, or 3.
46. A method of diagnosing an IL-lra-R polypeptide-related pathological condition or a susceptibility to a IL-lra-R polypeptide-related pathological condition in a subject comprising: determining the presence or amount of expression of the polypeptide of any of Claims 13, 14, 15, 53 or 54, or the polypeptide encoded by the nucleic acid molecule of any of Claims 1, 2, or 3 in a sample; and diagnosing a pathological condition or a susceptibility to a pathological condition based on the presence or amount of expression of the polypeptide.
47. A device, comprising: I a membrane suitable for implantation; and cells encapsulated within said membrane, wherein said cells secrete a ;protein of any of Claims 13, 14, 15, 53 or 54; and 20 said membrane is permeable to said protein and impermeable to materials detrimental to said cells. :48. A method of identifying a compound which binds to an IL-lra-R polypeptide comprising: *e 0 contacting the polypeptide of any of Claims 13, 14, 15, 53 or 54 with a compound; and determining the extent of binding of the IL-lra-R polypeptide to the compound.
49. The method of claim 48, further comprising determining the activity of the polypeptide when bound to the compound. A method of modulating levels of a polypeptide in an animal comprising administering to the animal the nucleic acid molecule of any of claims 1, 2, or 3.
51. A transgenic non-human mammal comprising the nucleic acid molecule. of any of claims 1, 2, or 3.
52. A process for determining whether a compound inhibits IL-lra-R polypeptide activity or IL-lra-R polypeptide production comprising exposing a transgenic mammal according to claim 51 to the compound, an measuring IL-lra-R polypeptide activity or IL-lra-R polypeptide production in said mammal.
53. An isolated polypeptide comprising the amino acid sequence as set forth in either SEQ ID NO: 2 or SEQ ID NO: 4, wherein the valine residue at position 29 may be substituted with a leucine residue; the alanine residue at position 34 may be substituted with a serine residue; the cysteine residue at position 38 may be substituted with a serine residue; 20 the alanine residue at position 39 may be substituted with a proline residue; the isoleucine residue at position 42 may be substituted with a valine residue; the threonine residue at position 53 may be substituted with a serine residue; 127 127 the isoleucine residue at position 61 may be substituted with a methionine residue; the glutamic acid residue at position 88 may be substituted with an aspartic acid residue; the glutamic acid residue at position 95 may be substituted with a glutamine residue; the alanine residue at either position 96 or 142 may be substituted with a threonine residue; the arginine residue at position 143 may be substituted with histidine residue; and the residues at any of positions 14, 16, 24, 33, 37, 44, 51, 66, 72, 74, 78, 104, 106, 117, 145, 150, or 152 may be substituted with any naturally occurring amino acid residue.
54. An isolated polypeptide comprising the amino acid sequence as set forth in either SEQ ID NO: 6, wherein the valine residue at position 48 may be substituted with a leucine residue; the alanine residue at position 53 may be substituted with a serine residue; the cysteine residue at position 57 may be substituted with a serine residue; the alanine residue at position 58 may be substituted with a proline residue; the isoleucine residue at position 61 may be substituted with a valine residue; 20 the threonine residue at position 72 may be substituted with a serine residue; the isoleucine residue at position 80 may be substituted with a methionine residue; the glutamic acid residue at position 107 may be substituted with an aspartic acid residue; the glutamic acid residue at position 114 may be substituted with a glutamine residue; the alanine residue at either position 115 or 161 may be substituted with a threonine residue; the arginine residue at position 162 may be substituted with histidine residue; and the residues at any of positions 33, 35, 43, 52, 56, 63, 70, 85, 91, 93, 97, 123, 125, 136, 164, 169, or 171 may be substituted with any naturally occurring amino acid residue. A nucleic acid molecule according to claim 1 substantially as hereinbefore described with reference to any one of the examples.
56. A vector according to claim 4 substantially as hereinbefore described with reference to any one of the examples.
57. A host cell according to claim 5 substantially as hereinbefore described with reference to any one of the examples. 0
58. A process according to claim 8 substantially as hereinbefore described with reference to any one of the examples.
59. A polypeptide according to any one of claims 9, 13 or 14 substantially as 20 hereinbefore described with reference to any one of the examples.
60. An antibody according to claim 18 substantially as hereinbefore described :with reference to Example 6. 0 4 Ao. S- 61. A selective binding agent according to claim 32 substantially as hereinbefore described with reference to Example 6. 1. DEC. 2004 14:59 W~RAY AND ASSOCIATES 'JO. 9706
62. A transgenic mammal according to claim 51 substantially as hereinbefore described with, reference to any one of Examples 7, 8 or 9.
63. Use of a region of the nucleotide sequence as set forth~ in any of SEQ ID NO: 1, SEQ ID NO: 3, SEQ ID NO: 5, SEQ ID NO: 35, the DNA insert in ATCC Deposit NO: PTA 1423 or any of claims 1(a) comprising a fragment of at least about 16 nucleotides; or a nucleotide sequence of any of claims 1 comprising a fragment of at least about 16 nucleotides; as a probe or primer for a nucleic acid molecule as set forth in any one of claims 1, 2(a) or 3(a) 3(c). Dated this FIRST day of DECEMBER 2004. Amgen Inc. Applicant Wray Associates Perth, Western Ausralia K Patent Attorneys for the Applicant 130 COMS tD No: SBMI-01021273 Received by IP Australia: Time 17.51 Date 2004-12-01
AU19450/01A 1999-12-10 2000-12-04 Interleukin-1 receptor antagonist-related molecules and uses thereof Ceased AU779534B2 (en)

