US20100233186A1 - Novel receptor that binds ctla-8 - Google Patents

Novel receptor that binds ctla-8 Download PDF

Info

Publication number
US20100233186A1
US20100233186A1 US12/722,477 US72247710A US2010233186A1 US 20100233186 A1 US20100233186 A1 US 20100233186A1 US 72247710 A US72247710 A US 72247710A US 2010233186 A1 US2010233186 A1 US 2010233186A1
Authority
US
United States
Prior art keywords
ctla
protein
dna
amino acid
proteins
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Abandoned
Application number
US12/722,477
Inventor
Zhengbin Yao
Melanie K. Spriggs
William C. Fanslow
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.)
Immunex Corp
Amgen K A Inc
Original Assignee
Immunex 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
Family has litigation
First worldwide family litigation filed litigation Critical https://patents.darts-ip.com/?family=27021039&utm_source=google_patent&utm_medium=platform_link&utm_campaign=public_patent_search&patent=US20100233186(A1) "Global patent litigation dataset” by Darts-ip is licensed under a Creative Commons Attribution 4.0 International License.
Priority claimed from US09/549,679 external-priority patent/US6680057B1/en
Application filed by Immunex Corp filed Critical Immunex Corp
Priority to US12/722,477 priority Critical patent/US20100233186A1/en
Publication of US20100233186A1 publication Critical patent/US20100233186A1/en
Assigned to KIRIN-AMGEN, INC. reassignment KIRIN-AMGEN, INC. ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: AMGEN INC.
Abandoned legal-status Critical Current

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/11DNA or RNA fragments; Modified forms thereof; Non-coding nucleic acids having a biological activity
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/52Cytokines; Lymphokines; Interferons
    • C07K14/54Interleukins [IL]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/16Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/16Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • A61K38/17Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • 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
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/02Immunomodulators
    • A61P37/06Immunosuppressants, e.g. drugs for graft rejection
    • 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
    • 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]
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/63Introduction of foreign genetic material using vectors; Vectors; Use of hosts therefor; Regulation of expression
    • C12N15/79Vectors or expression systems specially adapted for eukaryotic hosts
    • C12N15/85Vectors or expression systems specially adapted for eukaryotic hosts for animal cells
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K48/00Medicinal preparations containing genetic material which is inserted into cells of the living body to treat genetic diseases; Gene therapy
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2319/00Fusion polypeptide

