WO1998047921A9 - Mammalian cytokines; related reagents and methods - Google Patents
Mammalian cytokines; related reagents and methodsInfo
- Publication number
- WO1998047921A9 WO1998047921A9 PCT/US1998/006879 US9806879W WO9847921A9 WO 1998047921 A9 WO1998047921 A9 WO 1998047921A9 US 9806879 W US9806879 W US 9806879W WO 9847921 A9 WO9847921 A9 WO 9847921A9
- Authority
- WO
- WIPO (PCT)
- Prior art keywords
- protein
- seq
- leu
- polypeptide
- antibody
- Prior art date
Links
Classifications
-
- C—CHEMISTRY; METALLURGY
- C07—ORGANIC CHEMISTRY
- C07K—PEPTIDES
- C07K14/00—Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
- C07K14/435—Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
- C07K14/52—Cytokines; Lymphokines; Interferons
- C07K14/54—Interleukins [IL]
- C07K14/545—IL-1
-
- C—CHEMISTRY; METALLURGY
- C07—ORGANIC CHEMISTRY
- C07K—PEPTIDES
- C07K14/00—Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
- C07K14/435—Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
- C07K14/52—Cytokines; Lymphokines; Interferons
- C07K14/54—Interleukins [IL]
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K38/00—Medicinal preparations containing peptides
-
- C—CHEMISTRY; METALLURGY
- C07—ORGANIC CHEMISTRY
- C07K—PEPTIDES
- C07K2319/00—Fusion polypeptide
Definitions
- the present invention relates to compositions and methods for affecting mammalian physiology, including morphogenesis or immune system function.
- it provides nucleic acids, proteins, and antibodies which regulate development and/or the immune system. Diagnostic and therapeutic uses of these materials are also disclosed.
- Recombinant DNA technology refers generally to techniques of integrating genetic information from a donor source into vectors for subsequent processing, such as through introduction into a host, whereby the transferred genetic information is copied and/or expressed in the new environment.
- the genetic information exists in the form of complementary DNA (cDNA) derived from messenger RNA (mRNA) coding for a desired protein product.
- cDNA complementary DNA
- mRNA messenger RNA
- the carrier is frecjuently a plasmid having the capacity to incorporate cDNA for later replication in a host and, in some cases, actually to control expression of the cDNA and thereby direct synthesis of the encoded product in the host.
- Lymphokines apparently mediate cellular activities in a variety of ways . They have been shown to support the proliferation, growth, and/or differentiation of pluripotent hematopoietic stem cells into vast numbers of progenitors comprising diverse cellular lineages which make up a complex immune system. Proper and balanced interactions between the cellular components are necessary for a healthy immune response. The different cellular lineages often respond in a different manner when lymphokines are administered in conjunction with other agents .
- B-cells which can produce and secrete immunoglobulins (proteins with the capability of recognizing and binding to foreign matter to effect its removal)
- T-cells of various subsets that secrete lymphokines and induce or suppress the B-cells and various other cells (including other T- cells) making up the immune network.
- lymphocytes interact with many other cell types .
- mast cell which has not been positively identified in all mammalian species
- mast cell is a granule-containing connective tissue cell located proximal to capillaries throughout the body. These cells are found in especially high concentrations in the lungs, skin, and gastrointestinal and genitourinary tracts .
- Mast cells play a central role in allergy-related disorders, particularly anaphylaxis as follows: when selected antigens crosslink one class of immunoglobulins bound to receptors on the mast cell surface, the mast cell degranulates and releases mediators, e.g., histamine, serotonin, heparin, and prostaglandins, which cause allergic reactions, e.g., anaphylaxis.
- the interleukin-1 family of proteins includes the IL-l ⁇ , the IL-l ⁇ , the IL-lRA, and recently the IL-l ⁇
- IL-1 receptors various growth and regulatory factors exist which modulate morphogenetic development. This includes, e.g., the Toll ligands, which signal through binding to receptors which share structural, and mechanistic, features characteristic of the IL-1 receptors. See, e.g., Lemaitre, et al . (1996) Cell 86:973-983; and Belvin and Anderson (1996) Ann. Rev. Cell & Develop. Biol. 12:393-416.
- the present invention is based on the discovery, purification, and characterization of the biological activities of two novel mammalian, e.g., rodent, interleukin-1 like molecules, designated interleukin-l ⁇ (IL-l ⁇ ) and interleukin-l ⁇ (IL-l ⁇ ) .
- IL-l ⁇ interleukin-l ⁇
- IL-l ⁇ interleukin-l ⁇
- IL-l ⁇ interleukin-l ⁇
- IL-l ⁇ interleukin-l ⁇
- IL-l ⁇ interleukin-l ⁇
- nucleic acids of the invention are characterized, in part, by their homology to cloned complementary DNA (cDNA) sequences enclosed herein, and/or by functional assays for IL-l ⁇ or IL-l ⁇ activity applied to the polypeptides, which are typically encoded by these nucleic acids.
- Methods for modulating or intervening in the control of an immune response are also provided by the use of IL-l ⁇ or IL-l ⁇ either singly or in combination with other molecules .
- IL-l ⁇ or IL-l ⁇ gene products should be similar to, and may well share receptors or portions of the signaling pathways used by known IL-1 family members.
- Equivalent vectors may be constructed by using polymerase chain reaction (PCR) techniques and sequences of the inserts .
- the present invention provides isolated or recombinant IL-l ⁇ or IL-l ⁇ polypeptides that specifically bind polyclonal antibodies generated against a 12 consecutive amino residues of a defined amino acid segment (i.e., SEQ ID NO: 2 or 6). These IL-l ⁇ or IL-l ⁇ polypeptides are further defined by comprising a select sequence of additional defined amino acids. Further, in another embodiment, fusion proteins comprising IL-l ⁇ or IL-l ⁇ polypeptides are provided. In still another aspect, there are provided variants, including fragments, natural alleles, labels, and modifications of the IL-l ⁇ or IL-l ⁇ polypeptides. Also provided are the nucleic acids encoding such fragments, variants or modified polypeptides .
- polypeptide embodiments include an isolated or recombinant polypeptide that: A) specifically binds polyclonal antibodies generated against a 12 consecutive amino acid segment of SEQ ID NO: 2; and comprises at least one sequence selected from (see SEQ ID NO: 2) :
- Preferred embodiments include such a polypeptide: wherein the polypeptide comprises a plurality of the described sequences.
- the 12 consecutive amino acid segment comes from an IL-l ⁇ sequence:
- ValGln SerProVallleLeuGlyValGlnGlyGlySerGlnCys ;
- ThrSerSerPheGluSerAlaAlaTyrProGlyTrpPhe or an IL-l ⁇ sequence : ArgAlaAlaSerProSerLeuArgHisValGlnAspLeu; SerSerArgValTrpIleLeuGlnAsnAsnlleLeu; ProValThrlleThrLeuLeuProCysGlnTyrLeu; GlyValGlnArgProMetSerCysLeuPheCysThr; PheCysThrLysAspGlyGluGlnProValLeuGlnLeu; ThrSerThrPheGluSerAlaAlaPheProGlyTrpPhe; or
- the: polypeptide comprises a mature protein; lacks a post-translational modification; is from a rodent, including a mouse; is a natural allelic variant of IL-l ⁇ or IL-l ⁇ ; has a length at least about 30 amino acids; exhibits at least two non-overlapping epitopes that are specific for a rodent IL-l ⁇ ; exhibits a sequence identity at least about 90% over a length of at least about 20 amino acids to SEQ ID NO: 2; exhibits at least two non-overlapping epitopes which are specific for a rodent IL-l ⁇ ; exhibits a sequence identity at least about 90% over a length of at least about 20 amino acids to SEQ ID NO: 6; is glycosylated; has a molecular weight of at least 10 kD with natural glycosylation; is a synthetic polypeptide; is attached to a solid substrate; is conjugated
- a soluble polypeptide comprising: a sterile polypeptide; the sterile polypeptide and a carrier, wherein the carrier is: an aqueous compound, including water, saline, and/or buffer; and/or formulated for oral, rectal, nasal, topical, or parenteral administration.
- a fusion protein embodiment includes one having a polypeptide sequence as described, further comprising: a mature protein; a detection or purification tag, including a FLAG, His6, or Ig sequence; or sequence of another cytokine or chemokine .
- Kit embodiments includes those comprising a protein or polypeptide as described, and: a compartment comprising the protein or polypeptide; and/or instructions for use or disposal of reagents in the kit.
- Other embodiments include pharmaceutical compositions comprising a sterile IL-l ⁇ or IL-l ⁇ protein or peptide with a suitable carrier for use in various administrations .
- the invention also provides a binding compound comprising an antigen binding site from an antibody, which specifically binds to IL-l ⁇ or IL-l ⁇ protein or polypeptide sequence.
- Various preferred binding compounds comprise an antigen binding site from an antibody, which specifically binds to a mature protein of a polypeptide, as described, wherein: the mature protein is an IL-l ⁇ or IL-l ⁇ protein; the binding compound is an
- the binding compound is conjugated to another chemical moiety; or the antibody: is raised against a 12 consecutive amino acid segment of SEQ ID NO: 2 or 6; is raised against a mature IL-l ⁇ or IL-l ⁇ protein; is raised to a purified rodent IL-l ⁇ or IL- l ⁇ ; is immunoselected; is a polyclonal antibody; binds to a denatured IL-l ⁇ or IL-l ⁇ ; exhibits a Kd to antigen of at least 30
- binding compounds include those comprising an antigen binding portion from an antibody, which specifically binds to: a rodent protein , as described, wherein: the protein is a murine protein; the binding compound is an Fv, Fab, or Fab2 fragment; the binding compound is conjugated to another chemical moiety; or the antibody: is raised against a peptide sequence of a mature polypeptide comprising a 12 consecutive amino acid segment of SEQ ID NO: 2 or SEQ ID NO: 6; is raised against a mature rodent IL-l ⁇ or IL-l ⁇ ; is raised to a purified rodent IL-l ⁇ or IL-l ⁇ ; is immunoselected; is a polyclonal antibody; binds to a denatured rodent IL-l ⁇ or IL-l ⁇ ; exhibits a Kd to antigen of at least 30 ⁇ M; is attached to a solid substrate, including a bead or plastic membrane; is in a sterile composition; or is detectably labeled, including a radioactive or fluorescent
- binding compounds e.g., methods of: making an antibody, as described, comprising immunizing an immune system with an immunogenic -amount of: a rodent IL-l ⁇ polypeptide; a peptide sequence comprising a 12 consecutive amino acid segment of SEQ ID NO: 2; a rodent IL-l ⁇ polypeptide; a peptide sequence comprising a 12 consecutive amino acid segment of SEQ ID NO: 6; thereby causing the antibody to be produced; or producing an antigen: antibody complex, comprising contacting: a rodent IL-l ⁇ protein or peptide with an antibody, as described, or a rodent IL-l ⁇ protein or peptide with an antibody, as described, thereby allowing the complex to form.
- Kits comprising the binding compound, as described, and: a compartment comprising the binding compound; and/or instructions for use or disposal of reagents in the kit.
- Other forms of the compositions include those comprising: a sterile binding compound, as described, or the binding compound and a carrier, wherein the carrier is: an aqueous compound, including water, saline, and/or buffer; and/or formulated for oral, rectal, nasal, topical, or parenteral administration.
- the kit comprises the described binding compound and: a compartment comprising that binding compound; and/or instructions for use or disposal of reagents in the kit .
- the kit may also be capable of making a qualitative or quantitative analysis.
- compositions include: a sterile binding compound described above, or the binding compound and a carrier, wherein the carrier is: an aqueous compound, including water, saline, and/or buffer; and/or formulated for oral, rectal, nasal, topical, or parenteral administration.
- a sterile binding compound described above or the binding compound and a carrier, wherein the carrier is: an aqueous compound, including water, saline, and/or buffer; and/or formulated for oral, rectal, nasal, topical, or parenteral administration.
- Nucleic acid embodiments include an isolated or recombinant nucleic acid encoding a protein or peptide or fusion protein, as described, wherein: the IL-l ⁇ or IL-l ⁇ is from a mammal; the nucleic acid: encodes an antigenic peptide sequence of SEQ ID NO: 2, 4, or 6; encodes a plurality of antigenic peptide sequences of SEQ ID NO: 2, 4, or 6; exhibits at least about 80% identity to a natural cDNA encoding said segment; is an expression vector; further comprises an origin of replication; is from a natural source; comprises a detectable label; comprises synthetic nucleotide sequence; is less than 6 kb, preferably less than 3 kb; is from a mammal, including a rodent; comprises a natural full length coding sequence; is a hybridization probe for a gene encoding said IL-l ⁇ or IL-l ⁇ ; or is a primer, PCR product, or mutagenesis primer.
- the invention further embraces an isolated or recombinant nucleic acid encoding a protein or peptide or fusion protein, as described, wherein: the protein, peptide, or fusion protein is IL-l ⁇ or IL-l ⁇ from a rodent; or the nucleic acid: encodes an antigenic peptide sequence of SEQ ID NO: 2 or SEQ ID NO: 6; encodes a plurality of distinct antigenic peptide sequences of SEQ ID NO: 2 or 6; exhibits at least about 80% identity to a natural cDNA encoding the segment; is an expression vector; further comprises an origin of replication; is from a natural source; comprises a detectable label; comprises synthetic nucleotide sequence; is less than 6 kb, preferably less than 3 kb; is from a rodent; comprises a natural full length coding sequence; is a hybridization probe for a gene encoding the IL-l ⁇ or IL- l ⁇ ; or is a PCR primer, PCR product, or
- the cell is: a prokaryotic cell; a eukaryotic cell; a bacterial cell; a yeast cell; an insect cell; a mammalian cell; a mouse cell; a primate cell; or a human cell.
- kits include the described nucleic acid and: a compartment comprising the nucleic acid; a compartment comprising an IL-l ⁇ or IL-l ⁇ protein or polypeptide; and/or instructions for use or disposal of reagents in the kit.
- the kit is capable of making a qualitative or quantitative analysis.
- the invention provides a nucleic acid which: hybridizes under wash conditions of 30° C and less than 2M salt to SEQ ID NO: 1; hybridizes under wash conditions of 30° C and less than 2 M salt to SEQ ID NO: 3 or 5; exhibits at least about 85% identity over a stretch of at least about 30 nucleotides to a rodent IL- l ⁇ ; or exhibits at least about 85% identity over a stretch of at least about 30 nucleotides to a rodent IL-l ⁇ .
- the nucleic acid described will hybridize when wash conditions are at 45° C and/or 500 mM salt; or exhibits identity at least 90% and/or over a stretch of at least 55 nucleotides. More preferably, the nucleic acid above will: hybridize at wash conditions of 55° C and/or 150 mM salt; or exhibit an identity of at least 95% and/or over a stretch of at least 75 nucleotides.
- the invention also provides methods of making or using these compositions or compounds. Such include a method of modulating physiology or development of a cell or tissue culture cells comprising contacting said cell or cells with an agonist or antagonist of a mammalian IL- l ⁇ or IL-l ⁇ .
- the contacting is in combination with an agonist or antagonist of IL-l ⁇ , IL-1RA, IL-l ⁇ , IL-l ⁇ , IL-2, and/or IL-12; the contacting is with an antagonist, including binding composition comprising an antibody binding site which specifically binds an IL-l ⁇ or IL-l ⁇ ; or the modulating is regulation of IFN- ⁇ production.
- Figure IA is a cartoon depicting a top down view through the central axis of the predicted IL-l ⁇ or IL-l ⁇ protein demonstrating the characteristic tertiary ⁇ - trefoil structure with its 3-fold symmetric topology.
- Contact sites of the IL-l ⁇ or IL-l ⁇ protein that are predicted to bind the IL-1 receptor subunits are designated as sites A, B or C (Table 2) .
- Contact sites A and C bind to the first receptor subunit of IL-1 while contact site B binds to the IL-1 second receptor subunit.
- Figure IB is a cartoon depicting a side view of the predicted IL-l ⁇ and IL-l ⁇ protein demonstrating the barrel structure formed by the twelve ⁇ domains and a mushroomlike cap.
- the present invention provides the amino acid sequence and DNA sequence of mammalian, e.g., rodent, interleukin-1 like molecules having particular defined properties, both structural and biological. These have been designated herein as interleukin-l ⁇ (IL-l ⁇ ) and interleukin-l ⁇ (IL-l ⁇ ) , respectively, and increase the number of members of the IL-1 family from 4 to 6.
- IL-l ⁇ interleukin-l ⁇
- IL-l ⁇ interleukin-l ⁇
- IL-l ⁇ interleukin-l ⁇
- nucleic acids encompassed herein include DNA, cDNA, and RNA sequences which encode IL-l ⁇ and IL-l ⁇ . It is understood that nucleic acids encoding all or a portion of IL-l ⁇ and IL- l ⁇ polypeptides are also encompassed, so long as they encode a polypeptide with IL-l ⁇ or IL-l ⁇ activity. Such nucleic acids include both naturally occurring and intentionally manipulated nucleic acids. For example, IL-l ⁇ or IL-l ⁇ may be subjected to site-directed mutagenesis .
