EP0579739A1 - Interleukin-8-rezeptoren, damit in zusammenhang stehende moleküle und verfahrensweisen - Google Patents

Interleukin-8-rezeptoren, damit in zusammenhang stehende moleküle und verfahrensweisen

Info

Publication number
EP0579739A1
EP0579739A1 EP92910198A EP92910198A EP0579739A1 EP 0579739 A1 EP0579739 A1 EP 0579739A1 EP 92910198 A EP92910198 A EP 92910198A EP 92910198 A EP92910198 A EP 92910198A EP 0579739 A1 EP0579739 A1 EP 0579739A1
Authority
EP
European Patent Office
Prior art keywords
receptor
polypeptide
leu
ctg
binding
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Withdrawn
Application number
EP92910198A
Other languages
English (en)
French (fr)
Other versions
EP0579739A4 (de
Inventor
Javier Navarro
Kathleen M. Thomas
Daniel P. Witt
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Boston University
Repligen Corp
Original Assignee
Boston University
Repligen Corp
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Boston University, Repligen Corp filed Critical Boston University
Publication of EP0579739A1 publication Critical patent/EP0579739A1/de
Publication of EP0579739A4 publication Critical patent/EP0579739A4/de
Withdrawn legal-status Critical Current

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/705Receptors; Cell surface antigens; Cell surface determinants
    • C07K14/715Receptors; Cell surface antigens; Cell surface determinants for cytokines; for lymphokines; for interferons
    • C07K14/7155Receptors; Cell surface antigens; Cell surface determinants for cytokines; for lymphokines; for interferons for interleukins [IL]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P29/00Non-central analgesic, antipyretic or antiinflammatory agents, e.g. antirheumatic agents; Non-steroidal antiinflammatory drugs [NSAID]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides

Definitions

  • This invention relates to reducing inflammation.
  • an orderly progression of host defenses involving, e.g., T and B lymphocytes, macrophages, and neutrophils
  • a well-controlled immune and inflammatory response that protects the host from offending antigens.
  • regulatory dysfunction of any of the systems involved in the host defense can damage host tissue and produce clinically apparent disease.
  • inflammation a pathologic process consisting of a complex set of cytologic and histologic reactions. These reactions occur in the affected blood vessels and adjacent tissues in response to an injury or abnormal stimulation caused by a physical, chemical, or biological agent.
  • Inflammatory disorders include anaphylaxis, systemic necrotizing vasculitis, systemic lupus erythematosus, serum sickness syndromes, psoriasis, and rheumatoid arthritis, and reperfusion injury occurring following periods of ischemia, such as in myocardial infarction or shock. Inflammation may also play a role in ho ograft rejection.
  • Neutrophils are cellular components of the blood which play a role in the inflammatory process. When activated (e.g., following infection of the host by a pathogen) , neutrophils produce substances that are cytotoxic and amplify the inflammatory response. During intense inflammation, release of neutrophil proteolytic enzymes and oxygen free radicals may cause digestion of cartilage mucopolysaccharide, oxidation of synovial tissue, and widespread damage to the lungs. In addition, chemotactic factors at the site of inflammation induce neutrophil aggregation and adherence to endothelium, causing, e.g., leukostasis in the pulmonary vasculature and cardiopulmonary dysfunction (Jandl, Blood, Little, Brown & Co., Boston, 1987).
  • Interleukin-8 is a 72 amino acid peptide which is produced by a variety of cell types upon activation with interleukin-1 and other stimulatory cytokines (Westwick et al. , Immunology Today 10.:146, 1988) .
  • IL-8 has previously been known as neutrophil activating peptide-1 (NAP-1) , neutrophil activating factor (NAF) , and monocyte-derived neutrophil chemotactic f ctor (MDNCF) .
  • NAP-1 neutrophil activating peptide-1
  • NAF neutrophil activating factor
  • MDNCF monocyte-derived neutrophil chemotactic f ctor
  • IL-8 promotes che otaxis and degranulation of neutrophils (Djeu et al., J. Immunol . 144:2205. 1990).
  • IL-8 has been shown to be a potent chemoattractant for neutrophils in vitro and capable of producing a strong inflammatory effect in vivo (Colditz et al.. Am. J. Pathol . 134:755. 1989).
  • IL- 8 has been found to be present in significant quantities in naturally occurring inflammatory conditions such as psoriasis and rheumatoid arthritis. It is likely that IL-8 is a central factor in neutrophil-mediated inflammatory processes. For this reason, inhibitors or antagonists of IL-8 action can be expected to be useful anti-inflammatory agents.
  • IL-8 action on neutrophils is mediated by a specific receptor (Grob et al. , J " . Biol . Chem . 265:8311, 1990) .
  • This glycoprotein has been estimated to be of molecular mass 58,000 Daltons and is limited to granulocytic cells, especially neutrophils.
  • This receptor which has hitherto not been fully characterized or cloned, can be expected to be of particular utility in the development of IL-8 inhibitors and antagonists.
  • the invention features recombinant IL-
  • the receptor polypeptide may bind IL-8 with high affinity or with low affinity.
  • the receptor includes an amino acid sequence substantially identical to the amino acid sequence shown in Fig. 1 (SEQ ID NO: 1) , Fig. 2 (SEQ ID NO: 5) , or Fig.
  • the invention also features a substantially isolated polypeptide which is a fragment or analog of an IL-8 receptor and which includes a domain capable of binding IL-8.
  • the receptor is derived from a mammal, preferably, a human or a rabbit.
  • the invention further features a polypeptide including all or an IL-8-binding portion of an IL-8 receptor transmembrane domain or an IL-8 extracellular domain.
  • the polypeptide includes approximately amino acids 1-37 of the amino acid sequence shown in Fig. l (SEQ ID NO.:l) or an IL-8-binding fragment thereof; or approximately amino acids 1-50 of the amino acid sequence shown in Fig. 2 (SEQ ID NO.:5) or an IL-8-binding fragment thereof.
  • the polypeptide is a recombinant polypeptide or a synthetic polypeptide.
  • IL-8 receptor polypeptide is meant all or part of a cell surface protein which specifically binds IL-8 and signals the appropriate IL-8-mediated cascade of biological events; it includes receptors which bind IL-8 with either high or low affinity.
  • polypeptide is meant any chain of amino acids, regardless of length or post-translational modification (e.g., glycosylation) .
  • high affinity is meant having a K d which is lOnM or less (and, preferably, having a K d which is between 0.1 and lOnM) .
  • low affinity is meant having a K d which is greater than lOnM.
  • a “substantially isolated polypeptide” is one which is substantially free of other proteins, carbohydrates and lipids with which it is naturally associated.
  • a “substantially identical" amino acid sequence is meant an amino acid sequence which differs only by conservative amino acid substitutions, for example, substitution of one amino acid for another of the same class (e.g., valine for glycine, arginine for lysine, etc.) or by one or more non-conservative amino acid substitutions, deletions, or insertions located at positions of the amino acid sequence which do not destroy the biological activity of the receptor.
  • Such equivalent receptors can be isolated by extraction from the tissues or cells of any animal which naturally produce such a receptor or which can be induced to do so, using the methods described below, or their equivalent; or can be isolated by chemical synthesis; or can be isolated by standard techniques of recombinant DNA technology, e.g., by isolation of cDNA or genomic DNA encoding such a receptor.
  • derived from is meant encoded by the genome of that organism and present on the surface of a subset of that organism's cells.
  • synthetic peptide is meant one which is produced by chemical, e.g., peptide synthesis.
  • the invention features purified DNA which encodes a receptor (or receptor fragment or analog thereof) described above.
  • the purified DNA may encode a high affinity IL-8 receptor or it may encode a low affinity IL-8 receptor.
  • the purified DNA is cDNA; is cDNA which encodes a rabbit IL-8 receptor; is cDNA which encodes a human IL-8 receptor; is included in the plas id F3R; is included in the plasmid 5bla; is included in the plasmid 4AB.
  • purified DNA is meant a DNA molecule which encodes an IL-8 receptor (or an appropriate receptor fragment or analog) , but which is free of the genes that, in the naturally-occurring genome of the organism from which the DNA of the invention is derived, flank the gene encoding the IL-8 receptor.
  • the invention features vectors which contain such purified DNA and are capable of directing expression of the protein encoded by the DNA in a vector-containing cell; and cells containing such vectors (preferably eukaryotic cells, e.g., mammalian cells, e.g., myeloma cells or hamster lung fibroblast cells) .
  • cells containing such vectors preferably eukaryotic cells, e.g., mammalian cells, e.g., myeloma cells or hamster lung fibroblast cells.
  • eukaryotic cells e.g., mammalian cells, e.g., myeloma cells or hamster lung fibroblast cells
  • such cells are stably transfected with such purified DNA.
  • the expression vectors or vector-containing cells of the invention can be used in a method of the invention to produce recombinant IL-8 receptor polypeptide and the polypeptides described above.
  • the method involves providing a cell transformed with DNA encoding an IL-8 receptor or a fragment or analog thereof positioned for expression in the cell; culturing the transformed cell under conditions for expressing the DNA; and isolating the recombinant IL-8 receptor protein.
  • transformed cell is meant a cell into which (or into an ancestor of which) has been introduced, by means of genetic engineering, a DNA molecule encoding an IL-8 receptor (or a fragment or analog, thereof) .
  • Such a DNA molecule is "positioned for expression” meaning that the DNA molecule is positioned adjacent to a DNA sequence which directs transcription and translation of the sequence (i.e., facilitates the production of the IL-8 receptor protein, or fragment or analog, thereof) .
  • the invention features a purified antibody which binds preferentially to an IL-8 receptor (or a fragment or analog thereof) .
  • purified antibody is meant one which is sufficiently free of other proteins, carbohydrates, and lipids with which it is naturally associated to permit therapeutic administration.
  • Such an antibody "preferentially binds" to an IL-8 receptor (or fragment or analog, thereof) , i.e., does not substantially recognize and bind to other antigenically-unrelated molecules.
  • the antibody neutralizes the biological activity in vivo of the protein to which it binds.
  • biological activity is meant the ability of the IL-8 receptor to bind IL-8 and signal the appropriate cascade of biological events.
  • neutralize is meant to partially or completely block (e.g., the biological activity of an IL-8 receptor) .
  • the polypeptides or antibodies described above are used as the active ingredient of therapeutic compositions.
  • the active ingredient may be formulated with a physiologically-acceptable carrier or anchored within the membrane of a cell. These therapeutic compositions are used in a method of reducing inflammation.
  • the invention features a method of screening candidate compounds for their ability to antagonize interaction between IL-8 and an IL-8 receptor.
  • the method involves: a) mixing a candidate antagonist compound with a first compound which includes a recombinant IL-8 receptor (or IL-8-binding fragment or analog) on the one hand and with a second compound which includes IL-8 on the other hand; b) determining whether the first and second compounds bind; and c) identifying antagonistic compounds as those which interfere with the binding of the first compound to the second compound and/or which reduce the IL-8-mediated release of intracellular Ca ++ .
  • an “antagonist” is meant a molecule which inhibits a particular activity, in this case, the ability of IL-8 to interact with an IL-8 receptor and/or to trigger the biological events resulting from such an interaction (e.g., release of intracellular Ca ++ ) .
  • the invention features chimeric polypeptides, in particular, the chimeric polypeptides include an amino-terminal portion of the sequence shown in Fig. 1 (SEQ ID NO.:l) fused to a carboxy-terminal portion of the sequence shown in Fig. 2 (SEQ ID NO.:5).
  • the polypeptide includes approximately amino acids 1-58 of Fig. 1 (SEQ ID NO.:l) or an IL-8-binding fragment thereof fused to approximately amino acids 63- 360 of Fig. 2 (SEQ ID NO.:5) and is encoded by F3R/4AB.
  • the invention also features polypeptides which include an amino-terminal portion of the sequence shown in Fig.
  • the polypeptide includes approximately amino acids 1-62 of Fig. 2 (SEQ ID NO.:5) or an IL-8-binding fragment thereof fused to approximately amino acids 59-355 of Fig. 1 (SEQ ID NO.:l) and is encoded by 4AB/F3R.
  • the invention also features DNA encoding such chimeric polypeptides.