Definitions

  • the present invention relates generally to the field of cytokine receptors, and more specifically to cytokine receptor proteins having immunoregulatory activity.
  • Cytokines are hormone-like molecules that regulate various aspects of an immune or inflammatory response. Cytokines exert their effects by specifically binding receptors present on cells, and transducing a signal to the cells.
  • Rouvier et al. J. Immunol. 150:5445; 1993
  • the putative CTLA8 protein is 57% homologous to the predicted amino acid sequence of an open reading frame (ORF) present in Herpesvirus saimiri (HSV) referred to as HVS13 (Nicholas et al. Virol. 179:189, 1990; Albrecht et al., J. Virol. 66:5047; 1992).
  • CTLA-8 or HVS13 the function, if any of either CTLA-8 or HVS13 was not known, nor was a receptor or binding protein for CTLA-8 or HVS13 known.
  • CTLA-8 and HVS13 there was a need in the art to determine the function of CTLA-8 and HVS13, and to identify receptor molecules or binding proteins that play a role in the function of these proteins.
  • the present invention identifies a novel receptor that binds CTLA-8 and HVS13, a viral homolog of CTLA-8; DNAs encoding the novel receptor and novel receptor proteins are provided.
  • the receptor is a Type I transmembrane protein with 864 amino acid residues. Soluble forms of the receptor can be prepared and used to regulate immune responses in a therapeutic setting; accordingly, pharmaceutical compositions comprising soluble forms of the novel receptor are also provided. Deleted forms and fusion proteins comprising the novel receptor, and homologs thereof are also disclosed. Also provided are methods of regulating an immune response.
  • T cell thymoma EL4 cells were found to bind the HVS13/Fc as well as mCTLA8/Fc fusion protein.
  • a cDNA library from EL4 cells was prepared and screened for expression of the receptor.
  • the receptor is a Type I transmembrane protein with 864 amino acid residues, which is referred to as CTLA-8R.
  • CTLA-8R Various forms of CTLA-8R were prepared, including CTLA-8R/Fc protein, a soluble CTLA8R which contains the signal peptide and extracellular domain of CTLA-8R, and a soluble CTLA8/Flag® construct.
  • CTLA-8 refers to a cDNA cloned from an activated T cell hybridoma clone (Rouvier et al., J. Immunol. 150:5445; 1993). Northern blot analysis indicated that CTLA-8 transcription was very tissue specific. The CTLA-8 gene was found to map at chromosomal site 1a in mice, and at 2q31 in humans. Although a protein encoded by the CTLA-8 gene was never identified by Rouvier et al, the predicted amino acid sequence of CTLA-8 was found to be 57% homologous to the predicted amino acid sequence of an ORF present in HVS, HVS13.
  • HVS13 is a late gene which is present in the Hind III-G fragment of HVS. Antisera developed against peptides derived from HVS13 are believed to react with a late protein (Nicholas et al., supra).
  • CTLA-8R and homologs thereof having immunoregulatory activity.
  • Such proteins are substantially free of contaminating endogenous materials and, optionally, without associated native-pattern glycosylation.
  • Derivatives of CTLA-8R within the scope of the invention also include various structural forms of the primary protein which retain biological activity. Due to the presence of ionizable amino and carboxyl groups, for example, a CTLA-8R protein may be in the form of acidic or basic salts, or may be in neutral form. Individual amino acid residues may also be modified by oxidation or reduction.
  • the primary amino acid structure may be modified by forming covalent or aggregative conjugates with other chemical moieties, such as glycosyl groups, lipids, phosphate, acetyl groups and the like, or by creating amino acid sequence mutants.
  • Covalent derivatives are prepared by linking particular functional groups to amino acid side chains or at the N- or C-termini.
  • Soluble forms of CTLA-8R are also within the scope of the invention.
  • the nucleotide and predicted amino acid sequence is shown in SEQ ID NOs:1 and 2.
  • the signal peptide is followed by a 291 amino acid extracellular domain, a 21 amino acid transmembrane domain, and a 521 amino acid cytoplasmic tail.
  • Soluble CTLA-8R comprises the signal peptide and the extracellular domain (residues 1 to 322 of SEQ ID NO:1) or a fragment thereof.
  • a different signal peptide can be substituted for residues 1 through 31 of SEQ ID NO:1.
  • CTLA-8R protein and homologs thereof within the scope of this invention include covalent or aggregative conjugates of the protein or its fragments with other proteins or polypeptides, such as by synthesis in recombinant culture as N-terminal or C-terminal fusions.
  • the conjugated peptide may be a signal (or leader) polypeptide sequence at the N-terminal region of the protein which co-translationally or post-translationally directs transfer of the protein from its site of synthesis to its site of function inside or outside of the cell membrane or wall (e.g., the yeast ⁇ -factor leader).
  • Protein fusions can comprise peptides added to facilitate purification or identification of CTLA-8R proteins and homologs (e.g., poly-His).
  • the amino acid sequence of the inventive proteins can also be linked to an identification peptide such as that described by Hopp et al., Bio/Technology 6:1204 (1988).
  • Such a highly antigenic peptide provides an epitope reversibly bound by a specific monoclonal antibody, enabling rapid assay and facile purification of expressed recombinant protein.
  • the sequence of Hopp et al. is also specifically cleaved by bovine mucosal enterokinase, allowing removal of the peptide from the purified protein. Fusion proteins capped with such peptides may also be resistant to intracellular degradation in E. coli.
  • Fusion proteins further comprise the amino acid sequence of a CTLA-8R linked to an immunoglobulin Fc region.
  • An exemplary Fc region is a human IgG1 having a nucleotide and amino acid sequence set forth in SEQ ID NO:4. Fragments of an Fc region may also be used, as can Fc muteins such as those described in U.S. Ser. No. 08/145,830, filed Oct. 29, 1993.
  • a fusion protein may be expressed as a dimer, through formation of interchain disulfide bonds. If the fusion proteins are made with both heavy and light chains of an antibody, it is possible to form a protein oligomer with as many as four CTLA-8R regions.
  • CTLA-8R and homologs thereof further comprise an oligomerizing zipper domain.
  • Zipper domains are described in U.S. Ser. No. 08/107,353, filed Aug. 13, 1993, the relevant disclosure of which is incorporated by reference herein.
  • leucine zipper domains are those found in the yeast transcription factor GCN4 and a heat-stable DNA-binding protein found in rat liver (C/EBP; Landschulz et al., Science 243:1681, 1989), the nuclear transforming proteins, fos and jun, which preferentially form a heterodimer (O'Shea et al., Science 245:646, 1989; Turner and Tjian, Science 243:1689, 1989), and the gene product of the murine proto-oncogene, c-myc (Landschulz et al., Science 240:1759, 1988).
  • the fusogenic proteins of several different viruses including paramyxovirus, coronavirus, measles virus and many retroviruses, also possess leucine zipper domains (Buckland and Wild, Nature 338:547, 1989; Britton, Nature 353:394, 1991; Delwart and Mosialos, AIDS Research and Human Retroviruses 6:703, 1990).
  • CTLA-8R may also be used as immunogens, reagents in in vitro assays, or as binding agents for affinity purification procedures. Such derivatives may also be obtained by cross-linking agents, such as M-maleimidobenzoyl succinimide ester and N-hydroxysuccinimide, at cysteine and lysine residues.
  • inventive proteins may also be covalently bound through reactive side groups to various insoluble substrates, such as cyanogen bromide-activated, bisoxirane-activated, carbonyldiimidazole-activated or tosyl-activated agarose structures, or by adsorbing to polyolefin surfaces (with or without glutaraldehyde cross-linking).
  • proteins may be used to selectively bind (for purposes of assay or purification) antibodies raised against the CTLA-8R or against other proteins which are similar to the CTLA-8R, as well as other proteins that bind CTLA-8R or its homologous proteins.
  • the present invention also includes CTLA-8R with or without associated native-pattern glycosylation.
  • Proteins expressed in yeast or mammalian expression systems e.g., COS-7 cells, may be similar or slightly different in molecular weight and glycosylation pattern than the native molecules, depending upon the expression system.
  • Expression of DNAs encoding the inventive proteins in bacteria such as E. coli provides non-glycosylated molecules.
  • Functional mutant analogs of CTLA-8R protein or homologs thereof having inactivated N-glycosylation sites can be produced by oligonucleotide synthesis and ligation or by site-specific mutagenesis techniques. These analog proteins can be produced in a homogeneous, reduced-carbohydrate form in good yield using yeast expression systems.
  • N-glycosylation sites in eukaryotic proteins are characterized by the amino acid triplet Asn-A 1 -Z, where A 1 is any amino acid except Pro, and Z is Ser or Thr.
  • asparagine provides a side chain amino group for covalent attachment of carbohydrate.
  • Such a site can be eliminated by substituting another amino acid for Asn or for residue Z, deleting Asn or Z, or inserting a non-Z amino acid between A 1 and Z, or an amino acid other than Asn between Asn and A 1 .
  • CTLA-8R protein derivatives may also be obtained by mutations of the native CTLA-8R or its subunits.
  • a CTLA-8R mutated protein as referred to herein, is a polypeptide homologous to a CTLA-8R protein but which has an amino acid sequence different from the native CTLA-8R because of one or a plurality of deletions, insertions or substitutions.
  • any mutation made in a DNA encoding a CTLA-8R peptide may be easily determined by analyzing the ability of the mutated CTLA-8R peptide to inhibit costimulation of T or B cells by CTLA-8 or homologous proteins, or to bind proteins that specifically bind CTLA-8R (for example, antibodies or proteins encoded by the CTLA-8 cDNA or the HVS13 ORF).
  • activity of CTLA-8R analogs, muteins or derivatives can be determined by any of the assays methods described herein. Similar mutations may be made in homologs of CTLA-8R, and tested in a similar manner.
  • Bioequivalent analogs of the inventive proteins may be constructed by, for example, making various substitutions of residues or sequences or deleting terminal or internal residues or sequences not needed for biological activity.
  • cysteine residues can be deleted or replaced with other amino acids to prevent formation of incorrect intramolecular disulfide bridges upon renaturation.
  • Other approaches to mutagenesis involve modification of adjacent dibasic amino acid residues to enhance expression in yeast systems in which KEX2 protease activity is present.
  • substitutions should be made conservatively; i.e., the most preferred substitute amino acids are those having physicochemical characteristics resembling those of the residue to be replaced.
  • substitutions should be made conservatively; i.e., the most preferred substitute amino acids are those having physicochemical characteristics resembling those of the residue to be replaced.
  • the potential effect of the deletion or insertion on biological activity should be considered.
  • Subunits of viral proteins may be constructed by deleting terminal or internal residues or sequences. Additional guidance as to the types of mutations that can be made is provided by a comparison of the sequence of CTLA-8R to proteins that have similar structures, as well as by performing structural analysis of the inventive proteins.
  • Mutations in nucleotide sequences constructed for expression of analog CTLA-8R must, of course, preserve the reading frame phase of the coding sequences and preferably will not create complementary regions that could hybridize to produce secondary mRNA structures such as loops or hairpins which would adversely affect translation of the receptor mRNA.
  • a mutation site may be predetermined, it is not necessary that the nature of the mutation per se be predetermined. For example, in order to select for optimum characteristics of mutants at a given site, random mutagenesis may be conducted at the target codon and the expressed mutated viral proteins screened for the desired activity.
  • nucleotide sequence which encodes a CTLA-8R protein or homolog thereof will be expressed in the final product, for example, nucleotide substitutions may be made to enhance expression, primarily to avoid secondary structure loops in the transcribed mRNA (see EPA 75,444A, incorporated herein by reference), or to provide codons that are more readily translated by the selected host, e.g., the well-known E. coli preference codons for E. coli expression.
  • Mutations can be introduced at particular loci by synthesizing oligonucleotides containing a mutant sequence, flanked by restriction sites enabling ligation to fragments of the native sequence. Following ligation, the resulting reconstructed sequence encodes an analog having the desired amino acid insertion, substitution, or deletion.
  • oligonucleotide-directed site-specific mutagenesis procedures can be employed to provide an altered gene having particular codons altered according to the substitution, deletion, or insertion required.
  • Exemplary methods of making the alterations set forth above are disclosed by Walder et al. ( Gene 42:133, 1986); Bauer et al. ( Gene 37:73, 1985); Craik ( BioTechniques , January 1985, 12-19); Smith et al. ( Genetic Engineering: Principles and Methods , Plenum Press, 1981); and U.S. Pat. Nos. 4,518,584 and 4,737,462 disclose suitable techniques, and are incorporated by reference herein.
  • CTLA-8R analogs are at least about 70% identical in amino acid sequence to the amino acid sequence of CTLA-8R proteins as set forth in SEQ ID NO:1.
  • analogs of CTLA-8R homologs are at least about 70% identical in amino acid sequence to the amino acid sequence of the native, homologous proteins. In a most preferred embodiment, analogs of CTLA-8R or homologs thereof are at least about 80% identical in amino acid sequence to the native form of the inventive proteins.
  • Percent identity may be determined using a computer program, for example, the GAP computer program described by Devereux et al. ( Nucl. Acids Res. 12:387, 1984) and available from the University of Wisconsin Genetics Computer Group (UWGCG). For fragments derived from the CTLA-8R protein, the identity is calculated based on that portion of the CTLA-8R protein that is present in the fragment. Similar methods can be used to analyze homologs of CTLA-8R.
  • CTLA-8R analogs to bind CTLA-8 can be determined by testing the ability of the analogs to inhibit CTLA-8R-induced T cell proliferation.
  • suitable assays for example, an enzyme immunoassay or a dot blot, employing CTLA-8 or HSV13 (or a homolog thereof which binds native CTLA-8R) can be used to assess the ability of CTLA-8R analogs to bind CTLA-8.
  • Such methods are well known in the art.
  • the proteins of the present invention are preferably produced by recombinant DNA methods by inserting a DNA sequence encoding CTLA-8R protein or a homolog thereof into a recombinant expression vector and expressing the DNA sequence in a recombinant microbial expression system under conditions promoting expression.
  • DNA sequences encoding the proteins provided by this invention can be assembled from cDNA fragments and short oligonucleotide linkers, or from a series of oligonucleotides, to provide a synthetic gene which is capable of being inserted in a recombinant expression vector and expressed in a recombinant transcriptional unit.
  • Recombinant expression vectors include synthetic or cDNA-derived DNA fragments encoding CTLA-8R, homologs, or bioequivalent analogs, operably linked to suitable transcriptional or translational regulatory elements derived from mammalian, microbial, viral or insect genes.
  • suitable transcriptional or translational regulatory elements include a transcriptional promoter, an optional operator sequence to control transcription, a sequence encoding suitable mRNA ribosomal binding sites, and sequences which control the termination of transcription and translation, as described in detail below.
  • the ability to replicate in a host usually conferred by an origin of replication, and a selection gene to facilitate recognition of transformants may additionally be incorporated.
  • DNA regions are operably linked when they are functionally related to each other.
  • DNA for a signal peptide secretory leader
  • DNA for a polypeptide if it is expressed as a precursor which participates in the secretion of the polypeptide
  • a promoter is operably linked to a coding sequence if it controls the transcription of the sequence
  • a ribosome binding site is operably linked to a coding sequence if it is positioned so as to permit translation.
  • operably linked means contiguous and, in the case of secretory leaders, contiguous and in reading frame.
  • DNA sequences encoding CTLA-8R or homologs which are to be expressed in a microorganism will preferably contain no introns that could prematurely terminate transcription of DNA into mRNA.
  • Useful expression vectors for bacterial use can comprise a selectable marker and bacterial origin of replication derived from commercially available plasmids comprising genetic elements of the well known cloning vector pBR322 (ATCC 37017).
  • Such commercial vectors include, for example, pKK223-3 (Pharmacia Fine Chemicals, Uppsala, Sweden) and pGEM1 (Promega Biotec, Madison, Wis., USA). These pBR322 “backbone” sections are combined with an appropriate promoter and the structural sequence to be expressed.
  • E. coli is typically transformed using derivatives of pBR322, a plasmid derived from an E. coli species (Bolivar et al., Gene 2:95, 1977).
  • pBR322 contains genes for ampicillin and tetracycline resistance and thus provides simple means for identifying transformed cells.
  • Promoters commonly used in recombinant microbial expression vectors include the ⁇ -lactamase (penicillinase) and lactose promoter system (Chang et al., Nature 275:615, 1978; and Goeddel et al., Nature 281:544, 1979), the tryptophan (trp) promoter system (Goeddel et al., Nucl. Acids Res. 8:4057, 1980; and EPA 36,776) and tac promoter (Maniatis, Molecular Cloning: A Laboratory Manual , Cold Spring Harbor Laboratory, p. 412, 1982).
  • Plasmid vectors available from the American Type Culture Collection which incorporate derivatives of the ⁇ P L promoter include plasmid pHUB2, resident in E. coli strain JMB9 (ATCC 37092) and pPLc28, resident in E. coli RR1 (ATCC 53082).
  • Suitable promoter sequences in yeast vectors include the promoters for metallothionein, 3-phosphoglycerate kinase (Hitzeman et al., J. Biol. Chem. 255:2073, 1980) or other glycolytic enzymes (Hess et al., J. Adv. Enzyme Reg. 7:149, 1968; and Holland et al., Biochem.
  • enolase such as enolase, glyceraldehyde-3-phosphate dehydrogenase, hexokinase, pyruvate decarboxylase, phosphofructokinase, glucose-6-phosphate isomerase, 3-phosphoglycerate mutase, pyruvate kinase, triosephosphate isomerase, phosphoglucose isomerase, and glucokinase.
  • Suitable vectors and promoters for use in yeast expression are further described in R. Hitzeman et al., EPA 73,657.
  • Preferred yeast vectors can be assembled using DNA sequences from pBR322 for selection and replication in E. coli (Amp r gene and origin of replication) and yeast DNA sequences including a glucose-repressible ADH2 promoter and ⁇ -factor secretion leader.
  • the ADH2 promoter has been described by Russell et al. ( J. Biol. Chem. 258:2674, 1982) and Beier et al. ( Nature 300:724, 1982).
  • the yeast ⁇ -factor leader which directs secretion of heterologous proteins, can be inserted between the promoter and the structural gene to be expressed. See, e.g., Kurjan et al., Cell 30:933, 1982; and Bitter et al., Proc. Natl. Acad. Sci. USA 81:5330, 1984.
  • the leader sequence may be modified to contain, near its 3′ end, one or more useful restriction sites to facilitate fusion of the leader sequence to foreign genes.
  • the transcriptional and translational control sequences in expression vectors to be used in transforming vertebrate cells may be provided by viral sources.
  • viral sources for example, commonly used promoters and enhancers are derived from Polyoma, Adenovirus 2, Simian Virus 40 (SV40), and human cytomegalovirus.
  • DNA sequences derived from the SV40 viral genome for example, SV40 origin, early and late promoter, enhancer, splice, and polyadenylation sites may be used to provide the other genetic elements required for expression of a heterologous DNA sequence.
  • the early and late promoters are particularly useful because both are obtained easily from the virus as a fragment which also contains the SV40 viral origin of replication (Fiers et al., Nature 273:113, 1978).
  • Smaller or larger SV40 fragments may also be used, provided the approximately 250 by sequence extending from the Hind III site toward the BglI site located in the viral origin of replication is included. Further, viral genomic promoter, control and/or signal sequences may be utilized, provided such control sequences are compatible with the host cell chosen.
  • Exemplary vectors can be constructed as disclosed by Okayama and Berg ( Mol. Cell. Biol. 3:280, 1983).
  • a useful system for stable high level expression of mammalian receptor cDNAs in C127 murine mammary epithelial cells can be constructed substantially as described by Cosman et al. ( Mol. Immunol. 23:935, 1986).
  • a preferred eukaryotic vector for expression of CTLA-8R DNA is referred to as pDC406 (McMahan et al., EMBO J. 10:2821, 1991), and includes regulatory sequences derived from SV40, human immunodeficiency virus (HIV), and Epstein-Barr virus (EBV).
  • Other preferred vectors include pDC409 and pDC410, which are derived from pDC406.
  • pDC410 was derived from pDC406 by substituting the EBV origin of replication with sequences encoding the SV40 large T antigen.
  • pDC409 differs from pDC406 in that a Bgl II restriction site outside of the multiple cloning site has been deleted, making the Bgl II site within the multiple cloning site unique.