- a complete nucleotide and corresponding amino acid sequence of a mammalian (rodent) IL-l ⁇ coding segment is shown in SEQ ID NO: 1 and SEQ ID NO: 2, respectively.
- the coding sequence does not indicate a signal sequence, which has been reported for various forms of messages encoding other members of the IL-1 family.
- Another form of the message probably encodes a signal sequence much like the IL-l ⁇ prodomain which is cleaved by a convertase-like enzyme, see Dinarello (1994) FASEB J. 1314-1325) .
- SEQ ID NO: 3-6 show a partial nucleotide (SEQ ID NO: 3) and corresponding amino acid sequence (SEQ ID NO: 4) , as well as a full length nucleic acid (SEQ ID NO: 5) and corresponding amino acid sequence (SEQ ID NO: 6) of a mammalian (rodent) IL-l ⁇ coding segment.
- Table 1 shows ⁇ conformation boundaries for IL-l ⁇ and IL-l ⁇ .
- the presence of amino acid residues between ⁇ conformations ⁇ 4 and ⁇ 5 are characteristic of IL-1 agonists.
- IL-1 family molecules have highly conserved residues in the region encompassing ⁇ conformations ⁇ 9 and ⁇ lO.
- Table 2 shows relationship of IL-1 family members, and Table 3 provides an alignment of selected members.
- IL-l ⁇ shall be used to describe a protein comprising a protein or peptide segment having or sharing the amino acid sequence shown in SEQ ID NO: 2 or a substantial fragment thereof.
- the invention also includes protein variations of the IL-l ⁇ allele whose sequence is provided, e.g., a mutein agonist or antagonist. Typically, such agonists or antagonists will exhibit less than about 10% sequence differences, and thus will often have between 1- and 11-fold substitutions, e.g., 2-, 3-, 5-, 7-fold, and others. It also encompasses allelic and other variants, e.g., natural polymorphic variants, of the protein described.
- Natural as used herein means unmodified by artifice. Typically, it will bind to its corresponding biological receptor with high affinity, e.g., at least about 100 nM, usually better than about 30 nM, preferably better than about 10 nM, and more preferably at better than about 3 nM.
- the term shall also be used herein to refer to related naturally occurring forms, e.g., alleles, polymorphic variants, and metabolic variants of the mammalian protein.
- This invention also encompasses proteins or peptides having substantial amino acid sequence homology with the amino acid sequence in SEQ ID NO: 2, 4 or 6. It will include sequence variants with relatively few substitutions, e.g., preferably less than about 3-5.
- a substantial polypeptide "fragment”, or “segment” is a stretch of amino acid residues of at least about ⁇ amino acids, generally at least 10 amino acids, more generally at least 12 amino acids, often at least 14 amino acids, more often at least 16 amino acids, typically at least l ⁇ amino acids, more typically at least 20 amino acids, usually at least 22 amino acids, more usually at least 24 amino acids, preferably at least 26 amino acids, more preferably at least 2 ⁇ amino acids, and, in particularly preferred embodiments, at least about 30 or more amino acids. Sequences of segments of different proteins can be compared to one another over appropriate length stretches.
- Amino acid sequence homolo-gy, or sequence identity is determined by optimizing residue matches, if necessary, by introducing gaps as required. See, e.g., Needleham, et al . , (1970) J. Mol. Biol. 46:443-453: Sankoff, et al . , (1983) chapter one in Time Warps , String Edits, and Macromolecules : The Theory and Practice of Sequence Comparison, Addiso -Wesley, Reading, MA; and software packages from IntelliGenetics, Mountain View, CA; and the University of Wisconsin Genetics Computer Group (GCG) , Madison, WI; each of which is incorporated herein by reference. This changes when considering conservative substitutions as matches .
- Conservative substitutions typically include substitutions within the following groups: glycine, alanine; valine, isoleucine, leucine; aspartic acid, glutamic acid; asparagine, glutamine; serine, threonine; lysine, arginine; and phenylalanine, tyrosine.
- Homologous amino acid sequences are intended to include natural allelic and interspecies variations in the cytokine sequence. Typical homologous proteins or peptides will have from 50-100% homology (if gaps can be introduced) , to 60-100% homology (if conservative substitutions are included) with an amino acid sequence segments of Tables 1 or 2.
- Homology measures will be at least about 70%, generally at least 76%, more generally at least 81%, often at least 85%, more often at least 88%, typically at least 90%, more typically at least 92%, usually at least 94%, more usually at least 95%, preferably at least 96%, and more preferably at least 97%, and in particularly preferred embodiments, at least 98% or more.
- the degree of homology will vary with the length of the compared segments .
- Homologous proteins or peptides such as the allelic variants, will share most biological activities with the embodiments described in Tables 1 and/or 2.
- the term "biological activity" is used to describe, without limitation, effects on inflammatory responses and/or innate immunity.
- IL-l ⁇ may, like IL-l ⁇ , exhibit synergistic induction by splenocytes of IFN- ⁇ in combination with IL-12 or IL-2, with or without anti-type I or anti-type II IL-1 receptor antibodies, or more structural properties as receptor binding and cross-reactivity with antibodies raised against the same or a polymorphic variant of a mammalian IL-l ⁇ or IL-l ⁇ .
- ligand, agonist, antagonist, and analog of, e.g., IL-l ⁇ include molecules that modulate the characteristic cellular responses to IL-l ⁇ or IL-l ⁇ -like proteins, as well as molecules possessing the more standard structural binding competition features of ligand-receptor interactions, e.g., where the receptor is a natural receptor or an antibody.
- the cellular responses likely are mediated through binding of IL-l ⁇ or IL-l ⁇ to cellular receptors related to, but possibly distinct from, the type I or type II IL-1 receptors.
- a ligand is a molecule which serves either as a natural ligand to which said receptor, or an analog thereof, binds, or a molecule which is a functional analog of the natural ligand.
- the functional analog may be a ligand with structural modifications, or may be a wholly unrelated molecule which has a molecular shape which interacts with the appropriate ligand binding determinants.
- the ligands may serve as agonists or antagonists, see, e.g., Goodman, et al . (eds.) (1990) Goodman & Gilman's: The Pharmacological Bases of Therapeutics , Pergamon Press, New York.
- Rational drug design may also be based upon structural studies of the molecular shapes of a receptor or antibody and other effectors or ligands. Effectors may be other proteins which mediate other functions in response to ligand binding, or other proteins which normally interact with the receptor.
- One means for determining which sites interact with specific other proteins is a physical structure determination, e.g., x- ray crystallography or 2 dimensional NMR techniques . These will provide guidance as to which amino acid residues form molecular contact regions.
- x- ray crystallography or 2 dimensional NMR techniques .
- the IL-l ⁇ or IL-l ⁇ proteins will have a number of different biological activities, e.g., in the immune system, and will include inflammatory functions or other innate immunity responses.
- the IL-l ⁇ or IL-l ⁇ proteins are homologous to other IL-1 proteins, but each have structural differences.
- a human IL-l ⁇ gene coding sequence probably has about 70% identity with the nucleotide coding sequence of mouse IL-l ⁇ , and similar measures of similarity will apply to the IL-l ⁇ and IL-l ⁇ . At the amino acid level, there is also likely to be about 60% identity.
- the mouse IL-l ⁇ molecule has the ability to stimulate IFN- ⁇ production which augments NK activity in spleen cells. See Okamura, et al . (1995) Nature 378 : ⁇ -
- the activities of the mouse IL-l ⁇ , IL-l ⁇ , and IL-l ⁇ have been compared as to their ability to induce IFN- ⁇ , alone or in combination with IL-2 or IL-12 in SCID splenocytes and purified NK cells. See Hunter, et al . (1995) J. Immunol. 155:4347-4354; and Bancroft, et al . (1991) Immunol. Revs. 124:5-xxx.
- the IL-l ⁇ was found to be much more potent in stimulating IFN-l ⁇ than either IL- l ⁇ or IL-l ⁇ .
- IL-l ⁇ and IL-l ⁇ and their agonists or antagonists should have related activities, typically affecting similar immune functions, including inflammatory responses.
- IFN- ⁇ production is blocked by the addition of anti-IL-l ⁇ antibodies. See Hunter, et al . (1995) .
- mouse IL-l ⁇ can overcome this block and induce IFN- ⁇ . This is the only cytokine known to be able to do this.
- administration of mouse IL-l ⁇ to mice infected with the parasite T. Cruzi significantly decreases parasitemia.
- IL-l ⁇ and IL-l ⁇ and their agonists or antagonists should operate through related mechanisms and effectors.
- the present disclosure also describes new assays for activities predicted for the mouse IL-l ⁇ or IL-l ⁇ molecules . Corresponding activities should be found in other mammalian systems, including primates. It is likely that the new mouse IL-1-like molecules produced by similar recombinant means to the human IL-l ⁇ protein should exhibit a biological activity of modulating lymphocyte cells in production of IFN- ⁇ . See assays described, e.g., in de Waal Malefyt, et al . , in de Vries and de Waal Malefyt (eds. 1995) "Interleukin-10 " Austin Co., Austin, TX. Furthermore, there is substantial likelihood of synergy with other IL-1 or IL-12 related agonists or antagonists . It is likely that the receptors, which are expected to include multiple different polypeptide chains, exhibit species specificity for their corresponding ligands. The IL-l ⁇ and IL-l ⁇ ligands both signal through heterodimeric receptors.
- isolated nucleic acid or fragments e.g., which encode this or a closely related protein, or fragments thereof, e.g., to encode a biologically active corresponding polypeptide.
- isolated nucleic acid or fragments as used herein means a nucleic acid, e.g., a DNA or RNA molecule, that is not immediately contiguous when present in the naturally occurring genome of the organism from which it is derived.
- the term describes, for example, a nucleic acid that is incorporated into a vector, such as a plasmid or viral vector; a nucleic acid that is incorporated into the genome of a heterologous cell (or the genome of homologous cell, but at a site different from that at which it normally occurs) ; and a nucleic acid that exists as a separate molecule, e.g., a DNA fragment produced by PCR amplification or restriction enzyme digestion, or an RNA molecule produced by in vitro transcription.
- a vector such as a plasmid or viral vector
- a nucleic acid that is incorporated into the genome of a heterologous cell or the genome of homologous cell, but at a site different from that at which it normally occurs
- a nucleic acid that exists as a separate molecule e.g., a DNA fragment produced by PCR amplification or restriction enzyme digestion, or an RNA molecule produced by in vitro transcription.
- the term also describes a recombinant (i.e., genetically engineered) nucleic acid that forms part of a hybrid gene encoding additional polypeptide sequences that can be used, for example, in the production of a fusion protein.
- this invention embodies any engineered or nucleic acid molecule created by artifice that encodes a biologically active protein or polypeptide having characteristic IL-l ⁇ or IL-l ⁇ activity.
- the nucleic acid is capable of hybridizing, under appropriate conditions, with a nucleic acid sequence segment shown in SEQ ID NO: 1, 3 or 5.
- Said biologically active protein or polypeptide can be a full length protein, or fragment, and will typically have a segment of amino acid sequence highly homologous to one shown in SEQ ID NO: 2, 4 or 6.
- this invention covers the use of isolated or recombinant nucleic acid, or fragments thereof, which encode proteins having fragments which are homologous to the newly disclosed IL- 1-like proteins.
- the isolated nucleic acids can have the respective regulatory sequences in the 5' and 3' flanks, e.g. , promoters, enhancers, poly-A addition signals, and others from the natural gene.
- an "isolated" nucleic acid is a nucleic acid, e.g., an RNA, DNA, or a mixed polymer, which is substantially pure, e.g., separated from other components which naturally accompany a native sequence, such as ribosomes, polymerases, and flanking genomic sequences from the originating species.
- the term embraces a nucleic acid se-quence which has been removed from its naturally occurring environment, and includes recombinant or cloned DNA isolates, which are thereby distinguishable from naturally occurring compositions, and chemically synthesized analogs or analogs biologically synthesized by heterologous systems.
- a substantially pure molecule includes isolated forms of the molecule, either completely or substantially pure.
- An isolated nucleic acid will generally be a homogeneous composition of molecules, but will, in some embodiments, contain heterogeneity, preferably minor. This heterogeneity is typically found at the polymer ends or portions not critical to a desired biological function or activity.
- a "recombinant" nucleic acid is defined either by its method of production or its structure.
- the process is use of recombinant nucleic acid techniques, e.g., involving human intervention in the nucleotide sequence.
- this intervention involves in vitro manipulation, although under certain circumstances it may involve more classical animal breeding techniques.
- it can be a nucleic acid made by generating a sequence comprising fusion of two fragments which are not naturally contiguous to each other, but is meant to exclude products of nature, e.g., naturally occurring mutants as found in their natural state.
- nucleic acids comprising sequence derived using any synthetic oligonucleotide process.
- Such a process is often done to replace a codon with a redundant codon encoding the same or a conservative amino acid, while typically introducing or removing a restriction enzyme sequence recognition site.
- the process is performed to join together nucleic acid segments of desired functions to generate a single genetic entity comprising a desired combination of functions not found in the commonly available natural forms, e.g., encoding a fusion protein.
- Restriction enzyme recognition sites are often the target of such artificial manipulations, but other site specific targets, e.g., promoters, DNA replication sites, regulation sequences, control sequences, or other useful features may be incorporated by design.
- site specific targets e.g., promoters, DNA replication sites, regulation sequences, control sequences, or other useful features may be incorporated by design.
- a similar concept is intended for a recombinant, e.g., fusion, polypeptide. This will include a dimeric repeat.
- synthetic nucleic acids which, by genetic code redundancy, encode similar polypeptides to fragments of the IL-l ⁇ or IL-l ⁇ and fusions of sequences from various different interleukin or related molecules, e.g., growth factors.
- a "fragment" in a nucleic acid context is a contiguous segment of at least about 17 nucleotides, generally at least 21 nucleotides, more generally at least 25 nucleotides, ordinarily at least 30 nucleotides, more ordinarily at least 35 nucleotides, often at least 39 nucleotides, more often at least 45 nucleotides, typically at least 50 nucleotides, more typically at least 55 nucleotides, usually at least 60 nucleotides, more usually at least 66 nucleotides, preferably at least 72 nucleotides, more preferably at least 79 nucleotides, and in particularly preferred embodiments will be at least ⁇ 5 or more nucleotides including, e.g., 100, 150, 200, 250, etc. Typically, fragments of different genetic sequences can be compared to one another over appropriate length stretches, particularly defined segments such as the domains described below.
- a nucleic acid which codes for an IL-l ⁇ or IL-l ⁇ will be particularly useful to identify genes, mRNA, and cDNA species which code for itself or closely related proteins, as well as DNAs which code for polymorphic, allelic, or other genetic variants, e.g., from different individuals or related species .
- Preferred probes for such screens are those regions of the interleukin which are conserved between different polymorphic variants or which contain nucleotides which lack specificity, and will preferably be full length or nearly so. In other situations, polymorphic variant specific sequences will be more useful .
- This invention further covers recombinant nucleic acid molecules and fragments having a nucleic acid sequence identical to or highly homologous to the isolated DNA set forth herein.
- sequences will often be operably linked to DNA segments which control transcription, translation, and DNA replication. These additional segments typically assist in expression of the desired nucleic acid segment.
- homologous nucleic acid sequences when compared to one another or sequences shown in SEQ ID NO: 1, 3 or 5, exhibit significant similarity.
- the standards for homology in nucleic acids are either measures for homology generally used in the art by sequence comparison or based upon hybridization conditions. Comparative hybridization conditions are described in greater detail below.
- Substantial identity in the nucleic acid sequence comparison context means either that the segments, or their complementary strands, when compared, are identical when optimally aligned, with appropriate nucleotide insertions or deletions, in at least about 60% of the nucleotides, generally at least 66%, ordinarily at least 71%, often at least 76%, more often at least 80%, usually at least 84%, more usually at least 8 ⁇ %, typically at least 91%, more typically at least about 93%, preferably at least about 95%, more preferably at least about 96 to 96% or more, and in particular embodiments, as high at about 99% or more of the nucleotides, including, e.g., segments encoding structural domains such as the segments described below.
- segment will hybridize under selective hybridization conditions, to a strand or its complement, typically using a sequence derived from Table 1 or 2.
- selective hybridization will occur when there is at least about 55% homology over a stretch of at least about 14 nucleotides, more typically at least about 65%, preferably at least about 75%, and more preferably at least about 90%. See, Kanehisa (1964) Nuc . Acids Res. 12:203-213.
- the length of homology comparison may be over longer stretches, and in certain embodiments will be over a stretch of at least about 17 nucleotides, generally at least about 20 nucleotides, ordinarily at least about 24 nucleotides, usually at least about 28 nucleotides, typically at least about 32 nucleotides, more typically at least about 40 nucleotides, preferably at least about 50 nucleotides, and more preferably at least about 75 to 100 or more nucleotides .