  • the proteins of the invention are involved in the events leading to neutrophil activation and the inflammatory response. Such proteins are therefore useful to treat or, alternatively, to develop therapeutics to treat inflammation.
  • Particular disorders which may be treated using the proteins and/or the methods of the invention include psoriasis, rheumatoid arthritis, vasculitis, as well as reperfusion injury, or any neutrophil-mediated inflammatory disorder.
  • Preferred therapeutics include antagonists, e.g., peptide fragments (particularly, fragments derived from the N-terminal extracellular domain) , antibodies (particularly, antibodies which recognize and bind the N-terminal extracellular domain) , or drugs, which block IL-8 or IL- 8 receptor function by interfering with the interleukin: receptor interaction.
  • the instant invention provides a simple and rapid approach to the identification of useful therapeutics. Such an approach was previously difficult for several reasons: (1) because the interaction between IL-8 and its endogenous receptor on the surface of a neutrophil triggers a series of events leading to the release of proteolytic enzymes and oxygen free radicals, and the resultant destruction of the receptor-bearing neutrophil cell; and (2) because of the presence on the surface of neutrophils of related receptors.
  • Isolation of the IL-8 receptor gene allows its expression in a cell type remote from neutrophils (e.g., J558, SP2 myeloma cells, COS cells, or Chinese hamster lung fibroblast cells) , effectively uncoupling the IL-8 receptor from its normal cytotoxic signaling pathway and providing a system for assaying an IL-8:receptor interaction without associated cell death.
  • a cell type remote from neutrophils e.g., J558, SP2 myeloma cells, COS cells, or Chinese hamster lung fibroblast cells
  • a peptide- or antibody-based therapeutic may be produced, in large quantity and inexpensively, using recombinant and molecular biological techniques.
  • Fig. 1 shows the nucleic acid sequence and deduced amino acid sequence of a high affinity IL-8 receptor derived from a rabbit source.
  • Fig. 2 shows the nucleic acid sequence and deduced amino acid sequence of a low affinity IL-8 receptor derived from a human source.
  • Fig. 3 is a series of bar graphs which represent the extent of IL-8 binding to four independently-isolated cell lines which inducibly express a high affinity IL-8 receptor.
  • Fig. 4 is a graph showing IL-8 binding to a low affinity IL-8 receptor as a function of IL-8 concentration.
  • Fig. 5 is a graph showing MGSA/GRO ⁇ binding to a low affinity IL-8 receptor as a function of MGSA/GRO ⁇ concentration and competition by MGSA/GRO ⁇ with IL-8 for IL-8 receptor binding.
  • Fig. 6 is a graph showing competition by various ligands for binding to a low affinity IL-8 receptor.
  • Fig. 7 is a series of bar graphs which represent the extent of IL-8 binding to high affinity/low affinity and low affinity/high affinity chimeric receptors.
  • Fig. 8 is a graph showing competition by various ligands for binding to a low affinity IL-8 receptor and a high affinity/low affinity chimeric IL-8 receptor.
  • Fig. 9 shows the nucleic acid sequence and deduced amino acid sequence of a low affinity IL-8 receptor derived from a rabbit source.
  • Fig. 10 is a schematic drawing illustrating the structure of the IL-8 receptors.
  • Fig. 11 is a series of two graphs showing the percentage of total IL-8 binding to a high affinity IL-8 receptor as a function of agonist concentration.
  • Polypeptides according to the invention include the entire high affinity IL-8 receptor (as described in Fig. 1, SEQ ID NO: 1) and the entire low affinity IL-8 receptor (as described in Fig. 2, SEQ ID NO: 5 and Fig. 9, SEQ ID NO:6); high affinity receptors bind IL-8 with a K d of lOnM or less (and, preferably, with a K d of between 0.1 and lOnM) , and low affinity receptors bind IL-8 with a K d of greater than lOnM.
  • Such receptors may be derived from any source, but are preferably derived from a mammal, e.g., a human or a rabbit.
  • polypeptides are used, e.g., to screen for antagonists which disrupt an IL-8:receptor interaction (see below).
  • Polypeptides of the invention also include any analog or fragment of the high affinity or low affinity IL-8 receptors capable of interacting with IL-8 (e.g., those derived from the IL-8 receptor N-terminal extracellular domain) . Such analogs and fragments may also be used to screen for IL- 8 receptor antagonists.
  • that subset of receptor fragments or analogs which bind IL-8 and are, preferably, soluble (or insoluble and formulated in a lipid vesicle) may be used as antagonists to reduce inflammatory diseases (see below) .
  • the efficacy of a receptor analog or fragment is dependent upon its ability to interact with IL-8; such an interaction may be readily assayed using any of a number of standard in vitro binding methods and IL-8 receptor functional assays (e.g. , those described below) .
  • Specific receptor analogs of interest include full-length or partial (see below) receptor proteins including an amino acid sequence which differs only by conservative amino acid substitutions, for example, substitution of one amino acid for another of the same class (e.g., valine for glycine, arginine for lysine, etc.) or by one or more non-conservative amino acid substitutions, deletions, or insertions located at positions of the amino acid sequence which do not destroy the receptors 1 ability to bind IL-8 (as assayed below).
  • Specific receptor fragments of interest include any portions of the IL-8 receptor which are capable of interaction with IL-8, for example, all or part of the N- terminal extracellular domain.
  • Such portions include transmembrane segments 1-7 and portions of the receptor deduced to be extracellular (Fig. 10) .
  • Such fragments may be useful as antagonists (as described above) , and are also useful as immunogens for producing antibodies which neutralize the activity of the IL-8 receptor in vivo (e.g., by interfering with the interaction between the receptor and IL-8; see below) .
  • Extracellular regions may be identified by comparison with related proteins of similar structure (e.g., other members of the G-protein- coupled receptor superfamily) ; useful regions are those exhibiting homology to the extracellular domains of well- characterized members of the family.
  • the secondary protein structure and, therefore, the extracellular domain regions may be deduced semi- empirically using a hydrophobicity/hydrophilicity calculation such as the Chou-Fasman method (see, e.g., Chou and Fasman, Ann . Rev. Biochem. 42:251, 1978).
  • Hydrophilic domains particularly ones surrounded by hydrophobic stretches (e.g., transmembrane domains) present themselves as strong candidates for extracellular domains.
  • extracellular domains may be identified experimentally using standard enzymatic digest analysis, e.g., tryptic digest analysis.
  • Candidate fragments are tested for interaction with IL-8 by the assays described herein (e.g., the assay described above). Such fragments are also tested for their ability to antagonize the interaction between IL-8 and its endogenous receptor using the assays described herein. Analogs of useful receptor fragments (as described above) may also be produced and tested for efficacy as screening components or antagonists (using the assays described herein) ; such analogs are also considered to be useful in the invention.
  • receptor fragments encompassing the extracellular amino-terminal domain (or an IL-8-binding fragment thereof) ; this domain includes approximately amino acids 1-37 of the high affinity IL-8 receptor isolated from a rabbit source, approximately amino acids 1-49 of the low affinity IL-8 receptor isolated from a rabbit source, and approximately amino acids 1-50 of the low affinity IL-8 receptor isolated from a human source.
  • IL-8 receptor extracellular loops include approximately amino acids 94-113, 186-202, and 268-285 of the high affinity IL-8 receptor isolated from rabbits; approximately amino acids 106-118, 183-210, and 272-298 of the low affinity IL-8 receptor isolated from rabbits; and approximately amino acids 107-120, 184-213, and 274- 300 of the low affinity IL-8 receptor isolated from humans.
  • Peptide fragments derived from these extracellular loops may also be used as antagonists, particularly if the loops cooperate with the amino- terminal domain to facilitate IL-8 binding.
  • loops and extracellular N-terminal domain provide im unogens for producing anti-IL-8 receptor antibodies.
  • the rabbit high affinity IL-8 receptor gene was isolated as follows.
  • Rabbit peritoneal neutrophils were isolated from rabbits by the method of Zigmond and Tranquillo ( ,
  • This probe was designed based on the sequence derived from the second transmembrane domain of G-protein-coupled receptors (see, e.g., Hartig et al., TIBS 10:64, 1989) .
  • This probe was 5•-end-labeled with [ 32 P]ATP (Du Pont-New England Nuclear, Boston, MA) and T4 kinase (New England Biolabs, Beverly, MA) by the methods of Maniatis et al., supra.
  • the hybridization conditions were as follows: 6X SSPE, 1% SDS, 0.1% sodium pyrophosphate, IX Denhardt's, 100 ⁇ g/ml poly(A) , and 40 ⁇ g/ml denatured calf thymus DNA at 42°C for 12 h. Filters were washed with 2X SSC, 0.1% SDS at 50 ⁇ C. After tertiary screening, six plaques were isolated. The insert of one of these plaques, termed F3R was of 2.5 kb in size. This insert was sequenced using Sequenase 2.0 (U.S. Biochemical Corp., Cleveland, OH) according to the method of Sanger et al. Proc. Natl . Acad. Scl .
  • F3R Several structural features of the protein deduced from the F3R clone demonstrate that it belongs to the family of G-protein-coupled receptors.
  • a hydropathy plot of the deduced protein sequence indicates the existence of seven putative transmembrane segments.
  • the primary structure of F3R shows a high degree of similarity to other G-protein-coupled receptors.
  • the highest degree of homology is found to G- protein-coupled receptors that bind peptides such as the substance K and P receptors (Masu et al., Nature 329:836. 1987;Hershey and Krause, Science 247:958. 1990).
  • F3R exhibits several structural features attributed to G- protein-coupled receptors.
  • F3R contains two putative N-linked glycosylation sites in the N-terminus with no signal sequence. It also contains an aspartate at position 80 (i.e., in transmembrane segment II) which is conserved in all G-protein-coupled receptors, and the canonical Asp-Arg-Tyr tripeptide close to the putative transmembrane segment III. Like substance K and P receptors, F3R lacks Asp-113 in the putative transmembrane segment II which appears to be essential for binding of charged amines in adrenergic, muscarinic, dopaminergic, and serotonergic receptors (Dixon et al., Cold Spring Harbor Symp. Quant .
  • F3R G-protein-coupled receptors
  • F3R exhibits several critically-located serine and threonine residues which are potential substrates for protein kinases (Benovic et al., Ann. J?ev. Cell Biol . 4_:405, 1988).
  • the F3R cDNA was employed as a hybridization probe in Northern blot analysis of rabbit neutrophil RNA.
  • RNA was isolated from neutrophils and other tissues by cesium chloride gradient centrifugation (Glisin et al..
  • the hybridization solution contained 50% forma ide, 5X SSPE, 5X Denhardt's, 0.1% sodium pyrophosphate, 1 mg/ml heparin, 100 ⁇ g/ml poly(A) , 1% SDS, and 200 ⁇ g/ml denatured calf thymus DNA.