Applications Claiming Priority (11)

Application Number Priority Date Filing Date Title
US17019199P 1999-12-10 1999-12-10
US60/170191 1999-12-10
US18805300P 2000-03-09 2000-03-09
US60/188053 2000-03-09
US19452100P 2000-04-04 2000-04-04
US60/194521 2000-04-04
US19591000P 2000-04-10 2000-04-10
US60/195910 2000-04-10
USNOTGIVEN 2000-06-23
US72458300A 2000-11-28 2000-11-28
PCT/US2000/032940 WO2001042304A1 (en) 1999-12-10 2000-12-04 Interleukin-1 receptor antagonist-related molecules and uses thereof

Publications (2)

Publication Number Publication Date
AU1945001A AU1945001A (en) 2001-06-18
AU779534B2 true AU779534B2 (en) 2005-01-27

Family

ID=27538813

Family Applications (1)

Application Number Title Priority Date Filing Date
AU19450/01A Ceased AU779534B2 (en) 1999-12-10 2000-12-04 Interleukin-1 receptor antagonist-related molecules and uses thereof

Country Status (6)

Country Link
EP (1) EP1246844A1 (en)
JP (1) JP3842647B2 (en)
AR (1) AR026768A1 (en)
AU (1) AU779534B2 (en)
MX (1) MXPA02005775A (en)
TW (1) TWI283683B (en)

Families Citing this family (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP3551650A4 (en) * 2016-12-07 2020-07-08 The University of Florida Research Foundation Incorporated IL-1RA CDNAs
CN113940308A (en) * 2021-04-28 2022-01-18 重庆腾达牧业有限公司 Dazu black goat breeding house feeding management method

Also Published As

Publication number Publication date
EP1246844A1 (en) 2002-10-09
AU1945001A (en) 2001-06-18
MXPA02005775A (en) 2003-10-14
TWI283683B (en) 2007-07-11
JP3842647B2 (en) 2006-11-08
JP2003518371A (en) 2003-06-10
AR026768A1 (en) 2003-02-26

Similar Documents

Publication Publication Date Title
AU2001272968C1 (en) Il-17 molecules and uses thereof
US8148109B2 (en) Interferon-like proteins and uses thereof
AU2001259029B2 (en) Il-17 receptor like molecules and uses thereof
US20030166069A1 (en) Interleukin-1 receptor antagonist-like molecules and uses thereof
US20020045213A1 (en) IL-17 receptor like molecules and uses thereof
US20030004106A1 (en) Interleukin-1 receptor antagonist-related molecules and uses thereof
AU778676B2 (en) Interleukin-1 receptor antagonist-like molecules and uses thereof
AU782554B2 (en) Interleukin-1 receptor antagonist-like molecules and uses thereof
AU779534B2 (en) Interleukin-1 receptor antagonist-related molecules and uses thereof
CA2393661A1 (en) Interleukin-1 receptor antagonist-like molecules and uses thereof
AU2007202616A1 (en) IL-17 Like Molecules and Uses Thereof
AU1944901A (en) Interferon-like molecules and uses thereof
US20030099980A1 (en) IL-17 receptor like molecules and uses thereof
AU2005201491A1 (en) Interferon-Like Molecules and Uses Thereof
AU2007200885A1 (en) IL-17 Receptor Like Molecules and Uses Thereof