- Stringent conditions in referring to homology in the hybridization context, will be stringent combined conditions of salt, temperature, organic solvents, and other parameters typically controlled in hybridization reactions.
- Stringent temperature conditions will usually include temperatures in excess of about 30° C, more usually in excess of about 37° C, typically in excess of about 45° C, more typically in excess of about 55° C, preferably in excess of about 65° C, and more preferably in excess of about 70° C.
- Stringent salt conditions will ordinarily be less than about 500 mM, usually less than about 400 mM, more usually less than about 300 mM, typically less than about 200 mM, preferably less than about 100 mM, and more preferably less than about ⁇ O mM, even down to less than about 20 mM.
- the combination of parameters is much more important than the measure of any single parameter. See, e.g., Wetmur and Davidson (1968) J. Mol. Biol. 31:349-370, which is hereby incorporated herein by reference.
- the isolated DNA can be readily modified by nucleotide substitutions, nucleotide deletions, nucleotide insertions, and inversions of nucleotide stretches. These modifications result in novel DNA sequences which encode this protein or its derivatives . These modified sequences can be used to produce mutant proteins (muteins) or to enhance the expression of variant species . Enhanced expression may involve gene amplification, increased transcription, increased translation, and other mechanisms. Such mutant IL-1—like derivatives include predetermined or site-specific mutations of the protein or its fragments, including silent mutations using genetic code degeneracy.
- “Mutant IL-l ⁇ ” as used herein encompasses a polypeptide otherwise falling within the homology definition of the IL-l ⁇ as set forth above, but having an amino acid sequence which differs from that of other IL-1-like proteins as found in nature, whether by way of deletion, substitution, or insertion.
- site specific mutant IL-l ⁇ encompasses a protein having substantial homology with a protein of Table 1, and typically shares most of the biological activities of the form disclosed herein. Although site specific mutation sites are predetermined, mutants need not be site specific. Mammalian IL-l ⁇ mutagenesis can be achieved by making amino acid insertions or deletions in the gene, coupled with expression.
- Insertions include amino- or carboxy- terminal fusions.
- Random mutagenesis can be conducted at a target codon and the expressed mammalian IL-l ⁇ mutants can then be screened for the desired activity.
- Methods for making substitution mutations at predetermined sites in DNA having a known sequence are well known in the art, e.g., by Ml3 primer mutagenesis. See also Sambrook, et al. (1989) and Ausubel, et al . (1987 and periodic Supplements) .
- the mutations in the DNA normally should not place coding sequences out of reading frames and preferably will not create complementary regions that could hybridize to produce secondary mRNA structure such as loops or hairpins .
- the phosphoramidite method described by Beaucage and Carruthers (1981) Tetra. Letts. 22:1859-1862, will produce suitable synthetic DNA fragments .
- a double stranded fragment will often be obtained either by synthesizing the complementary strand and annealing the strand together under appropriate conditions or by adding the complementary strand using DNA polymerase with an appropriate primer sequence .
- PCR Polymerase chain reaction
- the present invention encompasses mammalian IL-l ⁇ or IL-l ⁇ , e.g., whose sequences are disclosed in SEQ ID NO: 2, 4 or 6, and described above. Allelic and other variants are also contemplated, including, e.g., fusion proteins combining portions of such sequences with others, including epitope tags and functional domains .
- the present invention also provides recombinant proteins, e.g., heterologous fusion proteins using segments from these rodent proteins .
- a heterologous fusion protein is a fusion of proteins or segments which are naturally not normally fused in the same manner.
- the fusion product of a growth factor with an interleukin is a continuous protein molecule having sequences fused in a typical peptide linkage, typically made as a single translation product and exhibiting properties derived from each source peptide.
- a similar concept applies to heterologous nucleic acid sequences.
- new constructs may be made from combining similar functional or structural domains from other related proteins, e.g., growth factors or other cytokines .
- receptor-binding or other segments may be "swapped" between different new fusion polypeptides or fragments. See, e.g., Cunningham, et al . (1989) Science 243:1330-1336; and O'Dowd, et al . (1988) J. Biol. Chem. 263:15985-15992, each of which is incorporated herein by reference.
- new chimeric polypeptides exhibiting new combinations of specificities will result from the functional linkage of receptor- binding specificities.
- a fusion protein may include a targeting domain which may serve to provide sequestering of the fusion protein to a particular organ, e.g. , a ligand portions which is specifically bound by spleen cells and would serve to accumulate in the spleen.
- a targeting domain which may serve to provide sequestering of the fusion protein to a particular organ, e.g. , a ligand portions which is specifically bound by spleen cells and would serve to accumulate in the spleen.
- Candidate fusion partners and sequences can be selected from various sequence data bases, e.g., GenBank, c/o IntelliGenetics, Mountain View, CA; and BCG, University of Wisconsin Biotechnology Computing Group, Madison, WI, which are each incorporated herein by reference.
- the present invention particularly provides muteins which act as agonists or antagonists of the IL-l ⁇ or IL- l ⁇ .
- Structural alignment of mouse IL-l ⁇ and mouse IL-l ⁇ with other members of the IL-1 family show conserved features/residues, particularly 12 ⁇ strands folded into a ⁇ -trefoil fold (see Fig IA; Table 1 and Table 3).
- the 12 IL-l ⁇ ⁇ strand domains are recited respectively (Table 1) as Leu8-Aspl4, Vall9-Asn24, Leu27-Gly31, Ile43-Asn4 ⁇ , Ser56-Val62, Gln67-Thr73, Pro77-Glu ⁇ 2, Phe99-Metl06, Leul0 ⁇ -Serll4, Phel21-Serl25, Glnl30-Thrl35, and Glnl53- Aspl56 of SEQ ID NO: 2; while the 12 IL-l ⁇ ⁇ strand domains are recited respectively (Table 1) as Serl3- Aspl9, Val24-Asn29, Ile31-Val35, Ile46-Cys51, Asp63- Val69, Ser74-Lys ⁇ 0, Pro ⁇ 5-Gly90, Serl07-Serll4, Thrll ⁇ - Serl22, Phel29-Cysl33 , Cysl3 ⁇ -Thrl43 , and
- mouse IL-l ⁇ does not bind to the known mouse IL- 1 receptor types I, II (decoy receptor), or III.
- mouse IGIF biological activity cannot be blocked with anti-type I, II, or III antibodies. This suggests that the related mouse IGIF binds to receptors related to the IL-1 receptors already isolated, but not yet identified as receptors for the IGIF.
- IL-l ⁇ the natural IL-1 receptor antagonist (IL-IRa)
- IL-lRa/IL-1 receptor type I suggest how to make a mouse IL-l ⁇ or IL-l ⁇ antagonist (See, e.g., accession numbers: U65590, gbUl9 ⁇ 44, gbUl9845, gi2173679, gi2170133, gi2172939, gbM15300, gbM289 ⁇ 3, gbU65590, gbM74294, embX04964, gi216969 ⁇ , gi216936 ⁇ emb270047, gi914939, gi2207 ⁇ 2, embX52731, embX56972 and embX12497, for various species examples of IL-1 family members) .
- This domain maps to a portion of site B in IL-l ⁇ or IL-l ⁇ (Table 2) that binds to the IL-1 second receptor subunit, suggesting that its absence confers antagonist activity as evidenced by homology comparison among other IL-1 family members.
- This loop portion of contact site B spans approximately 7-10 amino residues, while in IL-IRA the loop is "cut off” with only 2 residues remaining. Therefore, IL-IRA binds normally to receptor type I, but cannot interact with receptor type III. This makes IL- IRA into an effective IL-1 antagonist.
- the corresponding location in IL-l ⁇ or IL-l ⁇ defines a domain that forms a polypeptide loop which is part of a primary binding segment to the IL-1 receptor type (site B in Table 2) .
- the loop depicted pictorially in Figure IA as protruding into the central axis of the mature IL-l ⁇ or IL-l ⁇ protein, is located between arrows 4 and 5) . More precisely, the loop is defined for IL-l ⁇ by amino residues Pro47-Ala53 of SEQ ID NO: 2 and for IL-l ⁇ by amino residues Pro50-Glu5 ⁇ of SEQ ID NO: 6.
- IL-l ⁇ or IL-l ⁇ antagonist activity should be generated by removal all or an appropriate portion of a corresponding portion of amino acids located between ⁇ 4 and ⁇ 5. This suggests that analogous modifications to the loop between the ⁇ 4 and the ⁇ 5 strands will lead to variants with predictable biological activities.
- mouse IL-IRA it was shown that replacement of the mouse IL-IRA residues with those mouse IL-l ⁇ residues introduced IL-1 activity to the IL-IRA variant (IL-IRA could then bind type III receptor) . Similar substitutions will establish that type III receptor can probably be used by mouse IL-l ⁇ or IL-l ⁇ proteins or muteins.
- Additional site B contacts are defined in IL-l ⁇ by amino residues 8-11, 13, 112, 114-117, 158 and 160 of SEQ ID NO: 2.
- Corresponding additional site B contacts are defined in IL-l ⁇ by amino residues 3-6, ⁇ , 104, 106-109, 154 and 156 of SEQ ID NO: 2.
- Sites A and C mediate binding of IL-l ⁇ or IL-l ⁇ to the first IL-1 receptor subunit, e.g., an alpha receptor subunit.
- Site A contacts correspond in IL-l ⁇ to amino residues 13-16, 22-24, 29, 31-37, 39, 126-131, 151, and 153 of SEQ ID NO: 2; while site C contacts correspond in IL-l ⁇ to amino residues 74-98 of SEQ ID NO: 2.
- Site A contacts are defined in IL-l ⁇ by amino residues 18-21,
- site C contacts correspond in IL-l ⁇ to amino residues 81-106 of SEQ ID NO: 2.
- “Derivatives" of the mammalian IL-l ⁇ include amino acid sequence mutants, glycosylation variants, metabolic derivatives and covalent or aggregative conjugates with other chemical moieties .
- Covalent derivatives can be prepared by linkage of functionality's to groups which are found in the IL-l ⁇ amino acid side chains or at the
- N- or C- termini e.g., by means which are well known in the art.
- These derivatives can include, without limitation, aliphatic esters or amides of the carboxyl terminus, or of residues containing carboxyl side chains, O-acyl derivatives of hydroxyl group-containing residues, and N-acyl derivatives of the amino terminal amino acid or amino-group containing residues, e.g., lysine or arginine.
- Acyl groups are selected from the group of alkyl-moieties including C3 to Cl ⁇ normal alkyl, thereby forming alkanoyl aroyl species .
- glycosylation alterations are included, e.g., made by modifying the glycosylation patterns of a polypeptide during its synthesis and processing, or in further processing steps. Particularly preferred means for accomplishing this are by exposing the polypeptide to glycosylating enzymes derived from cells which normally provide such processing, e.g., mammalian glycosylation enzymes . Deglycosylation enzymes are also contemplated. Also embraced are versions of the same primary amino acid sequence which have other minor modifications, including phosphorylated .amino acid residues, e.g., phosphotyrosine, phosphoserine, or phosphothreonine .
- a major group of derivatives are covalent conjugates of the interleukin or fragments thereof with other proteins of polypeptides . These derivatives can be synthesized in recombinant culture such as N- or
- Preferred derivatization sites with cross-linking agents are at free amino groups, carbohydrate moieties, and cysteine residues.
- Fusion polypeptides between the interleukin and other homologous or heterologous proteins are also provided.
- Homologous polypeptides may be fusions between different growth factors, resulting in, for instance, a hybrid protein exhibiting ligand specificity for multiple different receptors, or a ligand which may have broadened or weakened specificity of binding to its receptor.
- heterologous fusions may be constructed which would exhibit a combination of properties or activities of the derivative proteins.
- Typical examples are fusions of a reporter polypeptide, e.g., luciferase, with a segment or domain of a receptor, e.g., a ligand-binding segment, so that the presence or location of a desired ligand may be easily determined.
- GST glutathione-S-transferase
- bacterial ⁇ -galactosidase bacterial ⁇ -galactosidase
- trpE bacterial ⁇ -galactosidase
- Protein A ⁇ -lactamase
- alpha amylase alpha amylase
- alcohol dehydrogenase yeast alpha mating factor
- a double stranded fragment will often be obtained either by synthesizing the complementary strand and annealing the strand together under appropriate conditions or by adding the complementary strand using DNA polymerase with an appropriate primer sequence .
- polypeptides may also have amino acid residues which have been chemically modified by phosphorylation, sulfonation, biotinylation, or the addition or removal of other moieties, particularly those which have molecular shapes similar to phosphate groups.
- the modifications will be useful labeling reagents, or serve as purification targets, e.g., affinity ligands.
- Fusion proteins will typically be made by either recombinant nucleic acid methods or by synthetic polypeptide methods . Techniques for nucleic acid manipulation and expression are described generally, for example, in Sambrook, et al . (1989) Molecular Cloning: A Laboratory Manual (2d ed. ) , Vols.
- the present invention relates to substantially purified peptide fragments of IL-l ⁇ or IL-l ⁇ that block binding between IL-1 family members and a target receptor.
- Such peptide fragments could represent research and diagnostic tools in the study of inflammatory reactions to antigenic challenge and the development of more effective anti-inflammatory therapeutics.
- pharmaceutical compositions comprising isolated and purified peptide fragments of IL- l ⁇ or IL-l ⁇ may represent effective anti-inflammatory therapeutics .
- substantially purified refers to a molecule, such as a peptide that is substantially free of other proteins, lipids, carbohydrates, nucleic acids, or other biological materials with which it is naturally associated.
- a substantially pure molecule such as a polypeptide, can be at least 60%, by dry weight, the molecule of interest.
- IL-l ⁇ or IL-l ⁇ peptides can be purified using standard protein purification methods and the purity of the polypeptides can be determined using standard methods including, e.g., polyacrylamide gel electrophoresis (e.g., SDS-PAGE), column chromatography (e.g., high performance liquid chromatography (HPLC) ) , and amino-terminal amino acid sequence analysis .
- the invention relates not only to fragments of naturally-occurring IL-l ⁇ or IL-l ⁇ , but also to IL-l ⁇ or IL-l ⁇ mutants and chemically synthesized derivatives of IL-l ⁇ or IL-l ⁇ that block binding between IL-1 family members and a target receptor.
- IL-l ⁇ or IL-l ⁇ changes in the amino acid se-quence of IL-l ⁇ or IL-l ⁇ are contemplated in the present invention.
- IL-l ⁇ or IL-l ⁇ can be altered by changing the nucleic acid sequence encoding the protein.
- Illustrative amino acid substitutions include the changes of: alanine to serine; arginine to lysine; asparagine to glutamine or histidine; aspartate to glutamate; cysteine to serine; glutamine to asparagine; glutamate to aspartate; glycine to proline; histidine to asparagine or glutamine; isoleucine to leucine or valine; leucine to valine or isoleucine; lysine to arginine, glutamine, or glutamate; methionine to leucine or isoleucine; phenylalanine to tyrosine, leucine or methionine; serine to threonine; threonine to serine; tryptophan to tyrosine; tyrosine to tryptophan or phenylalanine; valine to isoleucine or leucine.
- variants and fragments of IL-l ⁇ or IL-l ⁇ can be used in the present invention.
- Variants include analogs, homologues, derivatives, muteins, and mimetics of IL-l ⁇ or IL-l ⁇ that retain the ability to block binding between IL-1 family members and a target receptor.
- Fragments of the IL-l ⁇ or IL-l ⁇ refer to portions of the amino acid sequence of IL-l ⁇ or IL-l ⁇ as defined in SEQ ID NO: 2, 4 or 6 that also retain this ability.
- the variants and fragments can be generated directly from IL-l ⁇ or IL-l ⁇ itself by chemical modification, by proteolytic enzyme digestion, or by combinations thereof. Additionally, genetic engineering techniques, as well as methods of synthesizing polypeptides directly from amino acid residues, can be employed.
- Non-peptide compounds that mimic the binding and function of IL-l ⁇ or IL-l ⁇ can be produced by the approach outlined in Saragovi, et al . (1991) Science 253:792-95.
- Mimetics are molecules which mimic elements of protein secondary structure. See, e.g., Johnson et al . , "Peptide Turn Mimetics,” in Pezzuto, et al. (eds. 1993) Biotechnology and Pharmacy, Chapman and Hall, New York.
- the underlying rationale behind the use of peptide mimetics is that the peptide backbone of proteins exists chiefly to orient amino acid side chains in such a way as to facilitate molecular interactions.
- appropriate mimetics can be considered to be the equivalent of IL-l ⁇ or IL-l ⁇ itself.
- Variants and fragments also can be created by recombinant techniques employing genomic or cDNA cloning methods.
- Site-specific and region-directed mutagenesis techniques can be employed. See, e.g., vol. 1, ch. 8 in Ausubel, et al . (eds. 1989 and periodic updates) Current Protocols in Molecular Bioloov Wiley and Sons; and Oxender and Fox (eds.) Protein Engineering Liss, Inc.