  • CV-1/EBNA A useful cell line that allows for episomal replication of expression vectors, such as pDC406 and pDC409, which contain the EBV origin of replication, is CV-1/EBNA (ATCC CRL 10478).
  • the CV-1/EBNA cell line was derived by transfection of the CV-1 cell line with a gene encoding Epstein-Barr virus nuclear antigen-1 (EBNA-1) and constitutively express EBNA-1 driven from human CMV immediate-early enhancer/promoter.
  • EBNA-1 Epstein-Barr virus nuclear antigen-1
  • Transformed host cells are cells which have been transformed or transfected with expression vectors constructed using recombinant DNA techniques and which contain sequences encoding the proteins of the present invention.
  • Transformed host cells may express the desired protein (CTLA-8R or homologs thereof), but host cells transformed for purposes of cloning or amplifying the inventive DNA do not need to express the protein.
  • Expressed proteins will preferably be secreted into the culture supernatant, depending on the DNA selected, but may be deposited in the cell membrane.
  • Suitable host cells for expression of viral proteins include prokaryotes, yeast or higher eukaryotic cells under the control of appropriate promoters.
  • Prokaryotes include gram negative or gram positive organisms, for example E. coli or Bacillus spp.
  • Higher eukaryotic cells include established cell lines of mammalian origin as described below. Cell-free translation systems could also be employed to produce viral proteins using RNAs derived from the DNA constructs disclosed herein.
  • Appropriate cloning and expression vectors for use with bacterial, fungal, yeast, and mammalian cellular hosts are described by Pouwels et al. ( Cloning Vectors: A Laboratory Manual , Elsevier, N.Y., 1985), the relevant disclosure of which is hereby incorporated by reference.
  • Prokaryotic expression hosts may be used for expression of CTLA-8R or homologs that do not require extensive proteolytic and disulfide processing.
  • Prokaryotic expression vectors generally comprise one or more phenotypic selectable markers, for example a gene encoding proteins conferring antibiotic resistance or supplying an autotrophic requirement, and an origin of replication recognized by the host to ensure amplification within the host.
  • Suitable prokaryotic hosts for transformation include E. coli, Bacillus subtilis, Salmonella typhimurium , and various species within the genera Pseudomonas, Streptomyces , and Staphylococcus , although others may also be employed as a matter of choice.
  • Recombinant CTLA-8R may also be expressed in yeast hosts, preferably from the Saccharomyces species, such as S. cerevisiae .
  • yeast of other genera, such as Pichia or Kluyveromyces may also be employed.
  • Yeast vectors will generally contain an origin of replication from the 2 ⁇ yeast plasmid or an autonomously replicating sequence (ARS), promoter, DNA encoding the viral protein, sequences for polyadenylation and transcription termination and a selection gene.
  • yeast vectors will include an origin of replication and selectable marker permitting transformation of both yeast and E. coli , e.g., the ampicillin resistance gene of E. coli and S.
  • cerevisiae trp1 gene which provides a selection marker for a mutant strain of yeast lacking the ability to grow in tryptophan, and a promoter derived from a highly expressed yeast gene to induce transcription of a structural sequence downstream.
  • the presence of the trp1 lesion in the yeast host cell genome then provides an effective environment for detecting transformation by growth in the absence of tryptophan.
  • Suitable yeast transformation protocols are known to those of skill in the art; an exemplary technique is described by Hinnen et al., Proc. Natl. Acad. Sci. USA 75:1929, 1978, selecting for Trp + transformants in a selective medium consisting of 0.67% yeast nitrogen base, 0.5% casamino acids, 2% glucose, 10 ⁇ g/ml adenine and 20 ⁇ g/ml uracil.
  • Host strains transformed by vectors comprising the ADH2 promoter may be grown for expression in a rich medium consisting of 1% yeast extract, 2% peptone, and 1% glucose supplemented with 80 ⁇ g/ml adenine and 80 ⁇ g/ml uracil. Derepression of the ADH2 promoter occurs upon exhaustion of medium glucose. Crude yeast supernatants are harvested by filtration and held at 4° C. prior to further purification.
  • Suitable mammalian host cell lines include the COS-7 lines of monkey kidney cells, described by Gluzman ( Cell 23:175, 1981), and other cell lines capable of expressing an appropriate vector including, for example, CV-1/EBNA (ATCC CRL 10478), L cells, C127, 3T3, Chinese hamster ovary (CHO), HeLa and BHK cell lines.
  • CV-1/EBNA ATCC CRL 10478
  • L cells C127, 3T3, Chinese hamster ovary (CHO), HeLa and BHK cell lines.
  • Mammalian expression vectors may comprise nontranscribed elements such as an origin of replication, a suitable promoter and enhancer linked to the gene to be expressed, and other 5′ or 3′ flanking nontranscribed sequences, and 5′ or 3′ nontranslated sequences, such as necessary ribosome binding sites, a polyadenylation site, splice donor and acceptor sites, and transcriptional termination sequences.
  • nontranscribed elements such as an origin of replication, a suitable promoter and enhancer linked to the gene to be expressed, and other 5′ or 3′ flanking nontranscribed sequences, and 5′ or 3′ nontranslated sequences, such as necessary ribosome binding sites, a polyadenylation site, splice donor and acceptor sites, and transcriptional termination sequences.
  • Purified CTLA-8R, homologs, or analogs are prepared by culturing suitable host/vector systems to express the recombinant translation products of the DNAs of the present invention, which are then purified from culture media or cell extracts.
  • suitable host/vector systems to express the recombinant translation products of the DNAs of the present invention, which are then purified from culture media or cell extracts.
  • supernatants from systems which secrete recombinant protein into culture media can be first concentrated using a commercially available protein concentration filter, for example, an Amicon or Millipore Pellicon ultrafiltration unit.
  • a suitable affinity matrix can comprise a counter structure protein or lectin or antibody molecule bound to a suitable support.
  • an anion exchange resin can be employed, for example, a matrix or substrate having pendant diethylaminoethyl (DEAE) groups.
  • the matrices can be acrylamide, agarose, dextran, cellulose or other types commonly employed in protein purification.
  • a cation exchange step can be employed. Suitable cation exchangers include various insoluble matrices comprising sulfopropyl or carboxymethyl groups. Sulfopropyl groups are preferred. Gel filtration chromatography also provides a means of purifying the inventive proteins.
  • Affinity chromatography is a particularly preferred method of purifying CTLA-8R and homologs thereof.
  • a CTLA-8R expressed as a fusion protein comprising an immunoglobulin Fc region can be purified using Protein A or Protein G affinity chromatography.
  • a CTLA-8R protein comprising an oligomerizing zipper domain may be purified on a resin comprising an antibody specific to the oligomerizing zipper domain.
  • Monoclonal antibodies against the CTLA-8R protein may also be useful in affinity chromatography purification, by utilizing methods that are well-known in the art.
  • RP-HPLC reversed-phase high performance liquid chromatography
  • hydrophobic RP-HPLC media e.g., silica gel having pendant methyl or other aliphatic groups
  • Recombinant protein produced in bacterial culture is usually isolated by initial extraction from cell pellets, followed by one or more concentration, salting-out, aqueous ion exchange or size exclusion chromatography steps. Finally, high performance liquid chromatography (HPLC) can be employed for final purification steps.
  • Microbial cells employed in expression of recombinant viral protein can be disrupted by any convenient method, including freeze-thaw cycling, sonication, mechanical disruption, or use of cell lysing agents.
  • Fermentation of yeast which express the inventive protein as a secreted protein greatly simplifies purification.
  • Secreted recombinant protein resulting from a large-scale fermentation can be purified by methods analogous to those disclosed by Urdal et al. ( J. Chromatog. 296:171, 1984).
  • This reference describes two sequential, reversed-phase HPLC steps for purification of recombinant human GM-CSF on a preparative HPLC column.
  • Protein synthesized in recombinant culture is characterized by the presence of cell components, including proteins, in amounts and of a character which depend upon the purification steps taken to recover the inventive protein from the culture.
  • These components ordinarily will be of yeast, prokaryotic or non-human higher eukaryotic origin and preferably are present in innocuous contaminant quantities, on the order of less than about 1 percent by weight.
  • recombinant cell culture enables the production of the inventive proteins free of other proteins which may be normally associated with the proteins as they are found in nature in the species of origin.
  • the present invention provides methods of using therapeutic compositions comprising an effective amount of a protein and a suitable diluent and carrier, and methods for regulating an immune response.
  • CTLA-8R or homologs in conjunction with soluble cytokine receptors or cytokines, or other immunoregulatory molecules is also contemplated.
  • purified protein is administered to a patient, preferably a human, for treatment in a manner appropriate to the indication.
  • CTLA-8R protein compositions administered to regulate immune function can be given by bolus injection, continuous infusion, sustained release from implants, or other suitable technique.
  • a therapeutic agent will be administered in the form of a composition comprising purified CTLA-8R, in conjunction with physiologically acceptable carriers, excipients or diluents. Such carriers will be nontoxic to recipients at the dosages and concentrations employed.
  • the preparation of such protein compositions entails combining the inventive protein with buffers, antioxidants such as ascorbic acid, low molecular weight (less than about 10 residues) polypeptides, proteins, amino acids, carbohydrates including glucose, sucrose or dextrins, chelating agents such as EDTA, glutathione and other stabilizers and excipients.
  • buffers such as ascorbic acid, low molecular weight (less than about 10 residues) polypeptides, proteins, amino acids, carbohydrates including glucose, sucrose or dextrins, chelating agents such as EDTA, glutathione and other stabilizers and excipients.
  • Neutral buffered saline or saline mixed with conspecific serum albumin are exemplary appropriate diluents.
  • product is formulated as a lyophilizate using appropriate excipient solutions (e.g., sucrose) as diluents. Appropriate dosages can be determined in trials. The amount and frequency of administration will depend, of
  • Receptors for CTLA-8 can be administered for the purpose of inhibiting T cell proliferation, or for inhibiting T cell activation. Soluble CTLA-8Rs are thus likely to be useful in preventing or treating organ or graft rejection, autoimmune disease, allergy or asthma. The inventive receptor proteins will also be useful for prevention or treatment of inflammatory disease in which activated T cells play a role. Similarly, HVS13 and homologs thereof stimulate B cell proliferation and immunoglobulin secretion; thus, receptors that bind HVS13 or CTLA-8 will be useful in vivo to inhibit B cell proliferation or immunoglobulin secretion. Receptors for CTLA-8 will also be useful to inhibit the binding of HVS13 or CTLA-8 to cells expressing CTLA-8R.
  • This example describes identification of cells that express a receptor (or counterstructure) for HVS13/mCTLA8.
  • a chimeric protein HVS13 type II Fc consisting of an Fc region of a human immunoglobulin (SEQ ID NO:4) followed by the amino acid 19 to 151 of HVS 13 (SEQ ID NO:8) was prepared.
  • a murine CTLA8/Fc mCTLA8/Fc was constructed by fusing amino acid 22 to 150 of mCTLA8 (SEQ ID NO:6) to the Fc region of human IgG1.
  • a control Fc protein was constructed by a similar method.
  • the HVS13/Fc and mCTLA-8 proteins were expressed and used to identify cell sources by flow cytometry.
  • FACS buffer PBS, 1% FCS and 0.1% NaN 3
  • HVS 13/Fc, mCTLA-8/Fc or control/Fc protein 100 ⁇ l was added at 5 ⁇ g/ml and incubated on ice for 30 min. After washing, the cells were stained with biotin labeled anti human IgG (Fc specific) followed by PE-conjugated streptavidin (Becton Dickson & Co, Mountain View, Calif.) in 100 ⁇ l of FACS buffer.
  • This example describes cloning of the gene that encodes CTLA-8R.
  • an EL4 mammalian expression library was screened by a slide-binding autoradiographic method (Gearing et al., EMBO J. 8:3667, 1989).
  • CV1/EBNA cells were maintained in Dulbecco's modified Eagle's medium (DMEM) containing 10% (v/v) fetal calf serum (FCS) at 37° C. in a humidified atmosphere containing 10% CO 2 and passaged twice weekly.
  • DMEM Dulbecco's modified Eagle's medium
  • FCS fetal calf serum
  • Subconfluent CV1/EBNA cell monolayers on fibronectin-treated chamber slides were transfected by a chloroquine-mediated DEAE-dextran procedure with plasmid DNAs derived from pooled transformants (2,000 transformants per pool) of murine EL4 cDNA library.
  • the CV1/EBNA cells transfected with the murine EL4 cDNA pools were assayed for HVS13/Fc binding two days after transfection using [ 125 I] labeled goat anti-human IgG binding and slide autoradiography.
  • Transfected cell monolayers were washed with binding medium (RPMI 1640 containing 1% bovine serum albumin and 50 mg/ml non-fat dry milk), then incubated with 1 ⁇ g/ml of HVS13/Fc for one hour at room temperature. Cells were washed, incubated with 125 I-labeled goat anti-human IgG (New England nuclear, Cambridge, Mass.).
  • the open reading frame is predicted to encode a type I transmembrane protein of 864 amino acids.
  • the nucleotide and predicted amino acid sequence is shown in SEQ ID NOs:1 and 2.
  • the protein has an N-terminal signal peptide with a cleavage site between amino acid 31 and 32.
  • the signal peptide is followed by a 291 amino acid extracellular domain, a 21 amino acid transmembrane domain, and a 521 amino acid cytoplasmic tail.
  • the predicted molecular weight for this protein is 97.8 kilodaltons with an estimated isoelectric point of 4.85.
  • CTLA-8R/Flag® contains a leader sequence, and the region of CTLA-8R from amino acid 1 to amino acid 322 (SEQ ID NO:1), and the octapeptide referred to as Flag® (SEQ ID NO:3).
  • the construct is prepared essentially as described for other soluble constructs, by ligating a DNA fragment encoding amino acids 1 through 322 of SEQ ID NO:1 (prepared as described in Example 4) into an appropriate expression vector which contains a suitable leader sequence.
  • the resultant DNA construct is transfected into a suitable cell line such as the monkey kidney cell line CV-1/EBNA (ATCC CRL 10478).
  • CTLA-8R/Flag® may be purified using a Flag® antibody affinity column, and analyzed for biological activity using any of the methods described herein.
  • This example describes construction of a CTLA-8R DNA construct to express a CTLA-8R/Fc fusion protein.
  • a soluble form of CTLA-8R fused to the Fc region of human IgG1 was constructed in the mammalian expression vector pDC409 in the following way: A pair of oligonucleotide primers containing a sense sequence and an antisense sequence of CTLA-8R were synthesized. The sense primer contained a Sal I site at the 5′ end of the cDNA and antisense primer contained a Bgl II site and contained the CTLA-8R truncated just before the transmembrane region and a stop codon. A 980 by DNA fragment was amplified from CTLA-8R cDNA.
  • the PCR product was cut with Sal I and Bgl II and used in a three way ligation with a fragment carrying the human IgG1 region cut with Bgl II and Not I into a plasmid (pDC409; see U.S. Ser. No. 08/235,397) previously cut with Sal I and Not I.
  • the encoded insert contained the nucleotides encoding the amino acid sequence of residues 1 to 322 of CTLA-8R (SEQ ID NO:1). The sequence was confirmed by sequencing the whole region.
  • CTLA-8R/Fc expression plasmids were transfected into CV-1/EBNA cells, and supernatants were collected for 1 week.
  • the CTLA-8/Fc fusion proteins were purified on a protein A sepharose column (Pharmacia, Uppsala, Sweden) as described below. Protein concentration was determined by an enzyme-linked immunoadsorbent assay specific for the constant domain of human IgG1 and by BCA analysis (Pharmacia), and purity was confirmed by SDS-polyacrylamide gel electrophoresis analysis followed by silver stain of the gel.
  • CTLA-8R/Fc fusion protein is purified by conventional methods using Protein A or Protein G chromatography. Approximately one liter of culture supernatant containing CTLA-8R/Fc fusion protein is purified by filtering mammalian cell supernatants (e.g., in a 0.45 m filter) and applying filtrate to a protein A/G antibody affinity column (Schleicher and Schuell, Keene, N.H.) at 4° C. at a flow rate of 80 ml/hr for a 1.5 cm ⁇ 12.0 cm column. The column is washed with 0.5 M NaCl in PBS until free protein is not detected in the wash buffer. Finally, the column is washed with PBS. Bound fusion protein is eluted from the column with 25 mM citrate buffer, pH 2.8, and brought to pH 7 with 500 mM Hepes buffer, pH 9.1.
  • a CTLA-8R fusion protein comprising Flag® may also be detected and/or purified using an antibody that binds Flag®, substantially as described in Hopp et al., Bio/Technology 6:1204 (1988). Biological activity is measured by inhibition of CTLA-8 activity in any biological assay which quantifies the co-stimulatory effect of CTLA-8, for example, as described in the Examples herein.
  • This example illustrates the preparation of monoclonal antibodies against CTLA-8R.
  • Preparations of purified recombinant CTLA-8R, for example, or transfected cells expressing high levels of CTLA-8R are employed to generate monoclonal antibodies against CTLA-8R using conventional techniques, such as those disclosed in U.S. Pat. No. 4,411,993.
  • Such antibodies are likely to be useful in interfering with CTLA-8R binding to CTLA-8, as components of diagnostic or research assays for CTLA-8R, or in affinity purification of CTLA-8R.
  • CTLA-8R immunogen is emulsified in an adjuvant (such as complete or incomplete Freund's adjuvant, alum, or another adjuvant, such as Ribi adjuvant R700 (Ribi, Hamilton, Mont.), and injected in amounts ranging from 10-100 ⁇ g subcutaneously into a selected rodent, for example, BALB/c mice or Lewis rats.
  • an adjuvant such as complete or incomplete Freund's adjuvant, alum, or another adjuvant, such as Ribi adjuvant R700 (Ribi, Hamilton, Mont.)
  • Ribi adjuvant R700 Ribi, Hamilton, Mont.
  • Serum samples are periodically taken by retro-orbital bleeding or tail-tip excision for testing by dot-blot assay (antibody sandwich), ELISA (enzyme-linked immunosorbent assay), immunoprecipitation, or other suitable assays, including FACS analysis.
  • dot-blot assay antibody sandwich
  • ELISA enzyme-linked immunosorbent assay
  • immunoprecipitation or other suitable assays, including FACS analysis.
  • positive animals are given an intravenous injection of antigen in saline. Three to four days later, the animals are sacrificed, splenocytes harvested, and fused to a murine myeloma cell line (e.g., NS1 or preferably Ag 8.653 [ATCC CRL 1580]).
  • a murine myeloma cell line e.g., NS1 or preferably Ag 8.653 [ATCC CRL 1580]
  • Hybridoma cell lines generated by this procedure are plated in multiple microtiter plates in a selective medium (for example, one containing hypoxanthine, aminopterin, and thymidine, or HAT) to inhibit proliferation of non-fused cells, myeloma-myeloma hybrids, and splenocyte-splenocyte hybrids.
  • a selective medium for example, one containing hypoxanthine, aminopterin, and thymidine, or HAT
  • Hybridoma clones thus generated can be screened by ELISA for reactivity with CTLA-8R, for example, by adaptations of the techniques disclosed by Engvall et al., Immunochem. 8:871 (1971) and in U.S. Pat. No. 4,703,004.
  • a preferred screening technique is the antibody capture technique described by Beckman et al., J. Immunol. 144:4212 (1990).
  • Positive clones are then injected into the peritoneal cavities of syngeneic rodents to produce ascites containing high concentrations (>1 mg/ml) of anti-CTLA-8R monoclonal antibody.
  • the resulting monoclonal antibody can be purified by ammonium sulfate precipitation followed by gel exclusion chromatography.
  • affinity chromatography based upon binding of antibody to protein A or protein G can also be used, as can affinity chromatography based upon binding to CTLA-8R protein.
  • This example illustrates the ability of CTLA-8R to inhibit the proliferative response of T cells to mitogens.
  • Lymphoid organs were harvested aseptically and cell suspension was created. Splenic and lymph node T cells were isolated from the cell suspension. The purity of the resulting splenic T cell preparations was routinely >95% CD3 + and ⁇ 1% sIgM + .
  • Purified murine splenic T cells (2 ⁇ 10 5 /well) were cultured with either 1% PHA or 1 ⁇ g/ml Con A, and a soluble CTLA-8R was titered into the assay. Proliferation was determined after 3 days with the addition of 1 ⁇ Ci [ 3 H]thymidine. Soluble CTLA-8R/Fc inhibited the mitogen-induced proliferation of purified murine splenic T cells in a dose dependent manner, while a control Fc had no effect on the murine T cell proliferation.
  • This example presents the isolation of a DNA encoding human CTLA-8R by cross species hybridization.
  • a human peripheral blood lymphocyte library was prepared and screened substantially as described in U.S. Ser. No. 08/249,189, using murine CTLA-8R DNA under moderately high stringency conditions. Several clones of varying length were obtained. Initial sequencing data indicated that the human CTLA-8R was approximately 76% identical to murine CTLA-8R at the nucleotide level.