  • the blot was hybridized at 42°C for 16 h, and then washed with 0.1X SSC and 0.1% SDS at 65°C.
  • the F3R probe hybridized specifically to a neutrophil RNA molecule of 2.6 kilobases. This confirmed that F3R was expressed in neutrophils and indicated that the F3R clone was nearly full-length.
  • the F3R clone failed to hybridize to RNA isolated from rabbit uterine smooth muscle, skeletal muscle, lung, liver, or brain. It also failed to hybridize to poly(A) + RNA from fibroblasts, epithelial, and endothelial cells. Promyelocytic HL-60 cells exhibited very low levels of F3R mRNA; differentiated HL-60 cells expressed 20-fold higher levels of this RNA.
  • the F3R mRNA was translated in vitro in rabbit reticulocyte lysates by the method of Promega Corp. (Madison, WI) .
  • a protein of relative mass 30,000-32,000 Daltons was synthesized as determined by SDS- polyacrylamide gel electrophoresis (SDS-PAGE; carried out by standard techniques; see, e.g., Ausubel et al.. Current Protocols in Molecular Biology, Green Publishing Associates, New York, 1987) .
  • the difference between the calculated M r of 40,528 and the apparent M r of about 31,000 was likely due to the fact that membrane proteins frequently exhibit increased mobility relative to soluble protein standards on SDS-PAGE (Bonitz et al. , J. Biol . Chem.
  • a cDNA encoding the rabbit low affinity IL-8 receptor was also identified and isolated from the rabbit neutrophil library (described above) . This cDNA was subcloned into the E ⁇ oRI site of pUC19 to produce plasmid 5bla. Its nucleic acid sequence was determined by standard techniques and found to be similar, but not identical, to the high affinity receptor clone F3R.
  • Cloning of a Low Affinity IL-8 Receptor from a Human Source A human peripheral blood leukocyte ⁇ gtll cDNA library (5* stretch) obtained from Clontech (Palo Alto, CA) was screened with a 652 base pair JJcoRI/BamHI fragment (including nucleotides -27 to 625) of the rabbit F3R clone. This probe was labeled with [ 32 P]dCTP by random priming as described above.
  • Filters were hybridized with a solution containing 50% formamide, 200 ⁇ g/ml denatured calf thymus DNA, 5X SSPE, 1% SDS, 5X Denhardt's solution, and 0.1% sodium pyrophosphate, and incubated at 42°C for 16 hours. Filters were then washed with 0.1X SSC and 0.1% SDS at 65°C. After tertiary screening, several human clones which hybridized to the rabbit IL-8 probe were isolated. The insert of one such clone, termed 4AB, was found to be 4.0 kilobases in length; the insert was sequenced on both strands using Sequenase 2.0 (U.S.
  • a human IL-8 receptor-encoding gene may be isolated by hybridization with the full-length F3R probe. This probe is labelled (e.g., radiolabelled) by standard techniques (see, e.g., Ausubel et al..
  • a human peripheral blood leukocyte library e.g., the library described above
  • low stringency conditions e.g., hybridization in 50% formamide, 200 ⁇ g/ml denatured calf thymus DNA, 5X SSPE, 1% SDS, 5X Denhardt's solution, and 0.1% sodium pyrophosphate at an incubation temperature or 42°C for 16 hours.
  • the human IL-8 receptor gene may also be isolated by PCR cloning using primer sequences based either on the sequence of clone 4AB, for example:
  • Primers are synthesized by standard cyanoethyl phosphora idite chemistry using, e.g., an Applied Biosystems DNA Synthesizer (Foster City, CA) .
  • Human neutrophils are isolated by standard techniques and used as a source of polyA + RNA as described above.
  • cDNA is synthesized, also as described above, and a neutrophil cDNA library created by insertion of the cDNA fragments into any standard cloning vector, e.g., ⁇ gtll.
  • a human peripheral blood leukocyte ⁇ gtll cDNA library (5' stretch) may be purchased from Clontech (Palo Alto, CA) .
  • the resultant PCR product is isolated by electrophoresis and cloned, e.g., into the vector SK+ (Stratagene, LaJolla CA) and amplified in Escherichia coli XL-1 blue (Stratagene) .
  • Polypeptide Expression is isolated by electrophoresis and cloned, e.g., into the vector SK+ (Stratagene, LaJolla CA) and amplified in Escherichia coli XL-1 blue (Stratagene) .
  • Polypeptides according to the invention may be produced by transformation of a suitable host cell with all or part of an IL-8 receptor-encoding cDNA fragment (e.g., the cDNAs described above) in a suitable expression vehicle, and expression of the receptor.
  • a suitable host cell with all or part of an IL-8 receptor-encoding cDNA fragment (e.g., the cDNAs described above) in a suitable expression vehicle, and expression of the receptor.
  • any of a wide variety of expression systems may be used to provide the recombinant receptor protein.
  • the precise host cell used is not critical to the invention, however the following host cells are preferred: COS-7, SP-2, NIH 3T3, and Chinese Hamster Ovary cells, Chinese hamster lung fibroblast Dede cells. Such cells are available from a wide range of sources (e.g., the American Type Culture Collection, Rockville, MD) .
  • the method of transfection and the choice of expression vehicle will depend on the host system selected. Mammalian cell transfection methods are described, e.g., in Ausubel et al.
  • expression vehicles may be chosen from those provided, e.g., in Cloning Vectors : A Laboratory Manual (P.H. Pouwels et al., 1985, Supp. 1987).
  • One particularly preferred expression system is the mouse 3T3 fibroblast host cell transfected with a pMAMneo expression vector (Clontech, Palo Alto, CA) .
  • pMAMneo provides: an RSV-LTR enhancer linked to a dexamethasone-inducible MMTV-LTR promotor, an SV40 origin of replication which allows replication in mammalian systems, a selectable neomycin gene, and SV40 splicing and polyadenylation sites.
  • DNA encoding the human or rabbit IL-8 receptor or an appropriate receptor fragment or analog (as described above) would be inserted into the pMAMneo vector in an orientation designed to allow expression.
  • the recombinant receptor protein would be isolated as described below.
  • Other preferable host cells which may be used in conjunction with the pMAMneo expression vehicle include COS cells and CHO cells (ATCC Accession Nos. CRL 1650 and CCL 61, respectively) .
  • COS host cell ATCC Accession No. CRL 1650
  • pSVL vector Pharmacia
  • the high affinity or low affinity IL-8 receptor is produced by a stably-transfected mammalian cell line.
  • cDNA encoding the receptor is cloned into an expression vector which includes the dihydrofolate reductase (DHFR) gene.
  • DHFR dihydrofolate reductase
  • Integration of the plasmid and, therefore, the IL-8 receptor-encoding gene into the host cell chromosome is selected for by inclusion of 0.01-300 ⁇ M methotrexate in the cell culture medium (as described in Ausubel et al., supra ) .
  • This dominant selection can be accomplished in most cell types.
  • Recombinant protein expression can be increased by DHFR-mediated amplification of the transfected gene.
  • Methods for selecting cell lines bearing gene amplifications are described in Ausubel et al. (supra) ; such methods generally involve extended culture in medium containing gradually increasing levels of methotrexate.
  • DHFR-containing expression vectors commonly used for this purpose include pCVSEII-DHFR and pAdD26SV(A) (described in Ausubel et al. , supra) .
  • Any of the host cells described above or, preferably, a DHFR-deficient CHO cell line e.g., CHO DHFR cells, ATCC Accession No. CRL 9096
  • CHO DHFR cells ATCC Accession No. CRL 9096
  • One particularly preferred stable expression system is the myeloma cell line, J558 (ATCC Accession No. TIB6) or SP2 (ATCC Accession No.
  • pSV2-gpt provides: an SV40 early promotor and a selectable gpt marker (i.e., E. coli xanthine-guanine phosphoribosyl transferase) .
  • Another particularly preferred stable expression system is a Chinese hamster lung fibroblast Dede cell line (ATCC Accession No. CCL39, American Type Culture Collection, Rockville, MD) stably transfected with a pMAMneo vector.
  • This cell line has been used to inducibly express the rabbit IL-8 receptor as follows.
  • the F3R receptor cDNA (subcloned into a Bluescript vector, Stratagene, Jolla, CA) was cleaved with Xbal and Xhol, and a fragment of approximately 1700 bp was isolated and inserted into an Nhel/Xhol-digested pMAMneo expression vector (Clontech, Palo Alto, CA) , to create F3R-pMAMneo.
  • F3R-pMAMneo directs the expression of the rabbit high affinity IL-8 receptor protein under the control of the glucocorticoid-inducible mouse mammary tumor virus promoter.
  • F3R-pMAMneo was used to transfect Chinese hamster lung fibroblast Dede cells (ATCC No. CCL39, American Type Culture Collection, Rockville, MD) using the Lipofectin procedure of BRL (Gathersburg, MD) .
  • Transfected cells were selected by growth in medium which included 500 ⁇ g/ml Geneticin (Sigma Chemical Co., St. Louis, MO).
  • G418-resistant clones termed HI, H9, Hll, and H12, were isolated by standard techniques.
  • IL- 8 receptor protein was produced in such cells following a 24 hour treatment with l ⁇ M dexamethasone.
  • the ability of the receptor-expressing cells to bind IL-8 was assayed (as described below for Tables 1 and 2) , and the results are shown in Fig. 3.
  • This system may be used to inducibly express any polypeptide of the invention.
  • transfection of the Chinese hamster lung fibroblast Dede cell line (CCL39) with vector RC/CMV (Invitrogen, San Diego, CA) using the methods described above provides a preferred system for the constitutive expression of the polypeptides of the invention.
  • recombinant receptor e.g., produced by any of the expression systems described herein
  • immunological procedures such as Western blot or immunoprecipitation analysis of recombinant cell extracts, or by immunofluorescence of intact recombinant cells (using, e.g, the methods described in Ausubel et al., supra) .
  • Recombinant receptor protein is detected using an antibody directed to the receptor.
  • One such antibody is described below; also described below are methods for producing other IL- 8 receptor antibodies using, as an i munogen, the intact receptor or a peptide which includes a suitable IL-8 receptor epitope.
  • the antibody is preferably produced using, as an immunogen, an epitope included in the fragment or analog.
  • an immunogen an epitope included in the fragment or analog.
  • the recombinant IL-8 receptor protein or fragment or analog, thereof is expressed, it is isolated, e.g., using immunoaffinity chromatography.
  • IL-8 or an anti-IL-8 receptor antibody e.g., the IL-8 receptor antibody described below
  • Lysis and fractionation of receptor-harboring cells prior to affinity chromatography may be performed by standard methods (see, e.g., Ausubel et al. , supra) .
  • the recombinant protein can, if desired, be further purified, e.g., by high performance liquid chromatography (see, e.g., Fisher, Laboratory Techniques In Biochemistry And Molecular Biology, eds.. Work and Burdon, Elsevier, 1980) .
  • Receptors of the invention particularly short receptor fragments, can also be produced by chemical synthesis (e.g., by the methods described in Solid Phase Peptide Synthesis, 2nd ed. , 1984 The Pierce Chemical Co., Rockford, IL) .
  • Useful receptor fragments or analogs in the invention are those which interact with IL-8. Such an interaction may be detected by an in vitro binding assay (see below) .