- linker-scanning and PCR-mediated techniques can be employed for mutagenesis. See, e.g., Erlich (ed. 1989) PCR Technology Stockton Press.
- This invention also contemplates the use of derivatives of IL-l ⁇ other than variations in amino acid sequence or glycosylation.
- Such derivatives may involve covalent or aggregative association with chemical moieties .
- These derivatives generally fall into three classes: (1) salts, (2) side chain and terminal residue covalent modifications, and (3) adsorption complexes, for example with cell membranes.
- covalent or aggregative derivatives are useful as immunogens, as reagents in immunoassays, or in purification methods such as for affinity purification of a receptor or other binding molecule, e.g., an antibody.
- an IL- l ⁇ ligand can be immobilized by covalent bonding to a solid support such as cyanogen bromide-activated SEPHAROSE, by methods which are well known in the art, or adsorbed onto polyolefin surfaces, with or without glutaraldehyde cross-linking, for use in the assay or purification of IL-l ⁇ receptor, antibodies, or other similar molecules.
- the IL-l ⁇ can also be labeled with a detectable group, for example radio-iodinated by the chloramine T procedure, covalently bound to rare earth chelates, or conjugated to another fluorescent moiety for use in diagnostic assays.
- An IL-l ⁇ of this invention can be used as an immunogen for the production of antisera or antibodies specific, e.g., capable of distinguishing between other IL-1 family members and an IL-l ⁇ , for the interleukin or any fragments thereof .
- the purified interleukin can be used to screen monoclonal antibodies or antigen-binding fragments prepared by immunization with various forms of impure preparations containing the protein.
- the term "antibodies" also encompasses antigen binding fragments of natural antibodies .
- the purified interleukin can also be used as a reagent to detect any antibodies generated in response to the presence of elevated levels of expression, or immunological disorders which lead to antibody production to the endogenous cytokine.
- IL-l ⁇ fragments may also serve as immunogens to produce the antibodies of the present invention, as described immediately below.
- this invention contemplates antibodies having binding affinity to or being raised against the amino acid sequence shown in SEQ ID NO: 2, fragments thereof, or homologous peptides.
- this invention contemplates antibodies having binding affinity to, or having been raised against, specific fragments which are predicted to be, or actually are, exposed at the exterior protein surface of the native cytokine.
- in vitro assays of the present invention will often use antibodies or ligand binding segments of these antibodies, or fragments attached to solid phase substrates. These assays will also allow for the diagnostic determination of the effects of either binding region mutations and modifications, or ligand mutations and modifications, e.g., ligand analogs.
- This invention also contemplates the use of competitive drug screening assays, e.g., where neutralizing antibodies to the interleukin or fragments compete with a test compound for binding to a receptor or antibody.
- the neutralizing antibodies or fragments can be used to detect the presence of any polypeptide which shares one or more binding sites to a receptor and can also be used to occupy binding sites on a receptor that might otherwise bind an interleukin.
- Nucleic Acids and Protein DNA which encodes the protein or fragments thereof can be obtained by chemical synthesis, screening cDNA libraries, or by screening genomic libraries prepared from a wide variety of cell lines or tissue samples. Natural sequences can be isolated using standard methods and the sequences provided herein, e.g., in SEQ ID NO: 1. Other species counterparts can be identified by hybridization techniques, or by various PCR techniques, combined with or by searching in sequence databases.
- This DNA can be expressed in a wide variety of host cells for the synthesis of a full-length interleukin or fragments which can in turn, for example, be used to generate polyclonal or monoclonal antibodies; for binding studies; for construction and expression of modified agonist/antagonist molecules; and for structure/function studies.
- Each variant or its fragments can be expressed in host cells that are transformed or transfected with appropriate expression vectors.
- These molecules can be substantially free of protein or cellular contaminants, other than those derived from the recombinant host, and therefore are particularly useful in pharmaceutical compositions when combined with a pharmaceutically acceptable carrier and/or dilutent.
- the protein, or portions thereof, may be expressed as fusions with other proteins.
- Expression vectors are typically self-replicating DNA or RNA constructs containing the desired receptor gene or its fragments, usually operably linked to suitable genetic control elements that are recognized in a suitable host cell. These control elements are capable of effecting expression within a suitable host. The specific type of control elements necessary to effect expression will depend upon the eventual host cell used.
- the genetic control elements can include a prokaryotic promoter system or a eukaryotic promoter expression control system, and typically include a transcriptional promoter, an optional operator to control the onset of transcription, transcription enhancers to elevate the level of mRNA expression, a sequence that encodes a suitable ribosome binding site, and sequences that terminate transcription and translation.
- Expression vectors also usually contain an origin of replication that allows the vector to replicate independently of the host cell.
- the vectors of this invention include those which contain DNA which encodes a protein, as described, or a fragment thereof encoding a biologically active equivalent polypeptide.
- the DNA can be under the control of a viral promoter and can encode a selection marker.
- This invention further contemplates use of such expression vectors which are capable of expressing eukaryotic cDNA coding for such a protein in a prokaryotic or eukaryotic host, where the vector is compatible with the host and where the eukaryotic cDNA coding for the receptor is inserted into the vector such that growth of the host containing the vector expresses the cDNA in question.
- expression vectors are designed for stable replication in their host cells or for amplification to greatly increase the total number of copies of the desirable gene per cell. It is not always necessary to require that an expression vector replicate in a host cell, e.g. , it is possible to effect transient expression of the interleukin protein or its fragments in various hosts using vectors that do not contain a replication origin that is recognized by the host cell. It is also possible to use vectors that cause integration of the protein encoding portion or its fragments into the host DNA by recombination.
- Vectors as used herein, comprise plasmids, viruses, bacteriophage, integratable DNA fragments, and other vehicles which enable the integration of DNA fragments into the genome of the host.
- Expression vectors are specialized vectors which contain genetic control elements that effect expression of operably linked genes. Plasmids are the most commonly used form of vector but all other forms of vectors which serve an equivalent function and which are, or become, known in the art are suitable for use herein. See, e.g., Pouwels, et al. (1985 and Supplements) Cloning Vectors : A Laboratory Manual , Elsevier, N.Y. , and Rodriquez, et al . (eds) Vectors : A Survey of Molecular Cloning Vectors and Their Uses, Buttersworth, Boston, 19 ⁇ , which are incorporated herein by reference.
- Transformed cells are cells, preferably mammalian, that have been transformed or transfected with receptor vectors constructed using recombinant DNA techniques.
- Transformed host cells usually express the desired protein or its fragments, but for purposes of cloning, amplifying, and manipulating its DNA, do not need to express the subject protein.
- This invention further contemplates culturing transformed cells in a nutrient medium, thus permitting the interleukin to accumulate in the culture.
- the protein can be recovered, either from the culture or from the culture medium.
- nucleic sequences are operably linked when they are functionally related to each other.
- DNA for a pre-sequence or secretory leader is operably linked to a polypeptide if it is expressed as a pre-protein or participates in directing the polypeptide to the cell membrane or in secretion of the polypeptide.
- a promoter is operably linked to a coding sequence if it controls the transcription of the polypeptide;
- a ribosome binding site is operably linked to a coding sequence if it is positioned to permit translation.
- operably linked means contiguous and in reading frame, however, certain genetic elements such as repressor genes are not contiguously linked but still bind to operator sequences that in turn control expression.
- Suitable host cells include prokaryotes, lower eukaryotes, and higher eukaryotes .
- Prokaryotes include both gram negative and gram positive organisms, e.g., E. coli and B. subtilis .
- Lower eukaryotes include yeasts, e.g., S. cerevisiae and Pichia, and species of the genus Dictyostelium.
- Higher eukaryotes include established tissue culture cell lines from animal cells, both of non-mammalian origin, e.g., insect cells, and birds, and of mammalian origin, e.g., human, primates, and rodents.
- Prokaryotic host-vector systems include a wide variety of vectors for many different species. As used herein, E. coli and its vectors will be used generically to include equivalent vectors used in other prokaryotes.
- a representative vector for amplifying DNA is pBR322 or many of its derivatives .
- Vectors that can be used to express the receptor or its fragments include, but are not limited to, such vectors as those containing the lac promoter (pUC-series) ; trp promoter (pBR322-trp) ; Ipp promoter (the pIN-series) ; lambda-pP or pR promoters (pOTS) ; or hybrid promoters such as ptac (pDR540) .
- yeast vectors typically consist of a replication origin (unless of the integrating type) , a selection gene, a promoter, DNA encoding the receptor or its fragments, and sequences for translation termination, polyadenylation, and transcription termination.
- Suitable expression vectors for yeast include such constitutive promoters as 3-phosphoglycerate kinase and various other glycolytic enzyme gene promoters or such inducible promoters as the alcohol dehydrogenase 2 promoter or metallothionine promoter.
- Suitable vectors include derivatives of the following types: self-replicating low copy number (such as the YRp-series) , self-replicating high copy number (such as the YEp-series) ; integrating types (such as the Yip-series) , or mini-chromosomes (such as the YCp-series) .
- Higher eukaryotic tissue culture cells are normally the preferred host cells for expression of the functionally active interleukin protein.
- any higher eukaryotic tissue culture cell line is workable, e.g., insect baculovirus expression systems, whether from an invertebrate or vertebrate source.
- mammalian cells are preferred. Transformation or transfection and propagation of such cells has become a routine procedure.
- useful cell lines include HeLa cells, Chinese hamster ovary (CHO) cell lines, baby rat kidney (BRK) cell lines, insect cell lines, bird cell lines, and monkey (COS) cell lines.
- Expression vectors for such cell lines usually include an origin of replication, a promoter, a translation initiation site, RNA splice sites (if genomic DNA is used) , a polyadenylation site, and a transcription termination site. These vectors also usually contain a selection gene or amplification gene. Suitable expression vectors may be plasmids, viruses, or retroviruses carrying promoters derived, e.g., from such sources as from adenovirus, SV40, parvoviruses, vaccinia virus, or cytomegalovirus . Representative examples of suitable expression vectors include pCDNAl; pCD, see Okayama, et al. (1985) Mol. Cell Biol. 5:1136-1142; pMClneo PolyA, see Thomas, et al . (1987) Cell 51:503-512; and a baculovirus vector such as pAC 373 or pAC 610.
- an open reading frame usually encodes a polypeptide that consists of a mature or secreted product covalently linked at its N-terminus to a signal peptide.
- the signal peptide is cleaved prior to secretion of the mature, or active, polypeptide.
- the cleavage site can be predicted with a high degree of accuracy from empirical rules, e.g., von-Heijne (1986) Nucleic Acids Research 14:4683-4690, and the precise amino acid composition of the signal peptide does not appear to be critical to its function, e.g., Randall, et al. (1989) Science 243:1156-1159; Kaiser et al . (1987) Science 235:312-317.
- polypeptides it will often be desired to express these polypeptides in a system which provides a specific or defined glycosylation pattern.
- the usual pattern will be that provided naturally by the expression system.
- the pattern will be modifiable by exposing the polypeptide, e.g., an unglycosylated form, to appropriate glycosylating proteins introduced into a heterologous expression system.
- the interleukin gene may be co-transformed with one or more genes encoding mammalian or other glycosylating enzymes. Using this approach, certain mammalian glycosylation patterns will be achievable in prokaryote or other cells .
- the source of IL-l ⁇ or IL-l ⁇ can be a eukaryotic or prokaryotic host expressing recombinant IL-l ⁇ or IL-l ⁇ DNA, such as is described above.
- the source can also be a cell line such as mouse Swiss 3T3 fibroblasts, but other mammalian cell lines are also contemplated by this invention, with the preferred cell line being from the human species .
- the rodent IL-l ⁇ , fragments, or derivatives thereof can be prepared by conventional processes for synthesizing peptides. These include processes such as are described in Stewart and Young (1984) Solid Phase Peptide Synthesis, Pierce Chemical Co., Rockford, IL; Bodanszky and Bodanszky (1984) The Practice of Peptide Synthesis, Springer-Verlag, New York; and Bodanszky (1984) The Principles of Peptide Synthesis, Springer-Verlag, New York; all of each which are incorporated herein by reference.
- an azide process for example, an acid chloride process, an acid anhydride process, a mixed anhydride process, an active ester process (for example, p-nitrophenyl ester, N-hydroxysuccinimide ester, or cyanomethyl ester) , a carbodiimidazole process, an oxidative-reductive process, or a dicyclohexylcarbodiimide (DCCD) /additive process
- Solid phase and solution phase syntheses are both applicable to the foregoing processes. Similar techniques can be used with the partial IL-l ⁇ sequence.
- the IL-l ⁇ protein, fragments, or derivatives are suitably prepared in accordance with the above processes as typically employed in peptide synthesis, generally either by a so-called stepwise process which comprises condensing an amino acid to the terminal amino acid, one by one in sequence, or by coupling peptide fragments to the terminal .amino acid.
- Amino groups that are not being used in the coupling reaction typically must be protected to prevent coupling at an incorrect location. If a solid phase synthesis is adopted, the
- C-terminal amino acid is bound to an insoluble carrier or support through its carboxyl group.
- the insoluble carrier is not particularly limited as long as it has a binding capability to a reactive carboxyl group.
- examples of such insoluble carriers include halomethyl resins, such as chloromethyl resin or bromomethyl resin, hydroxymethyl resins, phenol resins, tert-alkyloxycarbonylhydrazidated resins, and the like.
- An amino group-protected amino acid is bound in sequence through condensation of its activated carboxyl group and the reactive amino group of the previously formed peptide or chain, to synthesize the peptide step by step. After synthesizing the complete sequence, the peptide is split off from the insoluble carrier to produce the peptide.
- This immunoabsorbant affinity chromatography is carried out by first linking the antibodies to a solid support and then contacting the linked antibodies with solubilized lysates of appropriate cells, lysates of other cells expressing the interleukin, or lysates or supernatants of cells producing the protein as a result of DNA techniques, see below.
- the purified protein will be at least about 40% pure, ordinarily at least about 50% pure, usually at least about 60% pure, typically at least about 70% pure, more typically at least about 80% pure, preferable at least about 90% pure and more preferably at least about 95% pure, and in particular embodiments, 97%- 99% or more. Purity will usually be on a weight basis, but can also be on a molar basis. Different assays will be applied as appropriate.
- antibody or "antibody molecule” as used in this invention includes intact molecules as well as fragments thereof, such as Fab, F(ab') 2 , and Fv which are capable of binding the epitopic determinant. These antibody fragments retain some ability to selectively bind with its antigen or receptor and are defined as follows: (1) Fab, the fragment which contains a monovalent antigen-binding fragment of an antibody molecule can be produced by digestion of whole antibody with the enzyme papain to yield an intact light chain and a portion of one heavy chain; (2) Fab', the fragment of an antibody molecule can be obtained by treating whole antibody with pepsin, followed by reduction, to yield an intact light chain and a portion of the heavy chain; two
- Fab' fragments are obtained per antibody molecule; (3) (Fab') 2/ the fragment of the antibody that can be obtained by treating whole antibody with the enzyme pepsin without subsequent reduction; F(ab')2 is a dimer of two Fab' fragments held together by two disulfide bonds; (4) Fv, defined as a genetically engineered fragment containing the variable region of the light chain and the variable region of the heavy chain expressed as two chains; and (5) Single chain antibody (“SCA”), defined as a genetically engineered molecule containing the variable region of the light chain, the variable region of the heavy chain, linked by a suitable polypeptide linker as a genetically fused single chain molecule.
- SCA Single chain antibody
- antibody molecule in its various forms as used herein contemplates both an intact antibody (immunoglobulin) molecule and an immunologically active portion of an antibody (immunoglobulin) molecule. Recombinant methods may be applied to make these fragments .
- the term "monoclonal antibody” refers to a population of one species of antibody molecule of antigen-specificity.
- a monoclonal antibody contains only one species of antibody combining site capable of immunoreacting with a particular antigen and thus typically displays a single binding affinity for that antigen.
- a monoclonal antibody may therefore contain a bispecific antibody molecule having two antibody combining sites, each immunospecific for a different antigen.
- the first antibody molecule is affixed to a solid support.
- the antibody molecules in a phage display combinatorial library are also monoclonal antibodies.
- epitopic determinants means any antigenic determinant on an antigen to which the paratope of an antibody binds .
- Epitopic determinants usually consist of chemically active surface groupings of molecules such as amino acids or sugar side chains and usually have specific three dimensional structural characteristics, as well as specific charge characteristics .
- complex refers to the product of a specific binding agent-ligand reaction.
- An exemplary complex is an immunoreaction product formed by an antibody-antigen reaction.
- antigen refers to a polypeptide or protein that is able to specifically bind to (immunoreact with) an antibody and form an immunoreaction product
- the site on the antigen with which the antibody binds is referred to as an antigenic determinant or epitope, and the labeling should be detectable, e.g., 2X, 5X or more above background.
- the method of the invention for detection of antibodies that bind to novel epitopes in a sample is performed in vitro, for example, in immunoassays in which the antibodies can be identified in liquid phase or bound to a solid phase carrier.