Abstract

Isolated receptors for CTLA-8, DNAs encoding such receptors, and pharmaceutical compositions made therefrom, are disclosed. The isolated receptors can be used to inhibit an immune response.

Description

    CROSS-REFERENCE TO RELATED APPLICATION
  • This application is a continuation of U.S. patent application Ser. No. 11/901,904, filed Sep. 19, 2007, which is a divisional of U.S. patent application Ser. No. 10/742,372, filed Dec. 18, 2003, now U.S. Pat. No. 7,273,843; which is a continuation of U.S. patent application Ser. No. 09/549,679, filed Apr. 14, 2000, now U.S. Pat. No. 6,680,057; which is a continuation of U.S. patent application Ser. No. 09/022,259, filed Feb. 11, 1998, now U.S. Pat. No. 6,191,104; which is a divisional of U.S. patent application Ser. No. 08/620,694, filed Mar. 21, 1996, now U.S. Pat. No. 5,869,286; which is a continuation-in-part of U.S. patent application Ser. No. 08/538,765, filed Aug. 7, 1995, now abandoned; which is a continuation-in-part of U.S. patent application Ser. No. 08/410,535, filed Mar. 23, 1995, now abandoned.
  • TECHNICAL FIELD OF THE INVENTION
  • The present invention relates generally to the field of cytokine receptors, and more specifically to cytokine receptor proteins having immunoregulatory activity.
  • BACKGROUND OF THE INVENTION
  • Cytokines are hormone-like molecules that regulate various aspects of an immune or inflammatory response. Cytokines exert their effects by specifically binding receptors present on cells, and transducing a signal to the cells. Rouvier et al. (J. Immunol. 150:5445; 1993) reported a novel cDNA which they termed CTLA-8. The putative CTLA8 protein is 57% homologous to the predicted amino acid sequence of an open reading frame (ORF) present in Herpesvirus saimiri (HSV) referred to as HVS13 (Nicholas et al. Virol. 179:189, 1990; Albrecht et al., J. Virol. 66:5047; 1992). However, the function, if any of either CTLA-8 or HVS13 was not known, nor was a receptor or binding protein for CTLA-8 or HVS13 known. Thus, prior to the present invention, there was a need in the art to determine the function of CTLA-8 and HVS13, and to identify receptor molecules or binding proteins that play a role in the function of these proteins.
  • SUMMARY OF THE INVENTION
  • The present invention identifies a novel receptor that binds CTLA-8 and HVS13, a viral homolog of CTLA-8; DNAs encoding the novel receptor and novel receptor proteins are provided. The receptor is a Type I transmembrane protein with 864 amino acid residues. Soluble forms of the receptor can be prepared and used to regulate immune responses in a therapeutic setting; accordingly, pharmaceutical compositions comprising soluble forms of the novel receptor are also provided. Deleted forms and fusion proteins comprising the novel receptor, and homologs thereof are also disclosed. Also provided are methods of regulating an immune response. These and other aspects of the present invention will become evident upon reference to the following detailed description of the invention.
  • DETAILED DESCRIPTION OF THE INVENTION
  • A soluble CTLA-8 protein and an ORF present in Herpesvirus saimiri (HVS13) expressed as a fusion protein comprising an immunoglobulin Fc region, and used to screen cells for expression of a receptor for CTLA-8. T cell thymoma EL4 cells were found to bind the HVS13/Fc as well as mCTLA8/Fc fusion protein. A cDNA library from EL4 cells was prepared and screened for expression of the receptor. The receptor is a Type I transmembrane protein with 864 amino acid residues, which is referred to as CTLA-8R. Various forms of CTLA-8R were prepared, including CTLA-8R/Fc protein, a soluble CTLA8R which contains the signal peptide and extracellular domain of CTLA-8R, and a soluble CTLA8/Flag® construct.
  • CTLA-8, HVS13 and Homologous Proteins
  • CTLA-8 refers to a cDNA cloned from an activated T cell hybridoma clone (Rouvier et al., J. Immunol. 150:5445; 1993). Northern blot analysis indicated that CTLA-8 transcription was very tissue specific. The CTLA-8 gene was found to map at chromosomal site 1a in mice, and at 2q31 in humans. Although a protein encoded by the CTLA-8 gene was never identified by Rouvier et al, the predicted amino acid sequence of CTLA-8 was found to be 57% homologous to the predicted amino acid sequence of an ORF present in HVS, HVS13.
  • The complete nucleotide sequence of the genome of HVS has been reported (Albrecht et al., J. Virol. 66:5047; 1992). Additional studies on one of the HVS open reading frames (ORFs), HVS13, are described in Nicholas et al., Virol. 179:1 89; 1990. HVS13 is a late gene which is present in the Hind III-G fragment of HVS. Antisera developed against peptides derived from HVS13 are believed to react with a late protein (Nicholas et al., supra).
  • As described U.S. Ser. No. 08/410,535, filed Mar. 23, 1995, full length murine CTLA-8 protein and a CTLA-8/Fc fusion protein were expressed, tested, and found to act as a costimulus for the proliferation of T cells. Human CTLA-8 was identified by probing a human T cell library using a DNA fragment derived from degenerate PCR; homologs of CTLA-8 are expected to exist in other species as well. A full length HVS13 protein, as well as an HVS13/Fc fusion protein, were also expressed, and found to act in a similar manner to CTLA-8 protein. Moreover, other species of herpesviruses are also likely to encode proteins homologous to that encoded by HVS 13.
  • Proteins and Analogs
  • The present invention provides isolated CTLA-8R and homologs thereof having immunoregulatory activity. Such proteins are substantially free of contaminating endogenous materials and, optionally, without associated native-pattern glycosylation. Derivatives of CTLA-8R within the scope of the invention also include various structural forms of the primary protein which retain biological activity. Due to the presence of ionizable amino and carboxyl groups, for example, a CTLA-8R protein may be in the form of acidic or basic salts, or may be in neutral form. Individual amino acid residues may also be modified by oxidation or reduction.
  • The primary amino acid structure may be modified by forming covalent or aggregative conjugates with other chemical moieties, such as glycosyl groups, lipids, phosphate, acetyl groups and the like, or by creating amino acid sequence mutants. Covalent derivatives are prepared by linking particular functional groups to amino acid side chains or at the N- or C-termini.
  • Soluble forms of CTLA-8R are also within the scope of the invention. The nucleotide and predicted amino acid sequence is shown in SEQ ID NOs:1 and 2. Computer analysis indicated that the protein has an N-terminal signal peptide with a cleavage site between amino acid 31 and 32. The signal peptide is followed by a 291 amino acid extracellular domain, a 21 amino acid transmembrane domain, and a 521 amino acid cytoplasmic tail. Soluble CTLA-8R comprises the signal peptide and the extracellular domain (residues 1 to 322 of SEQ ID NO:1) or a fragment thereof. Alternatively, a different signal peptide can be substituted for residues 1 through 31 of SEQ ID NO:1.
  • Other derivatives of the CTLA-8R protein and homologs thereof within the scope of this invention include covalent or aggregative conjugates of the protein or its fragments with other proteins or polypeptides, such as by synthesis in recombinant culture as N-terminal or C-terminal fusions. For example, the conjugated peptide may be a signal (or leader) polypeptide sequence at the N-terminal region of the protein which co-translationally or post-translationally directs transfer of the protein from its site of synthesis to its site of function inside or outside of the cell membrane or wall (e.g., the yeast α-factor leader).
  • Protein fusions can comprise peptides added to facilitate purification or identification of CTLA-8R proteins and homologs (e.g., poly-His). The amino acid sequence of the inventive proteins can also be linked to an identification peptide such as that described by Hopp et al., Bio/Technology 6:1204 (1988). Such a highly antigenic peptide provides an epitope reversibly bound by a specific monoclonal antibody, enabling rapid assay and facile purification of expressed recombinant protein. The sequence of Hopp et al. is also specifically cleaved by bovine mucosal enterokinase, allowing removal of the peptide from the purified protein. Fusion proteins capped with such peptides may also be resistant to intracellular degradation in E. coli.
  • Fusion proteins further comprise the amino acid sequence of a CTLA-8R linked to an immunoglobulin Fc region. An exemplary Fc region is a human IgG1 having a nucleotide and amino acid sequence set forth in SEQ ID NO:4. Fragments of an Fc region may also be used, as can Fc muteins such as those described in U.S. Ser. No. 08/145,830, filed Oct. 29, 1993. Depending on the portion of the Fc region used, a fusion protein may be expressed as a dimer, through formation of interchain disulfide bonds. If the fusion proteins are made with both heavy and light chains of an antibody, it is possible to form a protein oligomer with as many as four CTLA-8R regions.
  • In another embodiment, CTLA-8R and homologs thereof further comprise an oligomerizing zipper domain. Zipper domains are described in U.S. Ser. No. 08/107,353, filed Aug. 13, 1993, the relevant disclosure of which is incorporated by reference herein. Examples of leucine zipper domains are those found in the yeast transcription factor GCN4 and a heat-stable DNA-binding protein found in rat liver (C/EBP; Landschulz et al., Science 243:1681, 1989), the nuclear transforming proteins, fos and jun, which preferentially form a heterodimer (O'Shea et al., Science 245:646, 1989; Turner and Tjian, Science 243:1689, 1989), and the gene product of the murine proto-oncogene, c-myc (Landschulz et al., Science 240:1759, 1988). The fusogenic proteins of several different viruses, including paramyxovirus, coronavirus, measles virus and many retroviruses, also possess leucine zipper domains (Buckland and Wild, Nature 338:547, 1989; Britton, Nature 353:394, 1991; Delwart and Mosialos, AIDS Research and Human Retroviruses 6:703, 1990).
  • Derivatives of CTLA-8R may also be used as immunogens, reagents in in vitro assays, or as binding agents for affinity purification procedures. Such derivatives may also be obtained by cross-linking agents, such as M-maleimidobenzoyl succinimide ester and N-hydroxysuccinimide, at cysteine and lysine residues. The inventive proteins may also be covalently bound through reactive side groups to various insoluble substrates, such as cyanogen bromide-activated, bisoxirane-activated, carbonyldiimidazole-activated or tosyl-activated agarose structures, or by adsorbing to polyolefin surfaces (with or without glutaraldehyde cross-linking). Once bound to a substrate, proteins may be used to selectively bind (for purposes of assay or purification) antibodies raised against the CTLA-8R or against other proteins which are similar to the CTLA-8R, as well as other proteins that bind CTLA-8R or its homologous proteins.
  • The present invention also includes CTLA-8R with or without associated native-pattern glycosylation. Proteins expressed in yeast or mammalian expression systems, e.g., COS-7 cells, may be similar or slightly different in molecular weight and glycosylation pattern than the native molecules, depending upon the expression system. Expression of DNAs encoding the inventive proteins in bacteria such as E. coli provides non-glycosylated molecules. Functional mutant analogs of CTLA-8R protein or homologs thereof having inactivated N-glycosylation sites can be produced by oligonucleotide synthesis and ligation or by site-specific mutagenesis techniques. These analog proteins can be produced in a homogeneous, reduced-carbohydrate form in good yield using yeast expression systems. N-glycosylation sites in eukaryotic proteins are characterized by the amino acid triplet Asn-A1-Z, where A1 is any amino acid except Pro, and Z is Ser or Thr. In this sequence, asparagine provides a side chain amino group for covalent attachment of carbohydrate. Such a site can be eliminated by substituting another amino acid for Asn or for residue Z, deleting Asn or Z, or inserting a non-Z amino acid between A1 and Z, or an amino acid other than Asn between Asn and A1.
  • CTLA-8R protein derivatives may also be obtained by mutations of the native CTLA-8R or its subunits. A CTLA-8R mutated protein, as referred to herein, is a polypeptide homologous to a CTLA-8R protein but which has an amino acid sequence different from the native CTLA-8R because of one or a plurality of deletions, insertions or substitutions. The effect of any mutation made in a DNA encoding a CTLA-8R peptide may be easily determined by analyzing the ability of the mutated CTLA-8R peptide to inhibit costimulation of T or B cells by CTLA-8 or homologous proteins, or to bind proteins that specifically bind CTLA-8R (for example, antibodies or proteins encoded by the CTLA-8 cDNA or the HVS13 ORF). Moreover, activity of CTLA-8R analogs, muteins or derivatives can be determined by any of the assays methods described herein. Similar mutations may be made in homologs of CTLA-8R, and tested in a similar manner.
  • Bioequivalent analogs of the inventive proteins may be constructed by, for example, making various substitutions of residues or sequences or deleting terminal or internal residues or sequences not needed for biological activity. For example, cysteine residues can be deleted or replaced with other amino acids to prevent formation of incorrect intramolecular disulfide bridges upon renaturation. Other approaches to mutagenesis involve modification of adjacent dibasic amino acid residues to enhance expression in yeast systems in which KEX2 protease activity is present.
  • Generally, substitutions should be made conservatively; i.e., the most preferred substitute amino acids are those having physicochemical characteristics resembling those of the residue to be replaced. Similarly, when a deletion or insertion strategy is adopted, the potential effect of the deletion or insertion on biological activity should be considered. Subunits of viral proteins may be constructed by deleting terminal or internal residues or sequences. Additional guidance as to the types of mutations that can be made is provided by a comparison of the sequence of CTLA-8R to proteins that have similar structures, as well as by performing structural analysis of the inventive proteins.
  • Mutations in nucleotide sequences constructed for expression of analog CTLA-8R must, of course, preserve the reading frame phase of the coding sequences and preferably will not create complementary regions that could hybridize to produce secondary mRNA structures such as loops or hairpins which would adversely affect translation of the receptor mRNA. Although a mutation site may be predetermined, it is not necessary that the nature of the mutation per se be predetermined. For example, in order to select for optimum characteristics of mutants at a given site, random mutagenesis may be conducted at the target codon and the expressed mutated viral proteins screened for the desired activity.
  • Not all mutations in the nucleotide sequence which encodes a CTLA-8R protein or homolog thereof will be expressed in the final product, for example, nucleotide substitutions may be made to enhance expression, primarily to avoid secondary structure loops in the transcribed mRNA (see EPA 75,444A, incorporated herein by reference), or to provide codons that are more readily translated by the selected host, e.g., the well-known E. coli preference codons for E. coli expression.
  • Mutations can be introduced at particular loci by synthesizing oligonucleotides containing a mutant sequence, flanked by restriction sites enabling ligation to fragments of the native sequence. Following ligation, the resulting reconstructed sequence encodes an analog having the desired amino acid insertion, substitution, or deletion.
  • Alternatively, oligonucleotide-directed site-specific mutagenesis procedures can be employed to provide an altered gene having particular codons altered according to the substitution, deletion, or insertion required. Exemplary methods of making the alterations set forth above are disclosed by Walder et al. (Gene 42:133, 1986); Bauer et al. (Gene 37:73, 1985); Craik (BioTechniques, January 1985, 12-19); Smith et al. (Genetic Engineering: Principles and Methods, Plenum Press, 1981); and U.S. Pat. Nos. 4,518,584 and 4,737,462 disclose suitable techniques, and are incorporated by reference herein.