  • the receptor component may also be assayed functionally, i.e., for its ability to bind IL-8 and mobilize Ca ++ (see below) .
  • These assays include, as components, IL-8 and a recombinant IL-8 receptor (or a suitable fragment or analog) configured to permit detection of binding.
  • IL-8 may be obtained from Genzyme (Cambridge, MA) .
  • the IL-8 receptor component is produced by a cell that naturally presents substantially no receptor, e.g., by engineering such a cell to contain nucleic acid encoding the receptor component in an appropriate expression system. Suitable cells are, e.g., those discussed above with respect to the production of recombinant receptor, such as the myeloma cells, J558 or SP2.
  • In vitro assays to determine the extent of IL-8 binding to the IL-8 receptor may be carried out using either whole cells or membrane fractions.
  • a whole cell assay is preferably performed by fixing the cell expressing the IL-8 receptor component to a solid substrate (e.g., a test tube, a microtiter well, or a column) by means well known to those in the art (see, e.g., Ausubel et al., supra) . and presenting labelled IL- 8 (e.g., I-labelled IL-8) . Binding s assayed by the detection label in association with the receptor component (and, therefore, in association with the solid substrate) .
  • the assay format may be any of a number of suitable formats for detecting specific binding, such as a radioi munoassay format (see, e.g., Ausubel et al., supra) .
  • a radioi munoassay format see, e.g., Ausubel et al., supra
  • cells transiently or stably transfected with an IL-8 receptor expression vector are immobilized on a solid substrate (e.g., the well of a microtiter plate) and reacted with IL-8 which is detectably labelled, e.g., with a radiolabel or an enzyme which can be assayed, e.g., alkaline phosphatase or horseradish peroxidase.
  • COS cells were transiently transfected with varying amounts of the rabbit IL-8 receptor-expressing clone F3R- pSVL (see above) .
  • Membranes were harvested by standard techniques and used in an in vitro binding assay (see below) .
  • 125 I-labelled IL-8 was bound to the membranes and assayed for specific activity; specific binding was determined by comparison with binding assays performed in the presence of excess unlabelled IL-8. The results are shown in Table 1.
  • COS cells were transiently transfected with 8 ⁇ g of the human IL-8-expressing clone 4AB-pSVL (see above) .
  • Cells were harvested after three days and 2.5 nM I-labelled IL-8 was added to approximately 1 X 10 5 whole cells (in 200 ⁇ l PBS) .
  • Cells were incubated with IL-8 for 45 minutes at 4°C, pelleted by centrifugation, rinsed with cold phosphate buffered saline, and the cell-bound radioactivity measured in a gamma counter. Specific binding was determined by comparison with binding assays performed in the presence of excess (i.e., 250 nM) unlabelled IL-8.
  • IL-8 may be adhered to the solid substrate (e.g., to a microtiter plate using methods similar to those for adhering antigens for an ELISA assay; Ausubel et al., supra) and the ability of labelled IL-8 receptor-expressing cells to bind IL-8 (e.g., labelled with 3 H-thymidine; Ausubel et al., supra) can be used to detect specific receptor binding to the immobilized IL-8.
  • the solid substrate e.g., to a microtiter plate using methods similar to those for adhering antigens for an ELISA assay; Ausubel et al., supra
  • the ability of labelled IL-8 receptor-expressing cells to bind IL-8 e.g., labelled with 3 H-thymidine; Ausubel et al., supra
  • a vector expressing the IL-8 receptor is transfected into myeloma cells (e.g., J558 or SP2 cells) by the DEAE dextran-chloroquine method (Ausubel et al. , supra) .
  • Expression of the receptor protein confers binding of detectably-labelled IL-8 to the cells.
  • IL-8 does not bind significantly to untransfected host cells or cells bearing the parent vector alone; these cells are used as a "control" against which the binding assays are measured.
  • Tissue culture dishes e.g., 10 cm.
  • IL-8 receptor-expressing myeloma cells approximately 750,000 cells, dish 12-18h post- transfection. Forty-eight hours later, triplicate dishes are incubated with 0.5nM radioiodinated IL-8 (200
  • Ci/mmol binding to the receptor-bearing cells is assayed (e.g., by harvesting the cells and assaying the amount of detectable label in association with the cells) .
  • Cells which specifically bind labelled IL-8 are those which exhibit a level of binding (i.e., an amount of detectable label) which is greater than that of the control cells.
  • IL-8 receptor encoding RNA (prepared as described below) is injected into Xenopus laevis oocytes by standard methods. The RNA is translated in vivo in the oocytes, and the IL-8 receptor protein is inserted into the cell membrane.
  • oocyte membranes are prepared by sucrose gradient centrifugation (by the method of Colman, Transcription and Translation, IRL Press, Oxford, 1986) and 125 I-labelled IL-8 is added, and the membrane preparation subjected to vacuum filtration through Whatman GF/C filters (by the method of Williamson, Biochemistry, 22:5371, 1988).
  • a recombinant receptor may also be assayed functionally for its ability to mediate IL-8-dependent mobilization of calcium.
  • Cells preferably myeloma cells, transfected with an IL-8 expression vector (as described above) are loaded with FURA-2 or INDO-1 by standard techniques. Mobilization of calcium induced by IL-8 is measured by fluorescence spectroscopy as previously described (Grynkiewicz et al. , J. Biol . Chem . 160:3440, 1985).
  • Receptors Affinity of IL-8 Receptors for the IL-8 Ligand The K d of the high affinity F3R receptor was determined as follows.
  • pSVL-F3R-transfected COS-7 cell membranes (at a constant amount) were incubated in phosphate buffered saline containing either 125 I-labelled IL-8 at a concentration of between 0 and 50nM or were incubated in phosphate buffered saline containing 0.3nM
  • IL-8 binding to the low affinity IL-8 receptor (4AB) was measured as follows. 5 X 10 6 COS cells were transiently transfected with 8 ⁇ g of the human IL-8- expressing clone 4AB-pSVL (see above) . After 3 days, cells were rinsed twice with 7 ml phosphate buffered saline (PBS) and once with 7 ml PBS/lmM EDTA, and incubated in 7 ml of PBS/lmM EDTA at 37°C for 5-10 minutes.
  • PBS phosphate buffered saline
  • the cells were then collected, added to 25 ml of ice cold PBS/0.1% bovine serum albumin (BSA), counted, pelleted by centrifugation, and resuspended in ice cold PBS/0.1% BSA at a concentration of 2 X 10 7 cells/ml.
  • BSA bovine serum albumin
  • 1 5 I-labelled IL-8 (at a concentration of between 0 and 20nM) was added to 0.6-1 X 10 6 whole cells (in lOO ⁇ l PBS/0.1% BSA), incubation was allowed to proceed for 60 minutes at 0°C, and cells were filtered through GF/C filters soaked with 0.3% polyethylenimine (PEI; Sigma, St. Louis, MO), rinsed with cold PBS, and the cell-bound radioactivity measured in a gamma counter. Specific binding was determined by comparison with binding assays performed in the presence of a 300-fold excess of unlabelled IL-8.
  • PEI polyethylenimine
  • the K d for the low affinity receptor was calculated to be approximately 31nM; this may be compared with the K d of 1.4nM measured for the high affinity IL-8 receptor F3R (supra) .
  • IL-8 binding to the 4AB receptor was subsequently measured by the following assay using stably transfected cells, and the Kd was calculated to be approximately 8.4 nM.
  • the 4AB coding region was subcloned into the Hindlll/Xbal sites of the plasmid RC/CMV (Invitrogen) to create pRC.4AB.
  • CH0DG44 cells a double DHFR mutant cell line (gift of Lawrence Chaisen, Columbia University, New York, NY), were stably transfected with pRC.4AB expression vector and a subcloned expressing line was isolated (4ABCH033) . Two to three days after passage, the cells were rinsed twice with PBS and treated as described above. To test IL-8 binding, 125 I-labelled IL- 8 (1.0 to 2.0 nM) was added to samples containing 2.5 x 10 s cells and increasing amounts of unlabelled IL-8. Incubation was allowed to proceed for
  • Binding data was analyzed by non-linear least-squares curve fitting, using the generalized model for complex ligand-receptor systems (Hoffman et al., 1979, Life Sci. , 24:1739) and EBDA/LIGAND programs (McPherson, 1985, Kinetic, EBDA, Ligand, Lowry; A collection of radioligand binding analysis programs, Cambridge, U.K.; Biosoft) .
  • the results demonstrate saturable, specific binding of [ 125 I] IL-8, and Scatchard analysis of the binding data reveal a single binding site with a Kd of 8.4 nM.
  • MGSA/GRO ⁇ 125I-labelled MGSA/GRO ⁇ (Moser et al., J. Exp. Med. 171:1797. 1990; Richmond et al., EMBO J. 2 ⁇ 2025, 1988; and Anisowicz et al., Proc. Natl . Acad. Sci . USA 84:7188. 1987) was added at concentration of between 2 and 7.5nM. Nonspecific binding was determined by adding a 300-fold excess of unlabelled MGSA/GRO ⁇ or unlabelled IL-8. As shown in Fig.
  • the low affinity IL-8 receptor encoded by 4AB bound the ligand MGSA/GRO ⁇ and is displaced similarly with either unlabelled MGSA/GRO ⁇ or unlabelled IL-8. In contrast, no binding of MGSA/GRO ⁇ was detectable to the high affinity F3R receptor protein (not shown) .
  • COS cells were transiently transfected with 4AB-pSVL (as described above). After 3 days, cells were harvested as described above and resuspended in ice cold PBS/0.1% BSA at a concentration of 1.38 x 10 7 cells/ml.
  • 125 I-labelled IL-8 (at a concentration of 5nM) was added to a mixture of 6.9 x 10 5 whole cells expressing the low affinity receptor (in lOO ⁇ l PBS/0.1% BSA) and unlabelled ligand (specifically, IL-8 at a concentration of between 0 and 5000nM, PF4 at a concentration of between 50 and 5000nM, MGSA/GRO ⁇ at a concentration of 50 or 500nM, or FMLP at a concentration of between 50 and 5000nM) .
  • Cells were incubated in the presence of ligand for one hour at 4°C, filtered through GF/C filters which had been soaked in 0.3% PEI, rinsed with cold PBS/0.1% BSA, and the cell-bound radioactivity measured in a gamma counter.
  • IL-8 and MGSA/GRO ⁇ successfully competed with IL-8 for binding to the low affinity receptor.
  • Two other peptide ligands, PF4 and FMLP (Deuel et al., Proc. Natl . Acad. Sci . USA 78.:4585, 1981; Coats and Navarro, J. Biol . Chem . 265:5964. 1990) had little or no effect on IL-8 binding.
  • the low affinity receptor was not absolutely specific for IL-8; rather, it bound other closely-related members of the IL- 8 family.
  • the high affinity receptor was specific for IL-8 among the ligands measured.
  • COS cells were transiently transfected with vectors expressing each of the receptors. Cells were harvested as described above and resuspended in ice cold PBS/0.1% BSA. To test ligand binding, 2.5 x 10 5 cells were added to 2 nM [ 1 5 I] IL-8 in the presence of the unlabeled ligands PF4, MGSA/GROa, NAP-2 and fMLP.
  • the chimeric receptor containing the F3R extracellular N-terminal domain fused to the backbone of 4AB exhibits a ligand binding profile approximately the F3R subtype, whereas a receptor chimera containing the 4AB extracellular domain fused to the F3R backbone shows a ligand binding profile resembling the human 4AB receptor subtype.
  • the expression vectors F3R- pSVL and 4AB-pSVL (described below) were each digested with Xhol and Celll, and a fragment encoding the amino terminus of one receptor was exchanged for a fragment encoding the amino terminus of the other receptor.