- the method is performed with a capture antibody bound to a solid support.
- the capture antibody is a monoclonal antibody molecule.
- Examples of types of immunoassays which can be utilized to detect novel antibodies in a sample include competitive and non-competitive immunoassays in either a direct or indirect format .
- Examples of such immunoassays are the radioimmunoassay (RIA) and the sandwich (immunometric) assay.
- Detection of the antibodies can be done utilizing immunoassays which are run in either the forward, reverse, or simultaneous modes, including competition immunoassays and immunohistochemical assays on physiological samples.
- the method of the invention utilizes a forward immunoassay. Those of skill in the art will know, or can readily discern, other immunoassay formats without undue experimentation.
- Solid phase-bound antibody molecules are bound by adsorption from an aqueous medium, although other modes of affixation, such as covalent coupling or other well known means of affixation to the solid matrix can be used.
- the first antibody molecule is bound to a support before forming an immunocomplex with antigen, however, the immunocomplex can be formed prior to binding the complex to the solid support.
- Non-specific protein binding sites on the surface of the solid phase support are preferably blocked.
- an aqueous solution of a protein free from interference with the assay such as bovine, horse, or other serum albumin that is also free from contamination with the antigen is admixed with the solid phase to adsorb the admixed protein onto the surface of the antibody-containing solid support at protein binding sites on the surface that are not occupied by the antibody molecule.
- a typical aqueous protein solution contains about 2- 10 weight percent bovine serum albumin in PBS at a pH of about 7-8.
- the aqueous protein solution-solid support mixture is typically maintained for a time period of at least one hour at a temperature of about 4°-37°C and the resulting solid phase is thereafter rinsed free of unbound protein.
- the first preselected antibody can be bound to many different carriers and used to detect novel epitope binding antibodies in a sample.
- carriers include glass, polystyrene, polypropylene, polyethylene, dextran, nylon, amyloses, natural and modified celluloses, polyacrylamides , agaroses, and magnetite.
- the nature of the carrier can be either soluble or insoluble for purposes of the invention. Those skilled in the art will know of other suitable carriers for binding antibodies, or will be able to ascertain such, using routine experimentation.
- an antibody for detection in these immunoassays can be detectably labeled in various ways.
- labels and methods of labeling known to those of ordinary skill in the art.
- the types of labels which can be used in the present invention include enzymes, radioisotopes, fluorescent compounds, colloidal metals, chemiluminescent compounds, and bio-luminescent compounds.
- Those of ordinary skill in the art will know of other suitable labels for binding to the monoclonal antibodies of the invention, or will be able to ascertain such, using routine experimentation.
- the binding of these labels to the antibodies used in the method of the invention can be done using standard techniques common to those of ordinary skill in the art.
- Antibodies which bind to IL-l ⁇ or IL-l ⁇ polypeptides of the invention can be prepared using an intact polypeptide or fragments containing small peptides of interest as the immunizing antigen.
- the polypeptide or a peptide used to immunize an animal can be derived from translated cDNA or chemical synthesis which can be conjugated to a carrier protein, if desired.
- Such commonly used carriers which are chemically coupled to the peptide include keyhole limpet hemocyanin (KLH) , t yroglobulin, bovine serum albumin (BSA) , and tetanus toxoid.
- KLH keyhole limpet hemocyanin
- BSA bovine serum albumin
- the coupled peptide is then used to immunize the animal (e.g., a mouse, a rat, or a rabbit) .
- polyclonal or monoclonal antibodies can be further purified, for example, by binding to and elution from a matrix to which the polypeptide or a peptide to which the antibodies were raised is bound.
- a matrix to which the polypeptide or a peptide to which the antibodies were raised is bound.
- Those of skill in the art will know of various techniques common in the immunology arts for purification and/or concentration of polyclonal antibodies, as well as monoclonal antibodies See, e.g., Coligan, et al . (current ed.) Unit 9, Current Protocols in Immunology, Wiley Interscience.
- an anti-idiotypic monoclonal antibody made to a first monoclonal antibody will have a binding domain in the hypervariable region which is the "image" of the epitope bound by the first monoclonal antibody.
- the preparation of polyclonal antibodies is well- known to those skilled in the art. See, e.g., Green, et al . "Production of Polyclonal Antisera” pages 1-5 in Manson (ed. ) Immunochemical Protocols Humana Press; Production of Polyclonal Antisera in Rabbits, Rats, Mice and Hamsters section 2.4.1 in Coligan, et al . Current Protocols in Immunology.
- monoclonal antibodies can be obtained by injecting mice with a composition comprising an antigen, verifying the presence of antibody production by removing a serum sample, removing the spleen to obtain B lymphocytes, fusing the B lymphocytes with myeloma cells to produce hybridomas, cloning the hybridomas, selecting positive clones that produce antibodies to the antigen, and isolating the antibodies from the hybridoma cultures.
- Monoclonal antibodies can be isolated and purified from hybridoma cultures by a variety of well-established techniques. Such isolation techniques include affinity chromatography with Protein-A Sepharose, size-exclusion chromatography, and ion- exchange chromatography. See, e.g.
- Multiplication in vitro may be carried out in suitable culture media such as Dulbecco's Modified Eagle Medium or RPMI 1640 medium, optionally replenished, e.g., by a mammalian serum such as fetal calf serum or trace elements and growth- sustaining supplements such as normal mouse peritoneal exudate cells, spleen cells, bone marrow macrophages .
- suitable culture media such as Dulbecco's Modified Eagle Medium or RPMI 1640 medium
- a mammalian serum such as fetal calf serum or trace elements
- growth- sustaining supplements such as normal mouse peritoneal exudate cells, spleen cells, bone marrow macrophages .
- Production in vitro provides relatively pure antibody preparations and allows scale-up to yield large amounts of the desired antibodies.
- Large scale hybridoma cultivation can be carried out by homogenous suspension culture in an airlift reactor, in a continuous stirrer reactor, or in immobilized or entrapped cell
- Multiplication in vivo may be carried out by injecting cell clones into mammals histocompatible with the parent cells, e.g. , syngeneic mice, to cause growth of antibody- producing tumors.
- the animals are primed with a hydrocarbon, especially oils such as pristane (tetramethylpentadecane) prior to injection. After one to three weeks, the desired monoclonal antibody is recovered from the body fluid of the animal.
- antibodies of the present invention may also be derived from subhuman primate antibody.
- General techniques for raising therapeutically useful antibodies in baboons may be found, for example, in Goldenberg, et al . (1991) WO 91/11465; and Losman, et al . (1990) Int. J. Cancer 46:310.
- a therapeutically useful anti-IL-l ⁇ or anti-IL-l ⁇ antibody may be derived from a "humanized" monoclonal antibody.
- Humanized monoclonal antibodies are produced by transferring mouse complementary determining regions from heavy and light variable chains of the mouse immunoglobulin into a human variable domain, and then substituting human residues in the framework regions of the murine counterparts .
- the use of antibody components derived from humanized monoclonal antibodies obviates potential problems associated with the immunogenicity of murine constant regions.
- General techniques for cloning murine immunoglobulin variable domains are described, for example, by Orlandi, et al . (19 ⁇ 9) Proc. Nat ' 1 Acad. Sci. USA ⁇ 6:3 ⁇ 33. Techniques for producing humanized monoclonal antibodies are described, e.g., by Jones et al.
- Antibodies of the invention also may be derived from human antibody fragments isolated from a combinatorial immunoglobulin library. See, for example, Barbas, et al . (1991) Methods: A Companion to Methods in Enzymology, vol. 2, page 119; and Winter, et al . (1994) Ann. Rev.
- Cloning and expression vectors that are useful for producing a human immunoglobulin phage library can be obtained, e.g., from STRATAGENE Cloning Systems (La Jolla, CA) .
- antibodies of the present invention may be derived from a human monoclonal antibody.
- Such antibodies are obtained from transgenic mice that have been "engineered” to produce specific human antibodies in response to antigenic challenge.
- elements of the human heavy and light chain loci are introduced into strains of mice derived from embryonic stem cell lines that contain targeted disruptions of the endogenous heavy and light chain loci.
- the transgenic mice can synthesize human antibodies specific for human antigens, and the mice can be used to produce human antibody-secreting hybridomas .
- Antibody fragments of the present invention can be prepared by proteolytic hydrolysis of the antibody or by expression in E. coli of DNA encoding the fragment. Antibody fragments can be obtained by pepsin or papain digestion of whole antibodies by conventional methods .
- antibody fragments can be produced by enzymatic cleavage of antibodies with pepsin to provide a 5S fragment denoted F(ab')2- This fragment can be further cleaved using a thiol reducing agent, and optionally a blocking group for the sulfhydryl groups resulting from cleavage of disulfide linkages, to produce 3.5 S Fab' monovalent fragments.
- an enzymatic cleavage using papain produces two monovalent Fab fragments and an Fc fragment directly.
- Fv fragments comprise an association of V H and V L chains. This association may be noncovalent, as described in Inbar, et al . (1972) Proc . Nat ' 1 Acad. Sci. USA 69:2659.
- the variable chains can be linked by an intermolecular disulfide bond or crosslinked by chemicals such as glutaraldehyde . See, e.g., Sandhu (1992) Crit. Rev. Biotech. 12:437.
- the Fv fragments comprise V H and V L chains connected by a peptide linker.
- sFv single-chain antigen binding proteins
- Another form of an antibody fragment is a peptide coding for a single complementarity-determining region
- CDR CDR peptides
- minimal recognition units can be obtained by constructing genes encoding the CDR of an antibody of interest. Such genes are prepared, for example, by using the polymerase chain reaction to synthesize the variable region from RNA of antibody- producing cells. See, for example, Larrick, et al . (1991) Methods: A Companion to Methods in Enzymology, vol. 2, page 106.
- Antibodies can be raised to the various mammalian, e.g., rodent IL-l ⁇ and/or IL-l ⁇ proteins and fragments thereof, both in naturally occurring native forms and in their recombinant forms, the difference being that antibodies to the active ligand are more likely to recognize epitopes which are only present in the native conformations.
- Denatured antigen detection can also be useful in, e.g., Western analysis.
- Anti-idiotypic antibodies are also contemplated, which would be useful as agonists or antagonists of a natural receptor or an antibody.
- a number of immunogens may be used to produce antibodies specifically reactive with thymokine proteins. Recombinant protein is the preferred immunogen for the production of monoclonal or polyclonal antibodies.
- Naturally occurring protein may also be used either in pure or impure form.
- Synthetic peptides made using the human or mouse lymphotactin protein sequences described herein may also used as an immunogen for the production of antibodies to thymokines .
- Recombinant protein can be expressed in eukaryotic or prokaryotic cells as described herein, and purified as described. The product is then injected into an animal capable of producing antibodies. Either monoclonal or polyclonal antibodies may be generated for subsequent use in immunoassays to measure the protein.
- an immunogen preferably a purified protein
- animals are immunized with the mixture.
- the animal's immune response to the immunogen preparation is monitored by taking test bleeds and determining the titer of reactivity to the thymokine protein of interest .
- blood is collected from the animal and antisera are prepared. Further fractionation of the antisera to enrich for antibodies reactive to the protein can be done if desired. See Harlow and Lane.
- Monoclonal antibodies may be obtained by various techniques f-amiliar to those skilled in the art. Briefly, spleen cells from an animal immunized with a desired antigen are immortalized, commonly by fusion with a myeloma cell. Alternative methods of immortalization include transformation with Epstein Barr Virus, oncogenes, or retroviruses, or other methods well known in the art. Colonies arising from single immortalized cells are screened for production of antibodies of the desired specificity and affinity for the antigen, and yield of the monoclonal antibodies produced by such cells may be enhanced by various techniques, including injection into the peritoneal cavity of a vertebrate host.
- DNA se-quences which encode a monoclonal antibody or a binding fragment thereof by screening a DNA library from human B cells according to the general protocol outlined by Huse, et al. (1989) Science 246:1275-1281.
- Antibodies including binding fragments and single chain versions, against predetermined fragments of the protein can be raised by immunization of animals with conjugates of the fragments with immunogenic proteins.
- Monoclonal antibodies are prepared from cells secreting the desired antibody. These antibodies can be screened for binding to normal or defective protein, or screened for agonistic or antagonistic activity. These monoclonal antibodies will usually bind with at least a K D of about
- 1 mM more usually at least about 300 ⁇ M, typically at least about 100 ⁇ M, more typically at least about 30 ⁇ M, preferably at least about 10 ⁇ M, and more preferably at least about 3 ⁇ M or better; including 1 ⁇ M, 300 nM, 100 nM, 30 nM, etc.
- the antibodies, including antigen binding fragments, of this invention can have significant diagnostic or therapeutic value. They can be potent antagonists that bind to the interleukin and inhibit binding to the receptor or inhibit the ability of IL-l ⁇ or IL-l ⁇ to elicit a biological response. They also can be useful as non-neutralizing antibodies and can be coupled to toxins or radionuclides to bind producing cells, or cells localized to the source of the interleukin. Further, these antibodies can be conjugated to drugs or other therapeutic agents, either directly or indirectly by means of a linker.
- the antibodies of this invention can also be useful in diagnostic applications. As capture or non-neutralizing antibodies, they can bind to the interleukin without inhibiting receptor binding. As neutralizing antibodies, they can be useful in competitive binding assays. They will also be useful in detecting or quantifying IL-l ⁇ . They may be used as reagents for Western blot analysis, or for immunoprecipitation or immunopurification of the respective protein.
- Protein fra-gments may be joined to other materials, particularly polypeptides, as fused or covalently joined polypeptides to be used as immunogens.
- Mammalian IL-l ⁇ and its fra-gments may be fused or covalently linked to a variety of immunogens, such as keyhole limpet hemocyanin, bovine serum albumin, tetanus toxoid, etc. See Microbiology, Hoeber Medical Division, Harper and Row, 1969; Landsteiner (1962) Specificity of Serological Reactions , Dover Publications, New York; and Williams, et al . (1967) Methods in Immunology and Immunochemistry, Vol.
- a typical method involves hyperimmunization of an animal with an antigen. The blood of the animal is then collected shortly after the repeated immunizations and the gamma globulin is isolated.
- monoclonal antibodies from various mammalian hosts, such as mice, rodents, primates, humans, etc. Description of techniques for preparing such monoclonal antibodies may be found in, e.g., Stites, et al . (eds) Basic and Clinical Immunology (4th ed.
- hybrid cell or "hybridoma” that is capable of reproducing in vitro .
- the population of hybridomas is then screened to isolate individual clones, each of which secrete a single antibody species to the immunogen.
- the individual antibody species obtained are the products of immortalized and cloned single B cells from the immune animal generated in response to a specific site recognized on the immunogenic substance.
- the polypeptides and antibodies will be labeled by joining, either covalently or non-covalently, a substance which provides for a detectable signal .
- labels and conjugation techniques are known and are reported extensively in both the scientific and patent literature. Suitable labels include radionuclides, enzymes, substrates, cofactors, inhibitors, fluorescent moieties, chemiluminescent moieties, magnetic particles, and the like. Patents, teaching the use of such labels include U.S. Patent Nos. 3,817,837; 3,850,752; 3,939,350; 3,996,345; 4,277,437; 4,275,149; and 4,366,241.
- recombinant or chimeric immunoglobulins may be produced, see Cabilly, U.S. Patent No. 4,816,567; or made in transgenic mice, see Mendez, et al. (1997) Nature Genetics 15:146-156. These references are incorporated herein by reference.
- the antibodies of this invention can also be used for affinity chromatography in isolating the IL-l ⁇ .
- Columns can be prepared where the antibodies are linked to a solid support, e.g., particles, such as agarose, SEPHADEX, or the like, where a cell lysate may be passed through the column, the column washed, followed by increasing concentrations of a mild denaturant, whereby the purified protein will be released.
- the protein may be used to purify antibody.
- the antibodies may also be used to screen expression libraries for particular expression products. Usually the antibodies used in such a procedure will be labeled with a moiety allowing easy detection of presence of antigen by antibody binding. Antibodies raised against an IL-l ⁇ or IL-l ⁇ will also be used to raise anti-idiotypic antibodies.
- Binding Agent IL-l ⁇ /IL-l ⁇ Protein Complex
- the immunoassay typically uses a polyclonal antiserum which was raised, e.g., to a protein of SEQ ID NO: 2, 4, or 6. This antiserum is selected to have low crossreactivity against other IL-1 family members, preferably form the same species, and any such crossreactivity is removed by immunoabsorption prior to use in the immunoassay.
- the protein of SEQ ID NO: 2, 4, or 6 is isolated as described herein.
- recombinant protein may be produced in a mammalian cell line.
- An appropriate host e.g., an inbred strain of mice such as Balb/c, is immunized with the protein of SEQ ID NO: 2, 4, or 6 using a standard adjuvant, such as Freund's adjuvant, and a standard mouse immunization protocol (see Harlow and Lane) .