  • Due to code degeneracy, there can be considerable variation in nucleotide sequences encoding the same amino acid sequence. Other embodiments include sequences capable of hybridizing under moderately stringent conditions (prewashing solution of 5×SSC, 0.5% SDS, 1.0 mM EDTA (pH 8.0) and hybridization conditions of 50° C., 5×SSC, overnight) to the DNA sequences encoding CTLA-8R, and other sequences which are degenerate to those which encode the CTLA-8R. In a preferred embodiment, CTLA-8R analogs are at least about 70% identical in amino acid sequence to the amino acid sequence of CTLA-8R proteins as set forth in SEQ ID NO:1. Similarly, analogs of CTLA-8R homologs are at least about 70% identical in amino acid sequence to the amino acid sequence of the native, homologous proteins. In a most preferred embodiment, analogs of CTLA-8R or homologs thereof are at least about 80% identical in amino acid sequence to the native form of the inventive proteins.
  • Percent identity may be determined using a computer program, for example, the GAP computer program described by Devereux et al. (Nucl. Acids Res. 12:387, 1984) and available from the University of Wisconsin Genetics Computer Group (UWGCG). For fragments derived from the CTLA-8R protein, the identity is calculated based on that portion of the CTLA-8R protein that is present in the fragment. Similar methods can be used to analyze homologs of CTLA-8R.
  • The ability of CTLA-8R analogs to bind CTLA-8 can be determined by testing the ability of the analogs to inhibit CTLA-8R-induced T cell proliferation. Alternatively, suitable assays, for example, an enzyme immunoassay or a dot blot, employing CTLA-8 or HSV13 (or a homolog thereof which binds native CTLA-8R) can be used to assess the ability of CTLA-8R analogs to bind CTLA-8. Such methods are well known in the art.
  • Expression of Recombinant Receptors for CTLA-8
  • The proteins of the present invention are preferably produced by recombinant DNA methods by inserting a DNA sequence encoding CTLA-8R protein or a homolog thereof into a recombinant expression vector and expressing the DNA sequence in a recombinant microbial expression system under conditions promoting expression. DNA sequences encoding the proteins provided by this invention can be assembled from cDNA fragments and short oligonucleotide linkers, or from a series of oligonucleotides, to provide a synthetic gene which is capable of being inserted in a recombinant expression vector and expressed in a recombinant transcriptional unit.
  • Recombinant expression vectors include synthetic or cDNA-derived DNA fragments encoding CTLA-8R, homologs, or bioequivalent analogs, operably linked to suitable transcriptional or translational regulatory elements derived from mammalian, microbial, viral or insect genes. Such regulatory elements include a transcriptional promoter, an optional operator sequence to control transcription, a sequence encoding suitable mRNA ribosomal binding sites, and sequences which control the termination of transcription and translation, as described in detail below. The ability to replicate in a host, usually conferred by an origin of replication, and a selection gene to facilitate recognition of transformants may additionally be incorporated.
  • DNA regions are operably linked when they are functionally related to each other. For example, DNA for a signal peptide (secretory leader) is operably linked to DNA for a polypeptide if it is expressed as a precursor which participates in the secretion of the polypeptide; a promoter is operably linked to a coding sequence if it controls the transcription of the sequence; or a ribosome binding site is operably linked to a coding sequence if it is positioned so as to permit translation. Generally, operably linked means contiguous and, in the case of secretory leaders, contiguous and in reading frame. DNA sequences encoding CTLA-8R or homologs which are to be expressed in a microorganism will preferably contain no introns that could prematurely terminate transcription of DNA into mRNA.
  • Useful expression vectors for bacterial use can comprise a selectable marker and bacterial origin of replication derived from commercially available plasmids comprising genetic elements of the well known cloning vector pBR322 (ATCC 37017). Such commercial vectors include, for example, pKK223-3 (Pharmacia Fine Chemicals, Uppsala, Sweden) and pGEM1 (Promega Biotec, Madison, Wis., USA). These pBR322 “backbone” sections are combined with an appropriate promoter and the structural sequence to be expressed. E. coli is typically transformed using derivatives of pBR322, a plasmid derived from an E. coli species (Bolivar et al., Gene 2:95, 1977). pBR322 contains genes for ampicillin and tetracycline resistance and thus provides simple means for identifying transformed cells.
  • Promoters commonly used in recombinant microbial expression vectors include the β-lactamase (penicillinase) and lactose promoter system (Chang et al., Nature 275:615, 1978; and Goeddel et al., Nature 281:544, 1979), the tryptophan (trp) promoter system (Goeddel et al., Nucl. Acids Res. 8:4057, 1980; and EPA 36,776) and tac promoter (Maniatis, Molecular Cloning: A Laboratory Manual, Cold Spring Harbor Laboratory, p. 412, 1982). A particularly useful bacterial expression system employs the phage λPL promoter and cI857ts thermolabile repressor. Plasmid vectors available from the American Type Culture Collection which incorporate derivatives of the λPL promoter include plasmid pHUB2, resident in E. coli strain JMB9 (ATCC 37092) and pPLc28, resident in E. coli RR1 (ATCC 53082).
  • Suitable promoter sequences in yeast vectors include the promoters for metallothionein, 3-phosphoglycerate kinase (Hitzeman et al., J. Biol. Chem. 255:2073, 1980) or other glycolytic enzymes (Hess et al., J. Adv. Enzyme Reg. 7:149, 1968; and Holland et al., Biochem. 17:4900, 1978), such as enolase, glyceraldehyde-3-phosphate dehydrogenase, hexokinase, pyruvate decarboxylase, phosphofructokinase, glucose-6-phosphate isomerase, 3-phosphoglycerate mutase, pyruvate kinase, triosephosphate isomerase, phosphoglucose isomerase, and glucokinase. Suitable vectors and promoters for use in yeast expression are further described in R. Hitzeman et al., EPA 73,657.
  • Preferred yeast vectors can be assembled using DNA sequences from pBR322 for selection and replication in E. coli (Ampr gene and origin of replication) and yeast DNA sequences including a glucose-repressible ADH2 promoter and α-factor secretion leader. The ADH2 promoter has been described by Russell et al. (J. Biol. Chem. 258:2674, 1982) and Beier et al. (Nature 300:724, 1982). The yeast α-factor leader, which directs secretion of heterologous proteins, can be inserted between the promoter and the structural gene to be expressed. See, e.g., Kurjan et al., Cell 30:933, 1982; and Bitter et al., Proc. Natl. Acad. Sci. USA 81:5330, 1984. The leader sequence may be modified to contain, near its 3′ end, one or more useful restriction sites to facilitate fusion of the leader sequence to foreign genes.
  • The transcriptional and translational control sequences in expression vectors to be used in transforming vertebrate cells may be provided by viral sources. For example, commonly used promoters and enhancers are derived from Polyoma, Adenovirus 2, Simian Virus 40 (SV40), and human cytomegalovirus. DNA sequences derived from the SV40 viral genome, for example, SV40 origin, early and late promoter, enhancer, splice, and polyadenylation sites may be used to provide the other genetic elements required for expression of a heterologous DNA sequence. The early and late promoters are particularly useful because both are obtained easily from the virus as a fragment which also contains the SV40 viral origin of replication (Fiers et al., Nature 273:113, 1978). Smaller or larger SV40 fragments may also be used, provided the approximately 250 by sequence extending from the Hind III site toward the BglI site located in the viral origin of replication is included. Further, viral genomic promoter, control and/or signal sequences may be utilized, provided such control sequences are compatible with the host cell chosen. Exemplary vectors can be constructed as disclosed by Okayama and Berg (Mol. Cell. Biol. 3:280, 1983).
  • A useful system for stable high level expression of mammalian receptor cDNAs in C127 murine mammary epithelial cells can be constructed substantially as described by Cosman et al. (Mol. Immunol. 23:935, 1986). A preferred eukaryotic vector for expression of CTLA-8R DNA is referred to as pDC406 (McMahan et al., EMBO J. 10:2821, 1991), and includes regulatory sequences derived from SV40, human immunodeficiency virus (HIV), and Epstein-Barr virus (EBV). Other preferred vectors include pDC409 and pDC410, which are derived from pDC406. pDC410 was derived from pDC406 by substituting the EBV origin of replication with sequences encoding the SV40 large T antigen. pDC409 differs from pDC406 in that a Bgl II restriction site outside of the multiple cloning site has been deleted, making the Bgl II site within the multiple cloning site unique.
  • A useful cell line that allows for episomal replication of expression vectors, such as pDC406 and pDC409, which contain the EBV origin of replication, is CV-1/EBNA (ATCC CRL 10478). The CV-1/EBNA cell line was derived by transfection of the CV-1 cell line with a gene encoding Epstein-Barr virus nuclear antigen-1 (EBNA-1) and constitutively express EBNA-1 driven from human CMV immediate-early enhancer/promoter.
  • Host Cells
  • Transformed host cells are cells which have been transformed or transfected with expression vectors constructed using recombinant DNA techniques and which contain sequences encoding the proteins of the present invention. Transformed host cells may express the desired protein (CTLA-8R or homologs thereof), but host cells transformed for purposes of cloning or amplifying the inventive DNA do not need to express the protein. Expressed proteins will preferably be secreted into the culture supernatant, depending on the DNA selected, but may be deposited in the cell membrane.
  • Suitable host cells for expression of viral proteins include prokaryotes, yeast or higher eukaryotic cells under the control of appropriate promoters. Prokaryotes include gram negative or gram positive organisms, for example E. coli or Bacillus spp. Higher eukaryotic cells include established cell lines of mammalian origin as described below. Cell-free translation systems could also be employed to produce viral proteins using RNAs derived from the DNA constructs disclosed herein. Appropriate cloning and expression vectors for use with bacterial, fungal, yeast, and mammalian cellular hosts are described by Pouwels et al. (Cloning Vectors: A Laboratory Manual, Elsevier, N.Y., 1985), the relevant disclosure of which is hereby incorporated by reference.
  • Prokaryotic expression hosts may be used for expression of CTLA-8R or homologs that do not require extensive proteolytic and disulfide processing. Prokaryotic expression vectors generally comprise one or more phenotypic selectable markers, for example a gene encoding proteins conferring antibiotic resistance or supplying an autotrophic requirement, and an origin of replication recognized by the host to ensure amplification within the host. Suitable prokaryotic hosts for transformation include E. coli, Bacillus subtilis, Salmonella typhimurium, and various species within the genera Pseudomonas, Streptomyces, and Staphylococcus, although others may also be employed as a matter of choice.
  • Recombinant CTLA-8R may also be expressed in yeast hosts, preferably from the Saccharomyces species, such as S. cerevisiae. Yeast of other genera, such as Pichia or Kluyveromyces may also be employed. Yeast vectors will generally contain an origin of replication from the 2μ yeast plasmid or an autonomously replicating sequence (ARS), promoter, DNA encoding the viral protein, sequences for polyadenylation and transcription termination and a selection gene. Preferably, yeast vectors will include an origin of replication and selectable marker permitting transformation of both yeast and E. coli, e.g., the ampicillin resistance gene of E. coli and S. cerevisiae trp1 gene, which provides a selection marker for a mutant strain of yeast lacking the ability to grow in tryptophan, and a promoter derived from a highly expressed yeast gene to induce transcription of a structural sequence downstream. The presence of the trp1 lesion in the yeast host cell genome then provides an effective environment for detecting transformation by growth in the absence of tryptophan.
  • Suitable yeast transformation protocols are known to those of skill in the art; an exemplary technique is described by Hinnen et al., Proc. Natl. Acad. Sci. USA 75:1929, 1978, selecting for Trp+ transformants in a selective medium consisting of 0.67% yeast nitrogen base, 0.5% casamino acids, 2% glucose, 10 μg/ml adenine and 20 μg/ml uracil. Host strains transformed by vectors comprising the ADH2 promoter may be grown for expression in a rich medium consisting of 1% yeast extract, 2% peptone, and 1% glucose supplemented with 80 μg/ml adenine and 80 μg/ml uracil. Derepression of the ADH2 promoter occurs upon exhaustion of medium glucose. Crude yeast supernatants are harvested by filtration and held at 4° C. prior to further purification.
  • Various mammalian or insect cell culture systems can be employed to express recombinant protein. Baculovirus systems for production of heterologous proteins in insect cells are reviewed by Luckow and Summers, Bio/Technology 6:47 (1988). Examples of suitable mammalian host cell lines include the COS-7 lines of monkey kidney cells, described by Gluzman (Cell 23:175, 1981), and other cell lines capable of expressing an appropriate vector including, for example, CV-1/EBNA (ATCC CRL 10478), L cells, C127, 3T3, Chinese hamster ovary (CHO), HeLa and BHK cell lines. Mammalian expression vectors may comprise nontranscribed elements such as an origin of replication, a suitable promoter and enhancer linked to the gene to be expressed, and other 5′ or 3′ flanking nontranscribed sequences, and 5′ or 3′ nontranslated sequences, such as necessary ribosome binding sites, a polyadenylation site, splice donor and acceptor sites, and transcriptional termination sequences.
  • Purification of Receptors for CTLA-8
  • Purified CTLA-8R, homologs, or analogs are prepared by culturing suitable host/vector systems to express the recombinant translation products of the DNAs of the present invention, which are then purified from culture media or cell extracts. For example, supernatants from systems which secrete recombinant protein into culture media can be first concentrated using a commercially available protein concentration filter, for example, an Amicon or Millipore Pellicon ultrafiltration unit.
  • Following the concentration step, the concentrate can be applied to a suitable purification matrix. For example, a suitable affinity matrix can comprise a counter structure protein or lectin or antibody molecule bound to a suitable support. Alternatively, an anion exchange resin can be employed, for example, a matrix or substrate having pendant diethylaminoethyl (DEAE) groups. The matrices can be acrylamide, agarose, dextran, cellulose or other types commonly employed in protein purification. Alternatively, a cation exchange step can be employed. Suitable cation exchangers include various insoluble matrices comprising sulfopropyl or carboxymethyl groups. Sulfopropyl groups are preferred. Gel filtration chromatography also provides a means of purifying the inventive proteins.
  • Affinity chromatography is a particularly preferred method of purifying CTLA-8R and homologs thereof. For example, a CTLA-8R expressed as a fusion protein comprising an immunoglobulin Fc region can be purified using Protein A or Protein G affinity chromatography. Moreover, a CTLA-8R protein comprising an oligomerizing zipper domain may be purified on a resin comprising an antibody specific to the oligomerizing zipper domain. Monoclonal antibodies against the CTLA-8R protein may also be useful in affinity chromatography purification, by utilizing methods that are well-known in the art.
  • Finally, one or more reversed-phase high performance liquid chromatography (RP-HPLC) steps employing hydrophobic RP-HPLC media, e.g., silica gel having pendant methyl or other aliphatic groups, can be employed to further purify a CTLA-8R composition. Some or all of the foregoing purification steps, in various combinations, can also be employed to provide a homogeneous recombinant protein.
  • Recombinant protein produced in bacterial culture is usually isolated by initial extraction from cell pellets, followed by one or more concentration, salting-out, aqueous ion exchange or size exclusion chromatography steps. Finally, high performance liquid chromatography (HPLC) can be employed for final purification steps. Microbial cells employed in expression of recombinant viral protein can be disrupted by any convenient method, including freeze-thaw cycling, sonication, mechanical disruption, or use of cell lysing agents.