  • a 271 bp Xhol-Celll fragment of F3R containing the first 58 codons (i.e., up to and including Ser 58 of Fig. 1) was excised from F3R-pSVL and cloned into a Xhol-Celll ended 4AB-pSVL backbone.
  • a 283 bp Xhol-Celll fragment of 4AB containing the first 62 codons was likewise excised from 4AB-pSVL and cloned into a Xhol-Celll ended F3R-pSVL backbone.
  • Two chimeric IL-8 receptor genes were thus created; one encoding the amino-terminal 58 amino acids of rabbit F3R fused to the 298 carboxy-terminal amino acids of human 4AB (termed F3R/4AB) and the second encoding the amino- terminal 62 amino acids of human 4AB fused to the 297 carboxy-terminal amino acids of rabbit F3R (termed 4AB/F3R) .
  • IL-8 binding assays e.g., those described above
  • This difference in affinity was used to identify the IL-8 binding domain as follows.
  • COS cells were transiently transfected with F3R/4AB-pSVL or 4AB/F3R-pSVL chimeric receptor expression plasmids (described above) , and cells were harvested and washed as described above.
  • IL-8 bound F3R/4AB more 10 readily than it bound 4AB/F3R.
  • the amount of IL-8 binding the chimeric proteins mirrored the amount of IL- 8 binding to the amino-terminal portion of each protein; thus, the first 58 amino acids of the high affinity receptor conferred high affinity binding properties to 15 the low affinity receptor, and the first 62 amino acids of the low affinity receptor conferred low affinity binding properties to the high affinity receptor.
  • the high affinity IL-8 binding domain is contained in the amino terminus of the F3R 20 protein and the low affinity IL-8 binding domain is contained in the amino terminus of the 4AB protein.
  • the F3R/4AB chimera bound IL-8 more strongly than either the F3R or the 4AB receptor, indicating that interaction(s) between the amino-terminal 25 binding domain and other portions of the molecule may occur.
  • IL- 8 receptor Binding of ligand by the amino terminus of the IL- 8 receptor was also suggested by the experiment depicted in Fig. 8.
  • COS cells were transiently transfected with 30 4AB-pSVL or F3R/4AB-pSVL and harvested and washed as descri •bed above.
  • 1.2 ⁇ M 125I-labelled IL-8 was added to a mixture of 2 x 10 5 whole cells (in 50 ⁇ l PBS/0.1% BSA) and increasing concentrations of competing ligand (i.e., between 0 and lOOOnM unlabelled IL-8 or between 10 and 35 500nM MGSA/GRO ⁇ ) .
  • one aspect of the invention features screening for compounds that antagonize the interaction between IL-8 and the IL-8 receptor, thereby preventing or reducing the cascade of events that are mediated by that interaction.
  • the elements of the screen are IL-8 and recombinant IL-8 receptor (or a suitable receptor fragment or analog, as outlined above) configured to permit detection of binding.
  • IL-8 may be purchased from Genzyme and a full- length rabbit or human IL-8 receptor (or an IL-8-binding fragment or analog) may be produced as described herein. Binding of IL-8 to its receptor may be assayed by any of the methods described above.
  • cells expressing recombinant IL-8 receptor are immobilized on a solid substrate (e.g., the well of a microtiter plate or a column) and reacted with detectably-labelled IL-8 (as described above) . Binding is assayed by the detection label in association with the receptor component (and, therefore, in association with the solid substrate) . Binding of labelled IL-8 to receptor-bearing cells is used as a "control" against which antagonist assays are measured. The antagonist assays involve incubation of the IL-8 receptor-bearing cells with an appropriate amount of candidate antagonist. To this mix, an equivalent amount of labelled IL-8 is added. An antagonist useful in the invention specifically interferes with labelled IL-8 binding to the immobilized receptor-expressing cells.
  • a solid substrate e.g., the well of a microtiter plate or a column
  • detectably-labelled IL-8 as described above
  • Binding is assayed by the detection label in association with the receptor
  • IL-8 function i.e., to specifically interfere with labelled IL-8 binding without resulting in signal transduction normally mediated by the receptor.
  • stably transfected cell lines containing the IL-8 receptor can be produced as described herein and reporter compounds such as the calcium binding agent, FURA-2, loaded into the cytoplasm by standard techniques. Stimulation of the heterologous IL-8 receptor with IL-8 or another agonist leads to intracellular calcium release and the concomitant fluorescence of the calcium-FURA-2 complex. This provides a convenient means for measuring agonist activity.
  • IL-8 Inclusion of potential antagonists along with IL-8 allows for the screening and identification of authentic receptor antagonists as those which effectively block IL-8 binding without producing fluorescence (i.e., without causing the mobilization of intracellular Ca ++ ) .
  • Such an antagonist may be expected to be a useful therapeutic agent for inflammatory disorders.
  • candidate antagonists include IL-8 receptor fragments, particularly fragments containing an IL-8-binding portion adjacent to or including one or more transmembrane segments 2-7 or an extracellular domain of the receptor (described above) ; such fragments would preferably including five or more amino acids.
  • candidate antagonists include analogs of IL-8 and other peptides as well as non-peptide compounds and anti-IL-8 receptor antibodies designed or derived from analysis of the receptor.
  • High affinity or low affinity IL-8 receptors may be used to raise antibodies useful in the invention.
  • receptor fragments preferred for the production of antibodies are those fragments deduced or shown experimentally to be extracellular; such fragments include the extracellular N-terminal domain.
  • Antibodies directed to IL-8 receptor peptides are produced as follows. Peptides corresponding to all or part of the putative extracellular loops (approximately amino acids 94-113, 186-202, and 268-285 of the high affinity IL-8 receptor or approximately amino acids 107- 120, 184-213, and 274-300 of the low affinity IL-8 receptor) or to all or a portion of the extracellular N- terminal domain (approximately amino acids 1-37 of the high affinity IL-8 receptor or approximately amino acids 1-50 of the low affinity IL-8 receptor) are produced using a peptide synthesizer, by standard techniques. The peptides are coupled to KLH with -maleimide benzoic acid N-hydroxysuccinimide ester.
  • the KLH-peptide is mixed with Freund's adjuvant and injected into animals, e.g. guinea pigs or goats.
  • Antibodies are purified by peptide antigen affinity chromatography. Using such a method, polyclonal antisera were raised to peptides which included the N-terminal extracellular domain and also to loops 2 and 3. Additional peptides used for immunizations were the following:
  • antibodies to the IL-8 receptor are produced using whole cells expressing the IL-8 receptor, or membrance fractions of these cells (both described above) .
  • whole cells expressing the IL-8 receptor or membrance fractions of these cells (both described above) .
  • approximately 10 7 transiently transfected C0S7 cells, stably transfected CHO cells, or membrane fragments corresponding to 50 ⁇ g total membrane protein are injected into mice. After 2 weeks and 4 weeks the animals are boosted with approximately 10 7 cells or membrane fragments corresponding to 10-25 ⁇ g protein. Approximately 3 weeks following the second boost, the animals are boosted once again, and spleen cells are removed for the making of hybridomas using standard techniques.
  • Hybridomas producing antibodies that bind to the IL-8 receptor are screened by FACS (flourescence activated cell sorter) , by cell-based ELISA using untransfected versus transfected cells (preferably of a cell type different from the cells used in the immunization) , or using membranes.
  • Hybridomas producing antibodies that bind to tranfected cells are subcloned and tested for ability to block IL-8 binding to the receptor, or to block IL-8 dependent signal transduction. Once produced, antibodies are tested for specific IL-8 receptor recognition by Western blot or immunoprecipitation analysis (by the methods described in Ausubel et al. , supra) .
  • Antibodies which specifically recognize the IL-8 receptor are considered to be likely candidates for useful antagonists; such candidates are further tested for their ability to specifically interfere with the interaction between IL-8 and its receptor (as described above) .
  • Antibodies which antagonize IL-8/IL-8 receptor binding or IL-8 receptor function are considered to be useful as antagonists in the invention.
  • the soluble antagonistic receptor fragments described above formulated in an appropriate buffer such as physiological saline.
  • the fragment may include a sufficient number of adjacent transmembrane residues.
  • the fragment may be associated with an appropriate lipid fraction (e.g., in lipid vesicles or attached to fragments obtained by disrupting a cell membrane) .
  • anti-IL-8 receptor antibodies produced as described above may be used as a therapeutic. Again, the antibodies would be administered in a pharmaceutically-acceptable buffer (e.g., physiological saline). If appropriate, the antibody preparation may be combined with a suitable adjuvant.
  • the therapeutic preparation is administered in accordance with the condition to be treated. Ordinarily, it will be administered intravenously, at a dosage that provides suitable competition for IL-8 binding. Alternatively, it may be convenient to administer the therapeutic orally, nasally, or topically, e.g., as a liquid or a spray. Again, the dosages are as described above. Treatment may be repeated as necessary for alleviation of disease symptoms. Antagonists may also be administered to prevent (as well as treat) inflammation; the antagonist is administered as described above.
  • IL-8 receptor antagonists can be used to treat or prevent any inflammatory disease in which neutrophils play a principal role, such as psoriasis, rheumatoid arthritis, and other chronic disorders as well as acute inflammatory disorders such as reperfusion injury, septic shock, trauma shock, and pulmonary disorders such as adult respiratory distress syndrome (ARDS) and inflammatory airway disorders caused by bacterial infections in cystic fibrosis patients.
  • inflammatory disease in which neutrophils play a principal role such as psoriasis, rheumatoid arthritis, and other chronic disorders as well as acute inflammatory disorders such as reperfusion injury, septic shock, trauma shock, and pulmonary disorders such as adult respiratory distress syndrome (ARDS) and inflammatory airway disorders caused by bacterial infections in cystic fibrosis patients.
  • ARDS adult respiratory distress syndrome
  • the methods of the invention may be used to reduce inflammatory responses in any mammal, for example, humans, domestic pets, or livestock.
  • a non-human mammal is treated, the IL-8 receptor or receptor fragment or analog or the antibody employed is preferably specific for that species.
  • Val lie Tyr Ala Leu Val Phe Leu Leu Ser Leu Leu Gly Asn Ser Leu 50 55 60
  • Trp Ala Val Ser Lys Glu Lys Glu Trp lie Phe Gly Thr 100 105 110 CCC CTG TGC AAG GTG GGG TCG CTT GTG AAG GAA GTC AAC TTC TAC AGT 491
  • GGC ACG CCC CTG TGT AAG GTG GTC TCG CTT GTG AAG GAA GTC AAC TTC 454 Gly Thr Pro Leu Cys Lys Val Val Ser Leu Val Lys Glu Val Asn Phe 115 120 125