- a synthetic peptide derived from the sequences disclosed herein and conjugated to a carrier protein can be used an immunogen.
- Polyclonal sera are collected and titered against the immunogen protein in an immunoassay, e.g., a solid phase immunoassay with the immunogen immobilized on a solid support.
- Polyclonal antisera with a titer of 10 ⁇ or greater are selected and tested for their cross reactivity against other IL-1 family members, e.g., IL- l ⁇ , IL-l ⁇ , IL-IRA, and IL-l ⁇ , using a competitive binding immunoassay such as the one described in Harlow and Lane, supra, at pages 570-573.
- Preferably at least two IL-1 family members are used in this determination in conjunction with either IL-l ⁇ or IL-l ⁇ .
- These IL-1 family members can be produced as recombinant proteins and isolated using standard molecular biology and protein chemistry techniques as described herein.
- Immunoassays in the competitive binding format can be used for the crossreactivity determinations.
- the protein of SEQ ID NO: 2, 4, or 6 can be immobilized to a solid support. Proteins added to the assay compete with the binding of the antisera to the immobilized antigen. The ability of the above proteins to compete with the binding of the antisera to the immobilized protein is compared to the protein of SEQ ID NO: 2, 4, or 6. The percent crossreactivity for the above proteins is calculated, using standard calculations . Those antisera with less than 10% crossreactivity with each of the proteins listed above are selected and pooled. The cross-reacting antibodies are then removed from the pooled antisera by immunoabsorbtion with the above-listed proteins.
- the immunoabsorbed and pooled antisera are then used in a competitive binding immunoassay as described above to compare a second protein to the immunogen protein
- the two proteins are each assayed at a wide range of concentrations and the amount of each protein required to inhibit 50% of the binding of the antisera to the immobilized protein is determined. If the amount of the second protein required is less than twice the amount of the protein of SEQ ID NO: 2 that is required, then the second protein is said to specifically bind to an antibody generated to the immunogen.
- these IL-l ⁇ or IL-l ⁇ proteins are members of a family of homologous proteins that comprise at least 5 so far identified genes.
- the term refers not only to the amino acid sequences disclosed herein, but also to other proteins that are allelic, non-allelic or species variants.
- the term "IL-l ⁇ ” or "IL-l ⁇ ” includes nonnatural mutations introduced by deliberate mutation using conventional recombinant technology such as single site mutation, or by excising short sections of DNA encoding the respective proteins, or by substituting new amino acids, or adding new amino acids. Such minor alterations must substantially maintain the immunoidentity of the original molecule and/or its biological activity.
- these alterations include proteins that are specifically immunoreactive with a designated naturally occurring IL-1 related protein, for example, the IL-l ⁇ or IL-l ⁇ protein shown in SEQ ID NO: 2, 4, or 6.
- the biological properties of the altered proteins can be determined by expressing the protein in an appropriate cell line and measuring the appropriate effect upon lymphocytes .
- Particular protein modifications considered minor would include conservative substitution of .amino acids with similar chemical properties, as described above for the IL-1 family as a whole.
- kits and assay methods Both naturally occurring and recombinant forms of the IL-1 like molecules of this invention are particularly useful in kits and assay methods . For example, these methods would also be applied to screening for binding activity, e.g., receptors for these proteins.
- Several methods of automating assays have been developed in recent years so as to permit screening of tens of thousands of compounds per year. See, e.g., a BIOMEK automated workstation, Beckman Instruments, Palo Alto, California, and Fodor, et al . (1991) Science 251:767-773, which is incorporated herein by reference. The latter describes means for testing binding by a plurality of defined polymers synthesized on a solid substrate.
- suitable assays to screen for a receptor or agonist/antagonist homologous proteins can be greatly facilitated by the availability of large amounts of purified, soluble IL-l ⁇ or IL-l ⁇ in an active state such as is provided by this invention.
- Purified IL-l ⁇ can be coated directly onto plates for use in the aforementioned receptor screening techniques.
- non-neutralizing antibodies to these proteins can be used as capture antibodies to immobilize the respective interleukin on the solid phase, useful, e.g., in diagnostic uses.
- kits and methods for detecting the presence of the protein or its receptor.
- antibodies against the molecules may be incorporated into the kits and methods .
- the kit will have a compartment containing either a defined IL-l ⁇ peptide or gene segment or a reagent which recognizes one or the other.
- recognition reagents in the case of peptide, would be a receptor or antibody, or in the case of a gene segment, would usually be a hybridization probe.
- a preferred kit for determining the concentration of, for example, IL-l ⁇ , a sample would typically comprise a labeled compound, e.g., receptor or antibody, having known binding affinity for IL-l ⁇ , a source of IL-l ⁇
- Antibodies including antigen binding fragments, specific for mammalian IL-l ⁇ or a peptide fra-gment, or receptor fragments are useful in diagnostic applications to detect the presence of elevated levels of IL-l ⁇ and/or its fragments .
- Diagnostic assays may be homogeneous (without a separation step between free reagent and antibody-antigen complex) or heterogeneous (with a separation step) .
- unlabeled antibodies can be employed by using a second antibody which is labeled and which recognizes the antibody to IL-l ⁇ or to a particular fragment thereof.
- a second antibody which is labeled and which recognizes the antibody to IL-l ⁇ or to a particular fragment thereof.
- assays have also been extensively discussed in the literature. See, e.g. , Harlow and Lane (198 ⁇ ) Antibodies : A Laboratory Manual , CSH. , and Coligan (Ed.) (1991) and periodic supplements, Current Protocols In Immunology Greene/Wiley, New York.
- Anti-idiotypic antibodies may have similar use to serve as agonists or antagonists of IL-l ⁇ . These should be useful as therapeutic reagents under appropriate circumstances .
- the reagents for diagnostic assays are supplied in kits, so as to optimize the sensitivity of the assay.
- the protocol, and the label either labeled or unlabeled antibody, or labeled receptor is provided. This is usually in conjunction with other additives, such as buffers, stabilizers, materials necessary for signal production such as substrates for enzymes, and the like.
- the kit will also contain instructions for proper use and disposal of the contents after use.
- the kit has compartments for each useful reagent, and will contain instructions for proper use and disposal of reagents.
- the reagents are provided as a dry lyophilized powder, where the reagents may be reconstituted in an aqueous medium having appropriate concentrations for performing the assay.
- Any of the aforementioned constituents of the diagnostic assays may be used without modification or may be modified in a variety of ways.
- labeling may be achieved by covalently or non-covalently joining a moiety which directly or indirectly provides a detectable signal.
- a test compound, IL-l ⁇ , or antibodies thereto can be labeled either directly or indirectly.
- Possibilities for direct labeling include label groups: radiolabels such as 12 5 i, enzymes (U.S. Pat. No.
- the IL-l ⁇ can be immobilized on various matrixes followed by washing.
- Suitable matrices include plastic such as an ELISA plate, filters, and beads.
- Methods of immobilizing the receptor to a matrix include, without limitation, direct adhesion to plastic, use of a capture antibody, chemical coupling, and biotin-avidin.
- the last step in this approach involves the precipitation of antibody/antigen complex by any of several methods including those utilizing, e.g., an organic solvent such as polyethylene glycol or a salt such as ammonium sulfate.
- sequences can be used as probes for detecting levels of the IL-l ⁇ in patients suspected of having an immunological disorder.
- the preparation of both RNA and DNA nucleotide sequences, the labeling of the sequences, and the preferred size of the sequences has received ample description and discussion in the literature.
- an oligonucleotide probe should have at least about 14 nucleotides, usually at least about 18 nucleotides, and the polynucleotide probes may be up to several kilobases.
- Various labels may be employed, most commonly radionuclides, particularly 32p _ However, other techniques may also be employed, such as using biotin modified nucleotides for introduction into a polynucleotide.
- the biotin then serves as the site for binding to avidin or antibodies, which may be labeled with a wide variety of labels, such as radionuclides, fluorescers, enzymes, or the like.
- antibodies may be employed which can recognize specific duplexes, including DNA duplexes, RNA duplexes, DNA-RNA hybrid duplexes, or DNA-protein duplexes.
- the antibodies in turn may be labeled and the assay carried out where the duplex is bound to a surface, so that upon the formation of duplex on the surface, the presence of antibody bound to the duplex can be detected.
- probes to the novel anti-sense RNA may be carried out in any conventional techniques such as nucleic acid hybridization, plus and minus screening, recombinational probing, hybrid released translation (HRT) , and hybrid arrested translation (HART) .
- This also includes amplification techniques such as polymerase chain reaction (PCR) .
- kits which also test for the qualitative or quantitative presence of other markers are also contemplated. Diagnosis or prognosis may depend on the combination of multiple indications used as markers. Thus, kits may test for combinations of markers. See, e.g., Viallet, et al . (1989) Progress in Growth Factor Res. 1:89-97.
- This invention provides reagents with significant therapeutic value.
- the IL-l ⁇ or IL-l ⁇ (naturally occurring or recombinant) , fragments thereof, mutein agonists and antagonists, and antibodies, along with compounds identified as having binding affinity to the interleukin or its receptor or antibodies, should be useful in the treatment of conditions exhibiting abnormal expression of the interleukin. Such abnormality will typically be manifested by immunological disorders. Additionally, this invention should provide therapeutic value in various diseases or disorders associated with abnormal expression or abnormal triggering of response to the interleukin.
- the mouse IL- ⁇ has been suggested to be involved in tumors, allergies, and infectious diseases, e.g., pulmonary tuberculosis, leprosy, fulminant hepatitis, and viral infections, such as HIV.
- infectious diseases e.g., pulmonary tuberculosis, leprosy, fulminant hepatitis, and viral infections, such as HIV.
- the IL-l ⁇ and/or IL-l ⁇ or antagonist may have similar function.
- the dendritic cell expression profile shows human IL-l ⁇ primarily expressed in activated dendritic cells. Activated dendritic cells are also a major producer of IL-12, and it is thought that this dendritic cell produced IL-12 plays a major role in directing a Thl type response.
- IL-l ⁇ and IL-12 should be extremely potent in inducing IFN- ⁇ , suggesting that IL-l ⁇ or IL-l ⁇ , or antagonists thereof, may have similar function. It is possible that the combination of pro-inflammatory cytokines under certain circumstances could lead to septic shock. An antagonist, mutein or antibody, could prove very useful in this situation. See Rich (ed. ) Clinical Immunology: Principles and Practice, Mosby.
- IL-l ⁇ or IL-l ⁇ being homologous members of the IL-1 family likely play a role in modulating of local and systemic inflammatory processes (See, Durum, et al . (1986) Ann. Rev. Immunol. 3:253), through the enhancement of blood flow, induction of chemoattractants, and the enhancement and adherence of adhesion molecules resulting in the accumulation of inflammatory cells such as macrophages and neutrophils at the site of inflammation. Additionally, it is likely that IL-l ⁇ or IL-l ⁇ induce fibroblast growth and may play a role in contributing to the pathogenesis of chronic inflammation, as in rheumatoid arthritis or periodontal disease.
- IL-l ⁇ or IL-l ⁇ are also likely to play a role in systemic inflammatory reactions, such as fever, hypoglycemia, the acute phase response of the liver, reduced plasma iron and zinc, and increased plasma copper .
- a systemic reaction such as septic shock involves vasodilation, due to IL-1, most likely in combination with other cytokines, including, e.g., TNF, IFN- ⁇ , and leukemia inhibitory factor (LIF) .
- cytokines including, e.g., TNF, IFN- ⁇ , and leukemia inhibitory factor (LIF) .
- LIF leukemia inhibitory factor
- the newly described IL-l ⁇ or IL-l ⁇ are also likely to be similarly involved.
- IL-l ⁇ In the following, directed to IL-l ⁇ , similar substitution of IL-l ⁇ may be appropriate.
- Recombinant IL-l ⁇ , mutein agonists or antagonists, or IL-l ⁇ antibodies can be purified and then administered to a patient.
- These reagents can be combined for therapeutic use with additional active ingredients, e.g. , in conventional pharmaceutically acceptable carriers or diluents, along with physiologically innocuous stabilizers and excipients.
- additional active ingredients e.g.
- additional active ingredients e.g.
- additional active ingredients e.g.
- additional active ingredients e.g.
- additional active ingredients e.g., in conventional pharmaceutically acceptable carriers or diluents, along with physiologically innocuous stabilizers and excipients.
- These combinations can be sterile, e.g., filtered, and placed into dosage forms as by lyophilization in dosage vials or storage in stabilized aqueous preparations.
- Receptor screening using IL-l ⁇ or fragments thereof can be performed to identify molecules having binding affinity to the interleukin. Subsequent biological assays can then be utilized to determine if a receptor can provide competitive binding, which can block intrinsic stimulating activity. Receptor fragments can be used as a blocker or antagonist in that it blocks the activity of IL-l ⁇ . Likewise, a compound having intrinsic stimulating activity can activate the receptor and is thus an agonist in that it simulates the activity of IL- l ⁇ . This invention further contemplates the therapeutic use of antibodies to IL-l ⁇ as antagonists .
- reagents necessary for effective therapy will depend upon many different factors, including means of administration, target site, physiological state of the patient, and other medicants administered. Thus, treatment dosages should be titrated to optimize safety and efficacy. Typically, dosages used in vitro may provide useful guidance in the amounts useful for in situ administration of these reagents . Animal testing of effective doses for treatment of particular disorders will provide further predictive indication of human dosage. Various considerations are described, e.g., in Gilman, et al. (eds 1990) Goodman and Gilman's: The Pharmacological Bases of Therapeutics, 8th Ed., Pergamon Press; and Remington ' s Pharmaceutical Sciences, (current ed. ) , Mack Publishing Co., Easton, Penn.
- compositions for administration are discussed therein and below, e.g., for oral, intravenous, intraperitoneal, or intramuscular administration, transdermal diffusion, and others.
- Pharmaceutically acceptable carriers will include water, saline, buffers, and other compounds described, e.g., in the Merck Index,
- dosage ranges would ordinarily be expected to be in amounts lower than 1 mM concentrations, typically less than about 10 ⁇ M concentrations, usually less than about 100 nM, preferably less than about 10 pM (picomolar) , and most preferably less than about 1 fM (femtomolar) , with an appropriate carrier.
- Slow release formulations, or slow release apparatus will often be utilized for continuous administration.
- IL-l ⁇ fra-gments thereof, and antibodies or its fragments, antagonists, and agonists may be administered directly to the host to be treated or, depending on the size of the compounds, it may be desirable to conjugate them to carrier proteins such as ovalbumin or serum albumin prior to their administration.
- Therapeutic formulations may be administered in any conventional dosage formulation. While it is possible for the active ingredient to be administered alone, it is preferable to present it as a pharmaceutical formulation.
- Formulations comprise at least one active ingredient, as defined above, together with one or more acceptable carriers thereof . Each carrier must be both pharmaceutically and physiologically acceptable in the sense of being compatible with the other ingredients and not injurious to the patient .
- Formulations include those suitable for oral, rectal, nasal, or parenteral (including subcutaneous, intramuscular, intravenous and intradermal) administration.
- the formulations may conveniently be presented in unit dosage form and may be prepared by any methods well known in the art of pharmacy. See, e.g., Gilman, et al . (eds. 1990) Goodman and Gilman's: The Pharmacological Bases of Therapeutics, 8th Ed. , Pergamon Press; and Remington's Pharmaceutical Sciences, 17th ed. (1990), Mack Publishing Co., Easton, Penn.; Avis, et al . (eds. 1993) Pharmaceutical Dosage Forms : Parenteral Medications Dekker, NY; Lieberman, et al . (eds. 1990) Pharmaceutical Dosage Forms : Tablets Dekker, NY; and Lieberman, et al . (eds. 1990) Pharmaceutical Dosage Forms: Disperse Systems Dekker, NY.
- Another therapeutic approach included within the invention involves direct administration of reagents or compositions by any conventional administration techniques (for ex.am.ple but not restricted to local injection, inhalation, or administered systemically) , to the subject with an inflammatory disorder.
- the reagent, formulation or composition may also be targeted to specific cells or receptors by any of the methods described herein.
- the actual dosage of reagent, formulation or composition that modulates an inflammatory disorder depends on many factors, including the size and health of an organism, however one of one of ordinary- skill in the art can use the following teachings describing the methods and techniques for determining clinical dosages.
- the therapy of this invention may be combined with or used in association with other therapeutic agents, particularly agonists or antagonists of other IL-1 family members.
- IL-l ⁇ ligand herein provides means to identify a receptor, as described above. Such receptor should bind specifically to the IL-l ⁇ with reasonably high affinity.
- Various constructs are made available which allow either labeling of the IL-l ⁇ to detect its receptor. For example, directly labeling IL- l ⁇ , fusing onto it markers for secondary labeling, e.g.,
- a cytokine will bind to its receptor at a Kd of at least about 30 ⁇ M, preferably at least about 10 ⁇ M, and more preferably at least about 3 ⁇ M or better; including 1 ⁇ M, 300 nM, 100 nM, 30 nM, etc.