  • Fermentation of yeast which express the inventive protein as a secreted protein greatly simplifies purification. Secreted recombinant protein resulting from a large-scale fermentation can be purified by methods analogous to those disclosed by Urdal et al. (J. Chromatog. 296:171, 1984). This reference describes two sequential, reversed-phase HPLC steps for purification of recombinant human GM-CSF on a preparative HPLC column.
  • Protein synthesized in recombinant culture is characterized by the presence of cell components, including proteins, in amounts and of a character which depend upon the purification steps taken to recover the inventive protein from the culture. These components ordinarily will be of yeast, prokaryotic or non-human higher eukaryotic origin and preferably are present in innocuous contaminant quantities, on the order of less than about 1 percent by weight. Further, recombinant cell culture enables the production of the inventive proteins free of other proteins which may be normally associated with the proteins as they are found in nature in the species of origin.
  • Administration of CTLA-8R Compositions
  • The present invention provides methods of using therapeutic compositions comprising an effective amount of a protein and a suitable diluent and carrier, and methods for regulating an immune response. The use of CTLA-8R or homologs in conjunction with soluble cytokine receptors or cytokines, or other immunoregulatory molecules is also contemplated.
  • For therapeutic use, purified protein is administered to a patient, preferably a human, for treatment in a manner appropriate to the indication. Thus, for example, CTLA-8R protein compositions administered to regulate immune function can be given by bolus injection, continuous infusion, sustained release from implants, or other suitable technique. Typically, a therapeutic agent will be administered in the form of a composition comprising purified CTLA-8R, in conjunction with physiologically acceptable carriers, excipients or diluents. Such carriers will be nontoxic to recipients at the dosages and concentrations employed.
  • Ordinarily, the preparation of such protein compositions entails combining the inventive protein with buffers, antioxidants such as ascorbic acid, low molecular weight (less than about 10 residues) polypeptides, proteins, amino acids, carbohydrates including glucose, sucrose or dextrins, chelating agents such as EDTA, glutathione and other stabilizers and excipients. Neutral buffered saline or saline mixed with conspecific serum albumin are exemplary appropriate diluents. Preferably, product is formulated as a lyophilizate using appropriate excipient solutions (e.g., sucrose) as diluents. Appropriate dosages can be determined in trials. The amount and frequency of administration will depend, of course, on such factors as the nature and severity of the indication being treated, the desired response, the condition of the patient, and so forth.
  • Receptors for CTLA-8 can be administered for the purpose of inhibiting T cell proliferation, or for inhibiting T cell activation. Soluble CTLA-8Rs are thus likely to be useful in preventing or treating organ or graft rejection, autoimmune disease, allergy or asthma. The inventive receptor proteins will also be useful for prevention or treatment of inflammatory disease in which activated T cells play a role. Similarly, HVS13 and homologs thereof stimulate B cell proliferation and immunoglobulin secretion; thus, receptors that bind HVS13 or CTLA-8 will be useful in vivo to inhibit B cell proliferation or immunoglobulin secretion. Receptors for CTLA-8 will also be useful to inhibit the binding of HVS13 or CTLA-8 to cells expressing CTLA-8R.
  • The following examples are offered by way of illustration, and not by way of limitation. Those skilled in the art will recognize that variations of the invention embodied in the examples can be made, especially in light of the teachings of the various references cited herein, the disclosures of which are incorporated by reference.
  • Example 1
  • This example describes identification of cells that express a receptor (or counterstructure) for HVS13/mCTLA8. A chimeric protein (HVS13 type II Fc) consisting of an Fc region of a human immunoglobulin (SEQ ID NO:4) followed by the amino acid 19 to 151 of HVS 13 (SEQ ID NO:8) was prepared. A murine CTLA8/Fc (mCTLA8/Fc) was constructed by fusing amino acid 22 to 150 of mCTLA8 (SEQ ID NO:6) to the Fc region of human IgG1. A control Fc protein was constructed by a similar method. The HVS13/Fc and mCTLA-8 proteins were expressed and used to identify cell sources by flow cytometry.
  • Cells (1×106) were preincubated on ice for 30 minutes in 100 μl of FACS buffer (PBS, 1% FCS and 0.1% NaN3) containing 2% normal goat serum and 2% normal rabbit serum to block nonspecific binding. 100 μl of HVS 13/Fc, mCTLA-8/Fc or control/Fc protein was added at 5 μg/ml and incubated on ice for 30 min. After washing, the cells were stained with biotin labeled anti human IgG (Fc specific) followed by PE-conjugated streptavidin (Becton Dickson & Co, Mountain View, Calif.) in 100 μl of FACS buffer. Cells were then washed and analyzed using a FACScan (Becton Dickinson). A minimum of 5,000 cells were analyzed for each sample. More than a dozen cell lines were screened and it was found that both HVS13/Fc and mCTLA8/Fc fusion proteins bound specifically to the murine thymoma cell line EL4. These cells did not bind to the control/Fc fusion protein.
  • Example 2
  • This example describes cloning of the gene that encodes CTLA-8R. After identification of a source for HVS13 counterstructure, an EL4 mammalian expression library was screened by a slide-binding autoradiographic method (Gearing et al., EMBO J. 8:3667, 1989). CV1/EBNA cells were maintained in Dulbecco's modified Eagle's medium (DMEM) containing 10% (v/v) fetal calf serum (FCS) at 37° C. in a humidified atmosphere containing 10% CO2 and passaged twice weekly. Subconfluent CV1/EBNA cell monolayers on fibronectin-treated chamber slides (Labtek) were transfected by a chloroquine-mediated DEAE-dextran procedure with plasmid DNAs derived from pooled transformants (2,000 transformants per pool) of murine EL4 cDNA library.
  • The CV1/EBNA cells transfected with the murine EL4 cDNA pools were assayed for HVS13/Fc binding two days after transfection using [125I] labeled goat anti-human IgG binding and slide autoradiography. Transfected cell monolayers were washed with binding medium (RPMI 1640 containing 1% bovine serum albumin and 50 mg/ml non-fat dry milk), then incubated with 1 μg/ml of HVS13/Fc for one hour at room temperature. Cells were washed, incubated with 125I-labeled goat anti-human IgG (New England nuclear, Cambridge, Mass.). Cells were washed twice with binding medium, three times with PBS, and fixed in PBS containing 2.5% gluteraldehyde for 30 minutes, washed twice more with PBS and air dried. The chamber slides were then dipped in Kodak GTNB-2 photographic emulsion and exposed for 3 days at 4° C. before developing.
  • Forty pools of approximately 2,000 cDNA each were transfected into CV1/EBNA cells. Two pools of cDNA were found to confer binding to HVS13/Fc protein. These pools were broken down to pools of 100 cDNAs, and subsequently to individual clones. Two single cDNA clones were isolated. These clones were transfected into CV1/EBNA to determine whether the protein encoded thereby conferred binding to both HVS13/Fc and mCTLA8/Fc. Both HVS/Fc and mCTLA8/Fc bound to CV1/EBNA cells transfected with the cloned cDNA, but not to cells transfected with empty vector. Control/Fc did not bind to either of them.
  • Sequencing of these clones found that they contained a 3.2 kb and 1.7 kb insert derived from same mRNA. The 3.2 kb clone contained an open reading frame of 2595 by surrounded by 120 by at the 5′ noncoding sequence and 573 by of 3′ noncoding sequence. There were no in-frame stop codons upstream of the predicted initiator methionine, which is preceded by a purine residue (guanine) at −3 position, the most important indicator of a good translation initiation site (Kozak, Mol. Cell. Biol. 9:5134, 1989). It also has a guanine at +4 position, making it an optimal for translation initiation. The open reading frame is predicted to encode a type I transmembrane protein of 864 amino acids. The nucleotide and predicted amino acid sequence is shown in SEQ ID NOs:1 and 2.
  • Computer analysis indicated that the protein has an N-terminal signal peptide with a cleavage site between amino acid 31 and 32. The signal peptide is followed by a 291 amino acid extracellular domain, a 21 amino acid transmembrane domain, and a 521 amino acid cytoplasmic tail. There are eight potential N-linked glycosylation sites in the extracellular domain of the protein. The predicted molecular weight for this protein is 97.8 kilodaltons with an estimated isoelectric point of 4.85. Comparison of both nucleotide and amino acid sequences with the GenBank or EMBL databases found no significant homology with known nucleotide and protein sequences.
  • Example 3
  • This example describes construction of a construct to express a soluble CTLA-8R/Flag® protein referred to as CTLA-8R/Flag. CTLA-8R/Flag® contains a leader sequence, and the region of CTLA-8R from amino acid 1 to amino acid 322 (SEQ ID NO:1), and the octapeptide referred to as Flag® (SEQ ID NO:3). The construct is prepared essentially as described for other soluble constructs, by ligating a DNA fragment encoding amino acids 1 through 322 of SEQ ID NO:1 (prepared as described in Example 4) into an appropriate expression vector which contains a suitable leader sequence. The resultant DNA construct is transfected into a suitable cell line such as the monkey kidney cell line CV-1/EBNA (ATCC CRL 10478). CTLA-8R/Flag® may be purified using a Flag® antibody affinity column, and analyzed for biological activity using any of the methods described herein.
  • Example 4
  • This example describes construction of a CTLA-8R DNA construct to express a CTLA-8R/Fc fusion protein. A soluble form of CTLA-8R fused to the Fc region of human IgG1 was constructed in the mammalian expression vector pDC409 in the following way: A pair of oligonucleotide primers containing a sense sequence and an antisense sequence of CTLA-8R were synthesized. The sense primer contained a Sal I site at the 5′ end of the cDNA and antisense primer contained a Bgl II site and contained the CTLA-8R truncated just before the transmembrane region and a stop codon. A 980 by DNA fragment was amplified from CTLA-8R cDNA. The PCR product was cut with Sal I and Bgl II and used in a three way ligation with a fragment carrying the human IgG1 region cut with Bgl II and Not I into a plasmid (pDC409; see U.S. Ser. No. 08/235,397) previously cut with Sal I and Not I. The encoded insert contained the nucleotides encoding the amino acid sequence of residues 1 to 322 of CTLA-8R (SEQ ID NO:1). The sequence was confirmed by sequencing the whole region.
  • The CTLA-8R/Fc expression plasmids were transfected into CV-1/EBNA cells, and supernatants were collected for 1 week. The CTLA-8/Fc fusion proteins were purified on a protein A sepharose column (Pharmacia, Uppsala, Sweden) as described below. Protein concentration was determined by an enzyme-linked immunoadsorbent assay specific for the constant domain of human IgG1 and by BCA analysis (Pharmacia), and purity was confirmed by SDS-polyacrylamide gel electrophoresis analysis followed by silver stain of the gel.
  • Example 5
  • This example describes purification of CTLA-8R fusion proteins. CTLA-8R/Fc fusion protein is purified by conventional methods using Protein A or Protein G chromatography. Approximately one liter of culture supernatant containing CTLA-8R/Fc fusion protein is purified by filtering mammalian cell supernatants (e.g., in a 0.45 m filter) and applying filtrate to a protein A/G antibody affinity column (Schleicher and Schuell, Keene, N.H.) at 4° C. at a flow rate of 80 ml/hr for a 1.5 cm×12.0 cm column. The column is washed with 0.5 M NaCl in PBS until free protein is not detected in the wash buffer. Finally, the column is washed with PBS. Bound fusion protein is eluted from the column with 25 mM citrate buffer, pH 2.8, and brought to pH 7 with 500 mM Hepes buffer, pH 9.1.
  • A CTLA-8R fusion protein comprising Flag® may also be detected and/or purified using an antibody that binds Flag®, substantially as described in Hopp et al., Bio/Technology 6:1204 (1988). Biological activity is measured by inhibition of CTLA-8 activity in any biological assay which quantifies the co-stimulatory effect of CTLA-8, for example, as described in the Examples herein.
  • Example 6
  • This example illustrates the preparation of monoclonal antibodies against CTLA-8R. Preparations of purified recombinant CTLA-8R, for example, or transfected cells expressing high levels of CTLA-8R, are employed to generate monoclonal antibodies against CTLA-8R using conventional techniques, such as those disclosed in U.S. Pat. No. 4,411,993. Such antibodies are likely to be useful in interfering with CTLA-8R binding to CTLA-8, as components of diagnostic or research assays for CTLA-8R, or in affinity purification of CTLA-8R.
  • To immunize rodents, CTLA-8R immunogen is emulsified in an adjuvant (such as complete or incomplete Freund's adjuvant, alum, or another adjuvant, such as Ribi adjuvant R700 (Ribi, Hamilton, Mont.), and injected in amounts ranging from 10-100 μg subcutaneously into a selected rodent, for example, BALB/c mice or Lewis rats. Ten days to three weeks days later, the immunized animals are boosted with additional immunogen and periodically boosted thereafter on a weekly, biweekly or every third week immunization schedule. Serum samples are periodically taken by retro-orbital bleeding or tail-tip excision for testing by dot-blot assay (antibody sandwich), ELISA (enzyme-linked immunosorbent assay), immunoprecipitation, or other suitable assays, including FACS analysis. Following detection of an appropriate antibody titer, positive animals are given an intravenous injection of antigen in saline. Three to four days later, the animals are sacrificed, splenocytes harvested, and fused to a murine myeloma cell line (e.g., NS1 or preferably Ag 8.653 [ATCC CRL 1580]). Hybridoma cell lines generated by this procedure are plated in multiple microtiter plates in a selective medium (for example, one containing hypoxanthine, aminopterin, and thymidine, or HAT) to inhibit proliferation of non-fused cells, myeloma-myeloma hybrids, and splenocyte-splenocyte hybrids.
  • Hybridoma clones thus generated can be screened by ELISA for reactivity with CTLA-8R, for example, by adaptations of the techniques disclosed by Engvall et al., Immunochem. 8:871 (1971) and in U.S. Pat. No. 4,703,004. A preferred screening technique is the antibody capture technique described by Beckman et al., J. Immunol. 144:4212 (1990). Positive clones are then injected into the peritoneal cavities of syngeneic rodents to produce ascites containing high concentrations (>1 mg/ml) of anti-CTLA-8R monoclonal antibody. The resulting monoclonal antibody can be purified by ammonium sulfate precipitation followed by gel exclusion chromatography. Alternatively, affinity chromatography based upon binding of antibody to protein A or protein G can also be used, as can affinity chromatography based upon binding to CTLA-8R protein.
  • Example 6
  • This example illustrates the ability of CTLA-8R to inhibit the proliferative response of T cells to mitogens. Lymphoid organs were harvested aseptically and cell suspension was created. Splenic and lymph node T cells were isolated from the cell suspension. The purity of the resulting splenic T cell preparations was routinely >95% CD3+ and <1% sIgM+. Purified murine splenic T cells (2×105/well) were cultured with either 1% PHA or 1 μg/ml Con A, and a soluble CTLA-8R was titered into the assay. Proliferation was determined after 3 days with the addition of 1 μCi [3H]thymidine. Soluble CTLA-8R/Fc inhibited the mitogen-induced proliferation of purified murine splenic T cells in a dose dependent manner, while a control Fc had no effect on the murine T cell proliferation.
  • Example 7
  • This example presents the isolation of a DNA encoding human CTLA-8R by cross species hybridization. A human peripheral blood lymphocyte library was prepared and screened substantially as described in U.S. Ser. No. 08/249,189, using murine CTLA-8R DNA under moderately high stringency conditions. Several clones of varying length were obtained. Initial sequencing data indicated that the human CTLA-8R was approximately 76% identical to murine CTLA-8R at the nucleotide level.