Landscapes

  • Health & Medical Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Organic Chemistry (AREA)
  • Medicinal Chemistry (AREA)
  • General Health & Medical Sciences (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Rheumatology (AREA)
  • Biochemistry (AREA)
  • Biophysics (AREA)
  • Zoology (AREA)
  • Genetics & Genomics (AREA)
  • Toxicology (AREA)
  • Molecular Biology (AREA)
  • Immunology (AREA)
  • Cell Biology (AREA)
  • Pain & Pain Management (AREA)
  • Gastroenterology & Hepatology (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • General Chemical & Material Sciences (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Animal Behavior & Ethology (AREA)
  • Public Health (AREA)
  • Veterinary Medicine (AREA)
  • Peptides Or Proteins (AREA)
  • Preparation Of Compounds By Using Micro-Organisms (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)
EP19920910198 1991-04-10 1992-04-10 Interleukin-8-rezeptoren, damit in zusammenhang stehende moleküle und verfahrensweisen. Withdrawn EP0579739A4 (de)

Applications Claiming Priority (6)

Application Number Priority Date Filing Date Title
US685101 1984-12-21
US68510191A 1991-04-10 1991-04-10
US72660691A 1991-07-09 1991-07-09
US726606 1991-07-09
US80384291A 1991-12-09 1991-12-09
US803842 1991-12-09

Publications (2)

Publication Number Publication Date
EP0579739A1 true EP0579739A1 (de) 1994-01-26
EP0579739A4 EP0579739A4 (de) 1994-03-22

Family

ID=27418452

Family Applications (1)

Application Number Title Priority Date Filing Date
EP19920910198 Withdrawn EP0579739A4 (de) 1991-04-10 1992-04-10 Interleukin-8-rezeptoren, damit in zusammenhang stehende moleküle und verfahrensweisen.