- Kd a Kd of at least about 30 ⁇ M, preferably at least about 10 ⁇ M, and more preferably at least about 3 ⁇ M or better; including 1 ⁇ M, 300 nM, 100 nM, 30 nM, etc.
- descriptions of how to make IL-l ⁇ will be analogously applicable to embodiments directed to IL-l ⁇ reagents and compositions.
- Combination with recombinant techniques allow fusion to appropriate segments , e.g., to a FLAG sequence or an equivalent which can be fused via a protease-removable sequence.
- appropriate segments e.g., to a FLAG sequence or an equivalent which can be fused via a protease-removable sequence.
- OIAexpress The High Level Expression & Protein Purification System QIAGEN, Inc., Chatsworth, CA.
- IL-4 and IL-10 may be applied to IL-l ⁇ and/or IL-l ⁇ , as described, e.g., in U.S. Patent No. 5,017,691 (IL-4) , USSN 07/453,951 (IL-10) , and USSN 08/110,683 (IL-10 receptor), each of which is incorporated herein by reference for all purposes.
- DNA is isolated from a genomic or cDNA library using labeled oligonucleotide probes having sequences identical or complementary to the sequences disclosed herein. Full-length probes may be used, or oligonucleotide probes may be generated by comparison of the sequences disclosed. Such probes can be used directly in hybridization assays to isolate DNA encoding thymokine proteins, or probes can be designed for use in amplification techniques such as PCR, for the isolation of DNA encoding IL-l ⁇ and IL-l ⁇ proteins.
- Various methods of amplifying target sequences can also be used to prepare DNA encoding IL-l ⁇ and IL-l ⁇ proteins.
- Polymerase chain reaction (PCR) technology is used to amplify such nucleic acid sequences directly from mRNA, from cDNA, and from genomic libraries or cDNA libraries.
- the isolated sequences encoding IL-l ⁇ or IL-l ⁇ proteins may also be used as templates for PCR amplification.
- oligonucleotide primers complementary to two 5 ' regions in the DNA region to be amplified are synthesized. The polymerase chain reaction is then carried out using the two primers. See Innis et al . (Current eds.) PCR Protocols : A Guide to Methods and Applications Academic Press, San Diego, CA. Primers can be selected to amplify the entire regions encoding a full-length IL-l ⁇ or IL-l ⁇ proteins or to amplify smaller
- DNA segments as desired can be sequenced and oligonucleotide probes can be prepared from sequence obtained using standard techniques . These probes can then be used to isolate DNA's encoding tIL-l ⁇ or IL-l ⁇ proteins.
- Oligonucleotides for use as probes are chemically synthesized according to the solid phase phosphoramidite triester method first described by Beaucage and Carruthers (1983) Tetrahedron Lett . 22(20): 1859-l ⁇ 62, or using an automated synthesizer, as described in Needham-VanDevanter et al. (19 ⁇ 4) Nucleic Acids Res. 12: 6159-6166. Purification of oligonucleotides is performed e.g.j_ by native acrylamide gel electrophoresis or by anion-exchange HPLC as described in Pearson and Regnier (19 ⁇ 3) J. Chrom. 255: 137-149.
- the sequence of the synthetic oligonucleotide can be verified using the chemical degradation method of Maxam, A.M. and Gilbert, W. in Grossman, L. and Moldave (eds.) (19 ⁇ 0) Methods in Enzymology 65: 499-560 Academic Press, New York.
- the peptide segments can also be used to produce appropriate oligonucleotides to screen a library.
- the genetic code can be used to select appropriate oligonucleotides useful as probes for screening.
- synthetic oligonucleotides will be useful in selecting desired clones from a library.
- Complementary sequences will also be used as probes or primers. Based upon identification of the likely amino terminus, other peptides should be particularly useful, e.g. , coupled with anchored vector or poly-A complementary PCR techniques or with complementary DNA of other peptides .
- oligonucleotides are designed which corresponded to conserved regions among known IL-1 family members .
- the primers are used for polymerase chain reactions on mouse genomic DNA followed by subcloning the PCR products using restriction sites placed at the 5 ' ends of the primers, picking individual E. coli colonies carrying these subcloned inserts, and using a combination of random sequencing and hybridization analysis to eliminate known IL-1 family members.
- PCR products are gel-purified, digested with appropriate restriction enzymes, gel- purified again, and subcloned in the Bluescript vector (Stratagene, San Diego, CA) .
- Bacterial colonies carrying individual subclones are picked into 96 well microtiter plates, and multiple replicas are prepared by plating the cells onto nitrocellulose.
- the replicate filters are hybridized to probes representing known members of the IL-1 family, and DNA is prepared from non-hybridizing colonies for sequence analysis .
- RNA s-ample selected for the presence of message to produce a cDNA, e.g., a monocyte or macrophage cell sample.
- the original isolate of IL-l ⁇ was from a whole mouse cDNA library, and for the IL-l ⁇ from a mouse placenta.
- Full length clones may be isolated by hybridization of cDNA libraries from appropriate tissues pre-selected by PCR signal .
- PCR primers are typically designed to contain at least 15 nucleotides, e.g., 15-30 nucleotides.
- the design of IL-l ⁇ or IL-l ⁇ specific primers containing 21 nucleotides, e.g., that code for IL-l ⁇ or IL-l ⁇ polypeptides containing at least 4 amino acids from the IL-l ⁇ or IL-l ⁇ domains are described as follows.
- Other PCR primers designed to amplify other IL- l ⁇ or IL-l ⁇ polypeptide fragments will be designed in a similar fashion, e.g., mutagenesis primers.
- nucleotides in such a primer encode conserved amino acids, e.g., amino residues of SEQ. ID NO: 2, 4, 6, including IL-l ⁇ or IL-l ⁇ -specific amino acids as described herein.
- primers containing at least 40% IL-l ⁇ or IL-l ⁇ -conserved amino acids can be used.
- Such a primer, containing 21 nucleotides can include sequences encoding at least 3/7, 4/7, 5/7, 6/7 or 7/7 IL-l ⁇ or IL-l ⁇ -conserved amino acids.
- the primers can be synthesized using, e.g., standard chemical methods. Due to the degeneracy of the genetic code and the bias of preferred species variants, such primers should be designed to include appropriate degenerate sequences, as can be readily determined using common knowledge. Based on the guidelines presented above, examples of IL-l ⁇ or IL-l ⁇ -conserved amino acid peptides that can be used as templates for the design of IL-l ⁇ or IL-l ⁇ specific primers are as follows .
- Primers can be designed to amplify various structural features or domains, for example, a 4-10 amino acid region of either IL-l ⁇ or IL-l ⁇ peptide that corresponds to any one of the
- primers 12 ⁇ strands could be amplified using this strategy.
- primer desired primers can be designed, for example, to correspond to 4-7 consecutive amino acids of any of the segments shown below. 1.
- LeuCysPheArgMetLysAsp corresponding to .amino acid residues 8 to 14 of murine IL-l ⁇ (see SEQ ID NO: 2) .
- PheTyrPheGlnGln (corresponding to .amino acid residues 150 to 154 of murine IL-l ⁇ (see SEQ ID NO: 2) .
- ValTrpIleLeuGlnAsn (corresponding to amino acid residues 24 to 29 of murine IL-l ⁇ (SEQ ID NO: 6) .
- SerCysLeuPheCysThrLys (corresponding to amino acid residues 74 to 80 of murine IL-l ⁇ (SEQ ID NO: 6) .
- ProValLeuGlnLeuGly (corresponding to amino acid residues 85 to 90 of murine IL-l ⁇ (SEQ ID NO: 6) .
- ThrThrSerThrPheGluSer (corresponding to amino acid residues 116 to 122 of murine IL-l ⁇ (SEQ ID NO: 6) .
- PhelleAlaValCys (corresponding to amino acid residues 129 to 133 of murine IL-l ⁇ (SEQ ID NO: 6) .
- CysProLeuIleLeuThr (corresponding to .amino acid residues 138 to 143 of murine IL-l ⁇ (SEQ ID NO: 6) .
- PheGluMetlleVal (corresponding to amino acid residues 154 to 158 of murine IL-l ⁇ (SEQ ID NO: 6) .
- IL-l ⁇ or IL-l ⁇ primers for example primers based on IL-l ⁇ or IL-l ⁇ specific peptides shown above, or portions thereof, can be used in PCR reactions to generate IL-l ⁇ or IL-l ⁇ , probes which can be used in standard screening methods to identify nucleic acids encoding IL-l ⁇ or IL-l ⁇ family members (see e.g.,
- IL-l ⁇ Message for the gene encoding IL-l ⁇ has been detected in a mouse cDNA library. Message for IL-l ⁇ has been detected in placenta tissue.
- DNA (5 ⁇ g) from a primary amplified cDNA library is digested with appropriate restriction enzymes to release the inserts, run on a 1% agarose gel and transferred to a nylon membrane (Schleicher and Schuell, Keene, NH) .
- Samples for human mRNA isolation could include: peripheral blood mononuclear cells (monocytes, T cells, NK cells, granulocytes , B cells), resting (T100); peripheral blood mononuclear cells, activated with anti- CD3 for 2, 6, 12 h pooled (T101) ; T cell, TH0 clone Mot 72, resting (T102); T cell, TH0 clone Mot 72, activated with anti-CD28 and anti-CD3 for 3, 6, 12 h pooled (T103); T cell, THO clone Mot 72, anergic treated with specific peptide for 2, 7, 12 h pooled PT104) ; T cell, TH1 clone HY06, resting (T107); T cell, THl clone HY06, activated with anti-CD28 and anti-CD3 for 3, 6, 12 h pooled (T108) ; T cell, THl clone HY06, anergic treated with
- CD2 ⁇ , IL-4, and anti IFN- ⁇ , TH2 polarized, activated with anti-CD3 and anti-CD2 ⁇ 4 h T116
- T cell tumor lines Jurkat and Hut7 ⁇ , resting T117
- IL-10 for 1, 2, 6, 12, 24 h pooled M103
- elutriated monocytes, activated with LPS, IFN ⁇ , anti-IL-10 for 4, 16 h pooled M106
- elutriated monocytes activated LPS for 1 h (MlO ⁇ )
- DC 70% CDla ⁇ from CD34+ GM-CSF, TNF ⁇ 12 days, activated with PMA and ionomycin for 1 hr (D102);
- DC 70% CDla+ from CD34+ GM-CSF, TNF ⁇ 12 days, activated with PMA and ionomycin for 6 hr (D
- RNAse protection assay Tissue distribution of transcripts derived from IL- l ⁇ and IL-l ⁇ were determined in experiments using an RNAse protection assay.
- Total RNA was prepared from adult brain, spleen, lung, liver and kidney by homogenization in guanidium thiocyantae and extraction with phenol, followed by centrifugation through 5.7 M cesium chloride (Sambrook, et al . , Molecular Cloning: A laboratory- Manual, Current ed. , Cold Spring Harbor Laboratory Press, Cold Spring Harbor, New York) .
- Riboprobes were synthesized using either T7 or T3 RNA polymerase on linerized templates that were cloned into pBluescript. Each mouse IL-l ⁇ and IL-l ⁇ probe contained 150-200 nucleotides from the antisense strand, linked to 25-50 nucleotides of vector sequence. Reagents were obtained from Ambion (Austin, Texas) following standard manufacturer's protocols.
- IL-l ⁇ and IL-l ⁇ Various strategies are used to obtain species counterparts of mouse IL-l ⁇ and IL-l ⁇ .
- One method is by cross hybridization using closely related species DNA probes .
- the degree of identity between mouse and human IL-1 counterparts typically is as high as 70%. It may be useful to go into evolutionarily similar species as intermediate steps.
- Another method is by using specific PCR primers based on the identification of blocks of similarity between human and mouse IL-1 counterparts, e.g., areas of highly conserved polypeptide sequence.
- the IL-l ⁇ , IL-l ⁇ , and IL-IRA genes cluster on the s.ame human chromosome.
- IL-l ⁇ The fourth known member of the IL-1 family, IL-l ⁇ , which is most closely related to IL-l ⁇ , has been mapped to a different human chromosome.
- IL-l ⁇ and IL-l ⁇ are potential candidates, and sequencing of the human IL-l ⁇ locus may well lead to identification of the novel IL-1 genes.
- An appropriate, e.g., GST, fusion construct is engineered for expression, e.g., in E. coli.
- a mouse IGIF pGex plasmid is constructed and transformed into E. coli.
- Freshly transformed cells are grown in LB medium containing 50 ⁇ g/ml ampicillin and induced with IPTG (Sigma, St. Louis, MO) . After overnight induction, the bacteria are harvested and the pellets containing IL-l ⁇ are isolated. The pellets are homogenized in TE buffer (50 mM Tris-base pH 8.0, 10 mM EDTA and 2 mM pefabloc) in 2 liters.
- TE buffer 50 mM Tris-base pH 8.0, 10 mM EDTA and 2 mM pefabloc
- This material is passed through a microfluidizer (Microfluidics, Newton, MA) three times.
- the fluidized supernatant is spun down on a Sorvall GS-3 rotor for 1 h at 13,000 rpm.
- the resulting supernatant containing the IL-l ⁇ is filtered and passed over a glutathione-SEPHAROSE column equilibrated in 50 mM Tris-base pH 8.0.
- the fractions containing the IL-l ⁇ -GST fusion protein are pooled and cleaved with thrombin (Enzyme Research Laboratories,
- Balb/c mice are immunized intraperitoneally with recombinant forms of the protein, e.g., purified soluble IL-l ⁇ - or IL-l ⁇ -FLAG or stable transfected NIH-3T3 cells. Animals are boosted at appropriate time points with protein, with or without additional adjuvant, to further stimulate antibody production. Serum is collected, or hybridomas produced with harvested spleens. Alternatively, Balb/c mice are immunized with cells transformed with the gene or fragments thereof, either endogenous or exogenous cells, or with isolated membranes enriched for expression of the antigen. Serum is collected at the appropriate time, typically after numerous further administrations. Various gene therapy techniques may be useful, e.g., in producing protein in situ, for generating an immune response.
- recombinant forms of the protein e.g., purified soluble IL-l ⁇ - or IL-l ⁇ -FLAG or stable transfected NIH-3T3 cells. Animals are boosted at
- Monoclonal antibodies may be made. For example, splenocytes are fused with an appropriate fusion partner and hybridomas are selected in growth medium by standard procedures . Hybridoma supernatants are screened for the presence of antibodies which bind to the desired IL-l ⁇ , e.g., by ELISA or other assay. Antibodies which specifically recognize IL-l ⁇ or IL-l ⁇ may also be selected or prepared.
- binding reagent is either labeled as described above, e.g., fluorescence or otherwise, or immobilized to a substrate for panning methods.
- Nucleic acids may also be introduced into cells in an animal to produce the antigen, which serves to elicit an immune response. See, e.g., Wang, et al . (1993) Proc. Nat ' 1. Acad. Sci.
- fusion proteins with IL-l ⁇ or IL-l ⁇ .
- This portion of the gene is fused to an epitope tag, e.g., a FLAG tag, or to a two hybrid system construct. See, e.g., Fields and Song (1989) Nature 340:245-246.
- the epitope tag may be used in an expression cloning procedure with detection with anti-FLAG antibodies to detect a binding partner, e.g., receptor for the respective IL-1.
- the two hybrid system may also be used to isolate proteins which specifically bind to IL-l ⁇ or IL-l ⁇ .
- Chromosome spreads were prepared. In situ hybridization was performed on chromosome preparations obtained from phytohemagglutinin-stimulated lymphocytes cultured for 72 h. 5-bromodeoxyuridine was added for the final seven hours of culture (60 ⁇ g/ml of medium), to ensure a posthybridization chromosomal banding of good quality.
- An appropriate fragment e.g., a PCR fragment
- the vector was labeled by nick-translation with - .
- the radiolabeled probe was hybridized to metaphase spreads as described in Mattei, et al . (1985) Hum. Genet. 69:327- 331.
- Standard mutagenesis analysis is performed, e.g., by generating many different variants at determined positions, e.g., at the positions identified above, and evaluating biological activities of the variants. This may be performed to the extent of determining positions which modify activity, or to focus on specific positions to determine the residues which can be substituted to either retain, block, or modulate biological activity.
- analysis of natural variants can indicate what positions tolerate natural mutations. This may result from populational analysis of variation among individuals, or across strains or species. Samples from selected individuals are analyzed, e.g., by PCR analysis and sequencing. This allows evaluation of population polymorphisms .
- An IL-l ⁇ can be used as a specific binding reagent to identify its binding partner, by taking advantage of its specificity of binding, much like an antibody would be used.
- a binding reagent is either labeled as described above, e.g., fluorescence or otherwise, or immobilized to a substrate for panning methods.
- the binding composition is used to screen an expression library made from a cell line which expresses a binding partner, i.e. receptor.
- Standard staining techniques are used to detect or sort intracellular or surface expressed receptor, or surface expressing transformed cells are screened by panning. Screening of intracellular expression is performed by various staining or immunofluorescence procedures. See also McMahan, et al. (1991) EMBO J. 10:2821-2832.