Claims (19)

1. An isolated DNA selected from the group consisting of:
(a) a DNA having a nucleotide sequence encoding an amino acid sequence of amino acids 1 through 322 of SEQ ID NO.: 2; and
(b) DNA molecules capable of hybridization to the clones of (a) under stringent conditions and which encode CTLA-8R capable of binding CTLA-8.
2. An isolated DNA according to claim 1 which encodes a soluble CTLA-8R.
3. An isolated DNA selected from the group consisting of:
(a) a DNA having a nucleotide sequence encoding an amino acid sequence of amino acids 1 through 322 of SEQ ID NO.: 2; and
(b) DNA molecules encoding peptides that are at least about 70% identical in amino acid sequence to the amino acid sequence of SEQ ID NO:2.
4. A recombinant expression vector comprising a DNA sequence according to claim 1.
5. A recombinant expression vector comprising a DNA sequence according to claim 2.
6. A recombinant expression vector comprising a DNA sequence according to claim 3.
7. A host cell transformed or transfected with an expression vector according to claim 4.
8. A host cell transformed or transfected with an expression vector according to claim 5.
9. A host cell transformed or transfected with an expression vector according to claim 6.
10. A process for preparing a CTLA-8R or an analog thereof, comprising culturing a host cell according to claim 7 under conditions promoting expression.
11. A process for preparing a CTLA-8R or an analog thereof, comprising culturing a host cell according to claim 8 under conditions promoting expression.
12. A process for preparing a CTLA-8R or an analog thereof, comprising culturing a host cell according to claim 9 under conditions promoting expression.
13. An isolated and purified biologically active CTLA-8 receptor (CTLA-8R) protein.
14. An isolated and purified CTLA-8R protein according to claim 13, consisting essentially of human CTLA-8R.
15. An isolated and purified CTLA-8R according to claim 13, consisting essentially of soluble CTLA-8R.
16. A composition comprising a CTLA-8R protein according to claim 13, and a suitable diluent or carrier.
17. A method for regulating an immune or inflammatory response in a mammal, comprising administering an effective amount of a composition according to claim 16.
18. An assay method for detection of CTLA-8 and homologs thereof, or CTLA-8R and homologs thereof, or the interaction thereof, comprising use of a protein composition according to claim 16.
19. Antibodies immunoreactive with CTLA-8R.
US12/722,477 1995-03-23 2010-03-11 Novel receptor that binds ctla-8 Abandoned US20100233186A1 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
US12/722,477 US20100233186A1 (en) 1995-03-23 2010-03-11 Novel receptor that binds ctla-8

Applications Claiming Priority (8)

Application Number Priority Date Filing Date Title
US41053595A 1995-03-23 1995-03-23
US53876595A 1995-08-07 1995-08-07
US08/620,694 US5869286A (en) 1995-03-23 1996-03-21 Receptor that binds IL-17
US09/022,259 US6191104B1 (en) 1995-03-23 1998-02-11 Methods of treating graft rejection with the IL-17 receptor
US09/549,679 US6680057B1 (en) 1995-03-23 2000-04-14 Methods of treating autoimmune disease by administering interleukin-17 receptor
US10/742,372 US7273843B2 (en) 1995-03-23 2003-12-18 Methods of treating allergy by administering interleukin-17 receptor
US11/901,904 US20090068737A1 (en) 1995-03-23 2007-09-19 Methods of inhibiting interleukin-17 receptor
US12/722,477 US20100233186A1 (en) 1995-03-23 2010-03-11 Novel receptor that binds ctla-8