Country Status (4)

Country Link
EP (1) EP0579739A4 (de)
JP (1) JPH06506596A (de)
CA (1) CA2107682A1 (de)
WO (1) WO1992018641A1 (de)

Families Citing this family (22)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5840856A (en) * 1991-03-29 1998-11-24 Genentech, Inc. Antibodies to a human PF4 superfamily receptor
US5783415A (en) * 1991-03-29 1998-07-21 Genentech, Inc. Method of producing an IL-8 receptor polypeptide
US5440021A (en) * 1991-03-29 1995-08-08 Chuntharapai; Anan Antibodies to human IL-8 type B receptor
US5543503A (en) * 1991-03-29 1996-08-06 Genentech Inc. Antibodies to human IL-8 type A receptor
US5919896A (en) * 1991-03-29 1999-07-06 Genentech, Inc. PF4A receptor
US6087475A (en) * 1991-12-19 2000-07-11 Genentech, Inc. PF4A receptor
EP0669979B1 (de) * 1992-11-10 2005-10-12 Genentech, Inc. C-c ckr-1, ein c-c chemokin rezeptor
DE69317883T2 (de) * 1992-11-17 1998-11-12 Icos Corp Neue Sieben-Transmembran-Rezeptor V28
US6107475A (en) * 1992-11-17 2000-08-22 Icos Corporation Seven transmembrane receptors
US6448379B1 (en) * 1993-09-14 2002-09-10 Chiron Corporation IL8 inhibitors
AU1988795A (en) * 1994-03-15 1995-10-03 Repligen Corporation Antibodies to interleukin-8 receptors and methods of use
CA2242908A1 (en) * 1996-01-11 1997-07-17 Human Genome Sciences, Inc. Human g-protein chemokine receptor hsatu68
US7888466B2 (en) 1996-01-11 2011-02-15 Human Genome Sciences, Inc. Human G-protein chemokine receptor HSATU68
US7081360B2 (en) * 1998-07-28 2006-07-25 Cadus Technologies, Inc. Expression of G protein-coupled receptors with altered ligand binding and/or coupling properties
AU2003300396A1 (en) * 2002-12-24 2004-07-22 The Board Of Regents Of The University Of Texas System Chemokine antagonists and uses thereof
WO2005052132A2 (en) * 2003-11-24 2005-06-09 Exelixis, Inc Mbms as modifiers of branching morphogenesis and methods of use
WO2005103702A2 (en) * 2004-04-20 2005-11-03 Bayer Healthcare Ag Diagnostics and therapeutics for diseases associated with cxc chemokine receptor 2 (cxcr2)
TW200700727A (en) * 2005-03-23 2007-01-01 Astrazeneca Ab Screen
WO2012168484A1 (en) * 2011-06-10 2012-12-13 Université Libre de Bruxelles Targets and agents for the treatment of impaired bone fracture healing
RU2676317C1 (ru) * 2017-08-30 2018-12-27 федеральное государственное автономное образовательное учреждение высшего образования Первый Московский государственный медицинский университет имени И.М. Сеченова Министерства здравоохранения Российской Федерации (Сеченовский университет) ГЕНЕТИЧЕСКАЯ КОНСТРУКЦИЯ pCXCRs-Fc ДЛЯ ПОЛУЧЕНИЯ РЕКОМБИНАНТНОГО СЛИТОГО БЕЛКА, ОБЛАДАЮЩЕГО АНТИ-IL-8 АКТИВНОСТЬЮ
KR102049180B1 (ko) * 2018-04-02 2019-11-26 고려대학교 산학협력단 체세포에서 유도 만능 줄기세포로의 역분화 유도용 조성물 및 이를 이용한 역분화 유도방법
WO2019194549A1 (ko) 2018-04-02 2019-10-10 고려대학교 산학협력단 체세포에서 유도 만능 줄기세포로의 역분화 유도용 조성물 및 이를 이용한 역분화 유도방법

Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1992017497A1 (en) * 1991-03-29 1992-10-15 Genentech, Inc. Human pf4a receptors and their use

Patent Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1992017497A1 (en) * 1991-03-29 1992-10-15 Genentech, Inc. Human pf4a receptors and their use

Non-Patent Citations (3)

* Cited by examiner, † Cited by third party
Title
JOURNAL OF BIOLOGICAL CHEMISTRY. vol. 265, no. 33 , November 1990 , BALTIMORE US pages 20061 - 20064 THOMAS, K.M. ET AL.; 'Molecular cloning of the fMet-Leu-Phe receptor from neutrophils.' *
JOURNAL OF BIOLOGICAL CHEMISTRY. vol. 266, no. 23 , 15 August 1991 , BALTIMORE US pages 14839 - 14841 THOMAS, K.M. ET AL.; 'The Interleukin-8 receptor is encoded by a neutrophil-specific cDNA clone, F3R' *
See also references of WO9218641A1 *

Also Published As

Publication number Publication date
EP0579739A4 (de) 1994-03-22
WO1992018641A1 (en) 1992-10-29
JPH06506596A (ja) 1994-07-28
CA2107682A1 (en) 1992-10-11

Similar Documents

Publication Publication Date Title
EP0579739A1 (de) Interleukin-8-rezeptoren, damit in zusammenhang stehende moleküle und verfahrensweisen
US6329197B2 (en) DNA encoding galanin GALR3 receptors and uses thereof
WO1995003821A1 (en) Prosaposin and cytokine-derived peptides as therapeutic agents
CZ208197A3 (en) Side protein of human receptor for interleukin 1
JPH11502420A (ja) 哺乳動物ケモカインccf8およびケモカインレセプターcckr3
SK8752000A3 (en) Novel g protein-coupled receptor
US5892014A (en) DNA encoding a protease-activated receptor 3
KR20010041418A (ko) 프로테아제-활성화되는 수용체 4 및 그것의 사용
JP4208960B2 (ja) オピオイドレセプター遺伝子
WO1992016547A1 (en) Controlling nk-1 receptor-mediated responses and related diagnostics
JPH08502902A (ja) ヒト5−ht▲下2▼レセプター
AU741330B2 (en) Rat ST38.2 chemokine
JP3213946B2 (ja) 哺乳動物のチモカイン遺伝子
US20030166150A1 (en) Asthma associated factors as targets for treating atopic allergies including asthma and related disorders
EP0576609A1 (de) Rezeptoren für bombesin-ähnliche peptide
CA2290713A1 (en) A human edg-6 receptor homologue
WO2001046415A1 (fr) Polypeptides de type tachykinine et utilisation associee
CA2368548A1 (en) Dna molecules encoding human clax proteins and their soluble fusion proteins
WO1995025126A1 (en) Antibodies to interleukin-8 receptors and methods of use
WO1998022497A1 (en) Preprotachykinin-c gene encoding substance z tachykinin precursor
JPH06100595A (ja) Il−8受容体
US20030139589A1 (en) G protein coupled receptor A4
MXPA01006612A (en) Agonists or antagonists of cutaneous t cell-attracting chemokine (ctack) or vasoactive intestinal contractor (vic) chemokines

Legal Events

Date Code Title Description
PUAI Public reference made under article 153(3) epc to a published international application that has entered the european phase

Free format text: ORIGINAL CODE: 0009012

17P Request for examination filed

Effective date: 19931012

AK Designated contracting states

Kind code of ref document: A1

Designated state(s): AT BE CH DE DK ES FR GB GR IT LI LU MC NL SE

RHK1 Main classification (correction)

Ipc: C12N 15/12

STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: THE APPLICATION HAS BEEN WITHDRAWN

18W Application withdrawn

Withdrawal date: 19960307