- a positive control is prepared, e.g., of IL-l ⁇ -FLAG cDNA at 1 and
- HBSS HBSS.
- the slides may be stored at -80° C after all liquid is removed.
- 0.5 ml incubations are performed as follows. Add HBSS/saponin (0.1%) with 32 ⁇ l/ml of 1 M NaN 3 for 20 min. Cells are then washed with HBSS/saponin IX. Add appropriate IL-l ⁇ or IL- l ⁇ /antibody complex to cells and incubate for 30 min. Wash cells twice with HBSS/saponin. If appropriate, add first antibody for 30 min. Add second antibody, e.g., Vector anti-mouse antibody, at 1/200 dilution, and incubate for 30 min.
- second antibody e.g., Vector anti-mouse antibody
- ELISA solution e.g., Vector Elite ABC horseradish peroxidase solution, and preincubate for 30 min.
- Use e.g., 1 drop of solution A (avidin) and 1 drop solution B (biotin) per 2.5 ml HBSS/saponin. Wash cells twice with HBSS/saponin.
- IL-l ⁇ reagents are used to affinity purify or sort out cells expressing a receptor. See, e.g., Sambrook, et al . or Ausubel, et al .
- Another strategy is to screen for a membrane bound receptor by panning.
- the receptor cDNA is constructed as described above.
- the ligand can be immobilized and used to immobilize expressing cells. Immobilization may be achieved by use of appropriate antibodies which recognize, e.g., a FLAG sequence of a IL-l ⁇ fusion construct, or by use of antibodies raised against the first antibodies. Recursive cycles of selection and amplification lead to enrichment of appropriate clones and eventual isolation of receptor expressing clones.
- Phage expression libraries can be screened by mammalian IL-l ⁇ . Appropriate label techniques, e.g., anti-FLAG antibodies, will allow specific labeling of appropriate clones .
- SEQ ID NO: 1 provides rodent IL-l ⁇ nucleotide secjuence .
- SEQ ID NO: 2 provides rodent IL-l ⁇ polypeptide sequence
- SEQ ID NO: 3 provides partial rodent IL-l ⁇ nucleotide sequence
- SEQ ID NO: 4 provides partial rodent IL-l ⁇ polypeptide sequence
- SEQ ID NO: 5 provides full length rodent IL-l ⁇ nucleic acid sequence
- SEQ ID NO: 6 provides full length rodent IL-l ⁇ polypeptide sequence
- SEQ ID NO: 7 provides human IL-IRA precursor polypeptide sequence.
- S SEEQQ I IDD N NOO:: 8 8 provides human IL-l ⁇ (IGIF) precursor polypeptide sequence.
- SEQ ID NO: 9 provides mouse IL-l ⁇ (IGIF) precursor polypeptide sequence.
- SEQ ID NO: 11 provides human IL-l ⁇ precursor polypeptide sequence.
- SEQ ID NO: 1l1 provides human IL-l ⁇ precursor polypeptide sequence.
- GCT CCC ATC ACA GAC TTC TAC TTT CAG CAG TGT GAC TA 470
- MOLECULE TYPE protein
Landscapes
- Chemical & Material Sciences (AREA)
- Health & Medical Sciences (AREA)
- Life Sciences & Earth Sciences (AREA)
- Organic Chemistry (AREA)
- General Health & Medical Sciences (AREA)
- Molecular Biology (AREA)
- Biochemistry (AREA)
- Biophysics (AREA)
- Zoology (AREA)
- Genetics & Genomics (AREA)
- Medicinal Chemistry (AREA)
- Gastroenterology & Hepatology (AREA)
- Proteomics, Peptides & Aminoacids (AREA)
- Toxicology (AREA)
- Peptides Or Proteins (AREA)
- Apparatus Associated With Microorganisms And Enzymes (AREA)
- Preparation Of Compounds By Using Micro-Organisms (AREA)
- Micro-Organisms Or Cultivation Processes Thereof (AREA)
Abstract
Description
Claims
Priority Applications (4)
Application Number | Priority Date | Filing Date | Title |
---|---|---|---|
AU71031/98A AU7103198A (en) | 1997-04-21 | 1998-04-17 | Mammalian cytokines; related reagents and methods |
CA002287254A CA2287254A1 (en) | 1997-04-21 | 1998-04-17 | Mammalian cytokines; related reagents and methods |
JP54608798A JP2002501496A (en) | 1997-04-21 | 1998-04-17 | Mammalian cytokines; related reagents and methods |
EP98918031A EP0977779A1 (en) | 1997-04-21 | 1998-04-17 | Mammalian cytokines; related reagents and methods |
Applications Claiming Priority (4)
Application Number | Priority Date | Filing Date | Title |
---|---|---|---|
US83762797A | 1997-04-21 | 1997-04-21 | |
US08/837,627 | 1997-04-21 | ||
US5511197P | 1997-08-06 | 1997-08-06 | |
US60/055,111 | 1997-08-06 |
Publications (2)
Publication Number | Publication Date |
---|---|
WO1998047921A1 WO1998047921A1 (en) | 1998-10-29 |
WO1998047921A9 true WO1998047921A9 (en) | 1999-04-22 |
Family
ID=26733854
Family Applications (1)
Application Number | Title | Priority Date | Filing Date |
---|---|---|---|
PCT/US1998/006879 WO1998047921A1 (en) | 1997-04-21 | 1998-04-17 | Mammalian cytokines; related reagents and methods |
Country Status (5)
Country | Link |
---|---|
EP (1) | EP0977779A1 (en) |
JP (1) | JP2002501496A (en) |
AU (1) | AU7103198A (en) |
CA (1) | CA2287254A1 (en) |
WO (1) | WO1998047921A1 (en) |
Families Citing this family (37)
Publication number | Priority date | Publication date | Assignee | Title |
---|---|---|---|---|
US6197551B1 (en) * | 1998-01-27 | 2001-03-06 | Millennium Pharmaceuticals, Inc. | Spoil-1 protein and nucleic acid molecules and uses therefor |
EP1044271A1 (en) | 1998-01-09 | 2000-10-18 | Immunex Corporation | Il-1 delta dna and polypeptides |
US6962904B1 (en) | 1998-03-13 | 2005-11-08 | Connective Tissue Imagineering | Elastin peptide analogs and uses thereof |
US6294655B1 (en) | 1998-04-03 | 2001-09-25 | Hyseq, Inc. | Anti-interleukin-1 receptor antagonist antibodies and uses thereof |
US6426191B1 (en) * | 1998-04-03 | 2002-07-30 | Hyseq, Inc. | Assays involving an IL-1 receptor antagonist |
US6337072B1 (en) | 1998-04-03 | 2002-01-08 | Hyseq, Inc. | Interleukin-1 receptor antagonist and recombinant production thereof |
US6541623B1 (en) * | 1998-04-03 | 2003-04-01 | Hyseq, Inc. | Interleukin—1 receptor antagonist and uses thereof |
CA2326066A1 (en) * | 1998-04-03 | 1999-10-14 | Hyseq, Inc. | A interleukin-1 receptor antagonist and uses thereof |
AU5772299A (en) * | 1998-08-07 | 2000-02-28 | Millennium Pharmaceuticals, Inc. | Novel molecules of the tango-93-related protein family and uses thereof |
US6949359B1 (en) | 1998-08-21 | 2005-09-27 | Immunex Corporation | Human IL-1 epsilon DNA and polypeptides |
US6838290B2 (en) | 1998-08-21 | 2005-01-04 | Immunex Corporation | Methods for screening compounds that affect IL-1 epsilon activity |
CA2340790C (en) | 1998-08-21 | 2012-07-10 | Immunex Corporation | Human il-1 epsilon dna and polypeptides |
AU6296899A (en) * | 1998-10-08 | 2000-04-26 | Zymogenetics Inc. | Interleukin-1 homolog |
EP2330198A1 (en) * | 1998-12-23 | 2011-06-08 | Genentech, Inc. | IL-1 related polypeptides |
US7074556B2 (en) | 1999-03-02 | 2006-07-11 | Invitrogen Corporation | cDNA synthesis improvements |
ES2300266T3 (en) | 1999-05-20 | 2008-06-16 | Nuvelo, Inc. | INTERLEUQUINA-1 HY2, MATERIALS AND METHODS. |
US6339141B1 (en) | 1999-05-20 | 2002-01-15 | Hycey Inc. | Interleukin-1 Hy2 materials and methods |
WO2000071583A1 (en) * | 1999-05-24 | 2000-11-30 | Smithkline Beecham Corporation | Murine interleukin-1 homologue, il-1h3 |
EP1190097A2 (en) | 1999-06-22 | 2002-03-27 | Invitrogen Corporation | Improved primers and methods for the detection and discrimination of nucleic acids |
US6830902B1 (en) | 1999-07-02 | 2004-12-14 | Invitrogen Corporation | Compositions and methods for enhanced sensitivity and specificity of nucleic acid synthesis |
AU6219400A (en) * | 1999-07-16 | 2001-02-05 | Interleukin Genetics, Inc. | The il-1l1 gene and polypeptide products |
CA2393532A1 (en) * | 1999-12-10 | 2001-06-14 | Frank J. Calzone | Interleukin-1 receptor antagonist-like molecules and uses thereof |
US6794362B1 (en) * | 2000-05-30 | 2004-09-21 | Connective Tissue Imagineering Llc | Asparagine containing elastin peptide analogs |
NZ523403A (en) * | 2000-05-30 | 2005-04-29 | Connective Tissue Imagineering | Compounds that increase tissue elasticity and turgor which are analogous to portions of elastin |
US20030165859A1 (en) | 2001-10-23 | 2003-09-04 | Invitrogen Corporation | Primers and methods for the detection and discrimination of nucleic acids |
HUE029021T2 (en) * | 2005-06-21 | 2017-02-28 | Xoma (Us) Llc | IL-1beta binding antibodies and fragments thereof |
KR102080704B1 (en) * | 2010-07-29 | 2020-02-24 | 세센 바이오, 아이엔씨. | Chimeric IL-1 Receptor Type 1 Agonists and Antagonists |
EP2606150B1 (en) | 2010-08-20 | 2016-03-16 | Life Technologies Corporation | Quantitative real-time pcr assay using fret dual-labeled primers |
WO2012103240A2 (en) * | 2011-01-25 | 2012-08-02 | Eleven Biotherapeutics, Inc. | Receptor binding agents |
DK2914743T3 (en) | 2012-11-02 | 2019-10-14 | Life Technologies Corp | EXTRACTION, DETECTION AND QUANTIFICATION OF LITTLE RNA |
WO2014165210A2 (en) | 2013-03-12 | 2014-10-09 | Life Technologies Corporation | Universal reporter-based genotyping methods and materials |
JP6450364B2 (en) | 2013-03-13 | 2019-01-09 | セセン バイオ, インコーポレイテッド | Chimeric cytokine formulations for ocular delivery |
GB201511792D0 (en) * | 2015-07-06 | 2015-08-19 | Immatics Biotechnologies Gmbh | Novel peptides and combination of peptides for use in immunotherapy against esopageal cancer and other cancers |
PE20180259A1 (en) | 2015-07-06 | 2018-02-05 | Immatics Biotechnologies Gmbh | NEW PEPTIDES AND COMBINATION OF PEPTIDES FOR USE IN IMMUNOTHERAPY AGAINST ESOPHAGEAL AND OTHER CANCERS |
WO2017218938A1 (en) | 2016-06-16 | 2017-12-21 | Life Technologies Corporation | Novel compositions, methods and kits for microorganism detection |
EP3504342A1 (en) | 2016-08-26 | 2019-07-03 | Life Technologies Corporation | Nucleic acid extraction and amplification controls and methods of use thereof |
KR20200081476A (en) | 2017-11-13 | 2020-07-07 | 라이프 테크놀로지스 코포레이션 | Compositions, methods and kits for urinary tract microbial detection |
Family Cites Families (1)
Publication number | Priority date | Publication date | Assignee | Title |
---|---|---|---|---|
KR0148009B1 (en) * | 1988-05-27 | 1998-08-01 | 그래고리 비. 아보트 | Interleukin-1 inhibitors |
-
1998
- 1998-04-17 CA CA002287254A patent/CA2287254A1/en not_active Abandoned
- 1998-04-17 AU AU71031/98A patent/AU7103198A/en not_active Abandoned
- 1998-04-17 EP EP98918031A patent/EP0977779A1/en not_active Withdrawn
- 1998-04-17 JP JP54608798A patent/JP2002501496A/en active Pending
- 1998-04-17 WO PCT/US1998/006879 patent/WO1998047921A1/en not_active Application Discontinuation
Also Published As
Publication number | Publication date |
---|---|
JP2002501496A (en) | 2002-01-15 |
AU7103198A (en) | 1998-11-13 |
EP0977779A1 (en) | 2000-02-09 |
WO1998047921A1 (en) | 1998-10-29 |
CA2287254A1 (en) | 1998-10-29 |
Similar Documents
Publication | Publication Date | Title |
---|---|---|
WO1998047921A9 (en) | Mammalian cytokines; related reagents and methods | |
US7393530B2 (en) | IL-1-like cytokine antibodies | |
US20020173623A1 (en) | Mammalian cytokines; receptors; related reagents and methods | |
WO1999040195A1 (en) | Mammalian receptor proteins; related reagents and methods | |
CA2392109A1 (en) | Mammalian receptor proteins; related reagents and methods | |
CA2374391C (en) | Mammalian receptor proteins; related reagents and methods | |
CA2323236A1 (en) | Human receptor proteins; related reagents and methods | |
US7812126B2 (en) | DIRS1 polypeptides | |
US6326472B1 (en) | Human receptor proteins; related reagents and methods | |
US6843987B2 (en) | Mammalian cytokines; receptors; related reagents and methods | |
US20030082734A1 (en) | Mammalian receptor proteins; related reagents and methods | |
CA2255906A1 (en) | Human interleukin-1j and antagonists thereof | |
WO2000017363A2 (en) | Interleukin-1 like molecule: interleukin-1-zeta; nucleic acids, polypeptides, antibodies, their uses | |
US7205381B2 (en) | Mammalian cytokines; related reagents and methods | |
EP1029043A1 (en) | Human receptor proteins; related reagents and methods | |
MXPA99009646A (en) | Mammalian cytokines;related reagents and methods |
Legal Events
Date | Code | Title | Description |
---|---|---|---|
AK | Designated states |
Kind code of ref document: A1 Designated state(s): AL AM AU AZ BA BB BG BR BY CA CN CZ EE GE GW HU ID IL IS JP KG KR KZ LC LK LR LT LV MD MG MK MN MX NO NZ PL RO RU SG SI SK SL TJ TM TR TT UA UZ VN YU |
|
AL | Designated countries for regional patents |
Kind code of ref document: A1 Designated state(s): GH GM KE LS MW SD SZ UG ZW AM AZ BY KG KZ MD RU TJ TM AT BE CH CY DE DK ES FI FR GB GR IE IT LU MC NL PT SE BF BJ CF CG CI CM GA GN ML MR NE SN TD TG |
|
DFPE | Request for preliminary examination filed prior to expiration of 19th month from priority date (pct application filed before 20040101) | ||
121 | Ep: the epo has been informed by wipo that ep was designated in this application | ||
AK | Designated states |
Kind code of ref document: C2 Designated state(s): AL AM AU AZ BA BB BG BR BY CA CN CZ EE GE GW HU ID IL IS JP KG KR KZ LC LK LR LT LV MD MG MK MN MX NO NZ PL RO RU SG SI SK SL TJ TM TR TT UA UZ VN YU |
|
AL | Designated countries for regional patents |
Kind code of ref document: C2 Designated state(s): GH GM KE LS MW SD SZ UG ZW AM AZ BY KG KZ MD RU TJ TM AT BE CH CY DE DK ES FI FR GB GR IE IT LU MC NL PT SE BF BJ CF CG CI CM GA GN ML MR NE SN TD TG |
|
COP | Corrected version of pamphlet |
Free format text: PAGE 1/1, DRAWINGS, REPLACED BY A NEW PAGE 1/1; DUE TO LATE TRANSMITTAL BY THE RECEIVING OFFICE |
|
WWE | Wipo information: entry into national phase |
Ref document number: 1998918031 Country of ref document: EP |
|
ENP | Entry into the national phase in: |
Ref country code: JP Ref document number: 1998 546087 Kind code of ref document: A Format of ref document f/p: F |
|
ENP | Entry into the national phase in: |
Ref country code: CA Ref document number: 2287254 Kind code of ref document: A Format of ref document f/p: F Ref document number: 2287254 Country of ref document: CA |
|
WWE | Wipo information: entry into national phase |
Ref document number: PA/a/1999/009646 Country of ref document: MX |
|
WWP | Wipo information: published in national office |
Ref document number: 1998918031 Country of ref document: EP |
|
WWW | Wipo information: withdrawn in national office |
Ref document number: 1998918031 Country of ref document: EP |