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
US11/901,904 Continuation US20090068737A1 (en) 1995-03-23 2007-09-19 Methods of inhibiting interleukin-17 receptor

Publications (1)

Publication Number Publication Date
US20100233186A1 true US20100233186A1 (en) 2010-09-16

Family

ID=27021039

Family Applications (9)

Application Number Title Priority Date Filing Date
US08/620,694 Expired - Lifetime US5869286A (en) 1995-03-23 1996-03-21 Receptor that binds IL-17
US09/022,257 Expired - Lifetime US6197525B1 (en) 1995-03-23 1998-02-11 Assay kits for detection and methods of inhibiting IL-17 binding
US09/022,255 Expired - Lifetime US6072033A (en) 1995-03-23 1998-02-11 Receptor that binds IL-17
US09/022,253 Expired - Lifetime US6096305A (en) 1995-03-23 1998-02-11 Receptor that binds IL-17
US09/022,259 Expired - Lifetime US6191104B1 (en) 1995-03-23 1998-02-11 Methods of treating graft rejection with the IL-17 receptor
US09/022,260 Expired - Lifetime US6100235A (en) 1995-03-23 1998-02-11 Methods of treating immune and inflammatory response with IL-17 receptors
US09/022,696 Expired - Lifetime US6072037A (en) 1995-03-23 1998-02-12 Antibodies to the IL-17 receptor
US11/901,904 Abandoned US20090068737A1 (en) 1995-03-23 2007-09-19 Methods of inhibiting interleukin-17 receptor
US12/722,477 Abandoned US20100233186A1 (en) 1995-03-23 2010-03-11 Novel receptor that binds ctla-8

Family Applications Before (8)

Application Number Title Priority Date Filing Date
US08/620,694 Expired - Lifetime US5869286A (en) 1995-03-23 1996-03-21 Receptor that binds IL-17
US09/022,257 Expired - Lifetime US6197525B1 (en) 1995-03-23 1998-02-11 Assay kits for detection and methods of inhibiting IL-17 binding
US09/022,255 Expired - Lifetime US6072033A (en) 1995-03-23 1998-02-11 Receptor that binds IL-17
US09/022,253 Expired - Lifetime US6096305A (en) 1995-03-23 1998-02-11 Receptor that binds IL-17
US09/022,259 Expired - Lifetime US6191104B1 (en) 1995-03-23 1998-02-11 Methods of treating graft rejection with the IL-17 receptor
US09/022,260 Expired - Lifetime US6100235A (en) 1995-03-23 1998-02-11 Methods of treating immune and inflammatory response with IL-17 receptors
US09/022,696 Expired - Lifetime US6072037A (en) 1995-03-23 1998-02-12 Antibodies to the IL-17 receptor
US11/901,904 Abandoned US20090068737A1 (en) 1995-03-23 2007-09-19 Methods of inhibiting interleukin-17 receptor

Country Status (15)

Country Link
US (9) US5869286A (en)
EP (1) EP0817847B2 (en)
JP (1) JP4373495B2 (en)
KR (1) KR100467998B1 (en)
AT (1) ATE279517T1 (en)
AU (1) AU696775B2 (en)
CA (1) CA2215394C (en)
DE (1) DE69633617T3 (en)
DK (1) DK0817847T4 (en)
ES (1) ES2229264T5 (en)
MX (1) MX9707021A (en)
NO (1) NO974258L (en)
NZ (1) NZ306653A (en)
PT (1) PT817847E (en)
WO (1) WO1996029408A1 (en)

Cited By (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US10072085B2 (en) 2010-01-15 2018-09-11 Kirin-Amgen, Inc. Method of treating psoriasis using an IL-17 receptor antibody formulation
US10208122B2 (en) 2006-10-02 2019-02-19 Amgen K-A, Inc. IL-17 receptor A antigen binding proteins

Families Citing this family (87)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6274711B1 (en) * 1993-06-14 2001-08-14 Inserm, Institut National De La Sante Et De La Recherche Medicale Purified mammalian CTLA-8 antigens and related reagents
DE69633617T3 (en) * 1995-03-23 2010-05-20 Immunex Corp., Seattle IL-17 RECEPTOR
US6680057B1 (en) * 1995-03-23 2004-01-20 Immunex Corporation Methods of treating autoimmune disease by administering interleukin-17 receptor
US6902735B1 (en) 1995-07-19 2005-06-07 Genetics Institute, Llc Antibodies to human IL-17F and other CTLA-8-related proteins
ATE295414T1 (en) * 1995-07-19 2005-05-15 Inst Genetics Llc HUMAN CTLA-8 AND USE OF CTLA-8 SIMILAR PROTEINS
US6074849A (en) * 1995-07-19 2000-06-13 Genetics Institute, Inc. Polynucleotides encoding human CTLA-8 related proteins
DE69739375D1 (en) 1996-11-27 2009-06-04 Immunex Corp METHOD OF REGULATING NITROGEN MONOXIDE GENERATION
US7005412B1 (en) 1996-11-27 2006-02-28 Immunex Corporation Method of treating ulcerative colitis and Crohn's disease using IL-17 receptor proteins
US6579520B2 (en) * 1998-05-15 2003-06-17 Genentech, Inc. IL-17 related mammalian cytokine polypeptides (IL-17E)
WO1999014240A1 (en) * 1997-09-17 1999-03-25 Human Genome Sciences, Inc. Interleukin-17 receptor-like protein
US6482923B1 (en) * 1997-09-17 2002-11-19 Human Genome Sciences, Inc. Interleukin 17-like receptor protein
US6849719B2 (en) 1997-09-17 2005-02-01 Human Genome Sciences, Inc. Antibody to an IL-17 receptor like protein
WO1999032659A1 (en) * 1997-12-22 1999-07-01 Stiftelsen Universitetsforskning Bergen (Unifob) METHOD FOR DISEASE PROGNOSIS BASED ON Fc RECEPTOR GENOTYPING
EP1045905A2 (en) * 1998-01-09 2000-10-25 Immunex Corporation Il-17rh dna and polypeptides
US7771719B1 (en) * 2000-01-11 2010-08-10 Genentech, Inc. Pharmaceutical compositions, kits, and therapeutic uses of antagonist antibodies to IL-17E
CA2328496C (en) 1998-05-15 2016-01-05 Genentech, Inc. Il-17 homologous polypeptides and therapeutic uses thereof
US20050147609A1 (en) * 1998-05-15 2005-07-07 Genentech, Inc. Use of anti-IL-17 antibody for the treatment of cartilage damaged by osteoarthritis
US8133734B2 (en) * 1999-03-16 2012-03-13 Human Genome Sciences, Inc. Kit comprising an antibody to interleukin 17 receptor-like protein
WO2000061132A1 (en) * 1999-04-14 2000-10-19 The Government Of The United States Of America, As Represented By The Secretary, Department Of Health And Human Services Compositions containing immunotoxins and agents that inhibit dendritic cell maturation for inducing immune tolerance to a graft
WO2000064327A2 (en) * 1999-04-26 2000-11-02 Duke University Inhibition of complement action
US6936424B1 (en) 1999-11-16 2005-08-30 Matritech, Inc. Materials and methods for detection and treatment of breast cancer
US20040043397A1 (en) 2000-01-11 2004-03-04 Genentech, Inc. IL-17 homologous polypeptides and therapeutic uses thereof
PT1240325E (en) * 1999-12-23 2010-01-07 Genentech Inc Il-17 and il-17r homologous polypeptides and therapeutic uses thereof
EP1897946B1 (en) * 1999-12-23 2012-07-11 Genentech, Inc. IL-17 homologous polypeptides and therapeutic uses thereof
JP2016047051A (en) * 1999-12-23 2016-04-07 ジェネンテック, インコーポレイテッド Il-17 homologous polypeptides and therapeutic uses thereof
MXPA02009055A (en) * 2000-03-16 2003-03-12 Amgen Inc Il 17 receptor like molecules and uses thereof.
US7718397B2 (en) 2000-03-21 2010-05-18 Genentech, Inc. Nucleic acids encoding receptor for IL-17 homologous polypeptides and uses thereof
MXPA02011617A (en) * 2000-05-24 2003-03-10 Schering Corp Mammalian receptor proteins; related reagents and methods.
ES2614260T3 (en) 2000-05-26 2017-05-30 Immunex Corporation Use of antibodies against the interleukin-4 receptor and compositions thereof
US20030096969A1 (en) 2000-06-02 2003-05-22 Genentech, Inc. Secreted and transmembrane polypeptides and nucleic acids encoding the same
AU2001271860A1 (en) * 2000-07-06 2002-01-21 Zymogenetics Inc. Murine cytokine receptor
CA2427824A1 (en) * 2000-07-26 2002-01-31 Zymogenetics, Inc. Human cytokine receptor
EP1326626B1 (en) * 2000-10-18 2020-04-01 Immunex Corporation Methods for treating rheumatoid arthritis using il-17 antagonists
WO2002038764A2 (en) * 2000-11-10 2002-05-16 The Regents Of The University Of California Il-17 receptor-like protein, uses thereof, and modulation of catabolic activity of il-17 cytokines on bone and cartilage
AU2003259995B2 (en) 2002-08-28 2009-07-02 Immunex Corporation Compositions and methods for treating cardiovascular disease
EP2322201A3 (en) * 2002-10-29 2011-07-27 Genentech, Inc. Compositions and methods for the treatment of immune related diseases
JP4441170B2 (en) * 2002-11-28 2010-03-31 独立行政法人理化学研究所 Escherichia coli cell extract having mutation in S12 ribosomal protein and cell-free protein production method using the same
CA2539116C (en) * 2003-09-15 2014-11-18 Research Development Foundation Cripto antagonism of activin and tgf-b signaling
WO2005117979A2 (en) * 2004-05-28 2005-12-15 Applied Research Systems Ars Holding N.V. Use of il-17 in the treatment of fertility-related disorders
AU2005254998B2 (en) 2004-06-10 2011-10-27 Zymogenetics, Inc. Soluble ZcytoR14, anti-ZcytoR14 antibodies and binding partners and methods of using in inflammation
GB0417487D0 (en) 2004-08-05 2004-09-08 Novartis Ag Organic compound
WO2006034278A2 (en) * 2004-09-21 2006-03-30 Matritech, Inc. Methods and compositions for detecting cancer using components of the u2 spliceosomal particle
EA200700823A1 (en) * 2004-10-18 2008-02-28 Займодженетикс, Инк., Сша SOLUBLE ZCYTOR21, ANTI-ZCYTOR21 ANTIBODIES AND BINDING PARTNERS AND METHODS OF APPLICATION FOR INFLAMMATION
JP4590249B2 (en) * 2004-11-17 2010-12-01 独立行政法人理化学研究所 Cell-free protein synthesis system for glycoprotein synthesis
JP4868731B2 (en) 2004-11-17 2012-02-01 独立行政法人理化学研究所 Cell-free protein synthesis system derived from cultured mammalian cells
JP4787488B2 (en) * 2004-11-19 2011-10-05 独立行政法人理化学研究所 Cell-free protein synthesis method using linear template DNA and cell extract therefor
JP2008532936A (en) * 2005-02-14 2008-08-21 ワイス Interleukin-17F antibody and other IL-17F signaling antagonists and uses thereof
JP2008532493A (en) * 2005-02-14 2008-08-21 ワイス Characterization of the interaction between IL-17F and IL-17R
GB0504436D0 (en) * 2005-03-03 2005-04-06 Glaxosmithkline Biolog Sa Vaccine
KR20080031684A (en) 2005-06-14 2008-04-10 암젠 인코포레이티드 Self-buffering protein formulations
AU2006284841B2 (en) 2005-09-01 2012-11-08 Merck Sharp & Dohme Corp. Use of IL-23 and IL-17 antagonists to treat autoimmune ocular inflammatory disease
US20070249533A1 (en) 2005-09-28 2007-10-25 Levin Steven D Il-17a and il-17f antagonists and methods of using the same
AU2007235212A1 (en) * 2006-02-10 2007-10-18 Zymogenetics, Inc. Truncated IL-17RA soluble receptor and methods of using in inflammation
JP2009526084A (en) 2006-02-10 2009-07-16 ザイモジェネティクス, インコーポレイテッド Soluble IL-17RCx4 and uses in inflammation
CA2652924A1 (en) * 2006-06-19 2007-12-27 Wyeth Methods of modulating il-22 and il-17
FR2908999B1 (en) * 2006-11-29 2012-04-27 Biomerieux Sa NOVEL DRUG FOR THE INHIBITION, PREVENTION OR TREATMENT OF RHEUMATOID ARTHRITIS.
FR2910324B1 (en) * 2006-12-21 2009-03-06 Biomerieux Sa NEW MEDICINE FOR THE TREATMENT OF GASTRIC CANCER
CN101668531B (en) 2007-02-28 2014-05-07 默沙东公司 Combination therapy for treatment of immune disorders
WO2008118930A1 (en) 2007-03-26 2008-10-02 Zymogenetics, Inc. Soluble il-17ra/rc fusion proteins and related methods
CL2008000883A1 (en) * 2007-03-28 2008-10-03 Wyeth6 3 METHOD OF DETECTION OF CAPABLE COMPOUNDS TO ANTAGONIZE THE SIGNALING OF IL-17F / IL-17A; COMPOUND IDENTIFIED BY SUCH METHOD; USE OF A QUANTITY OF AN IL-17F / IL-17A SENALIZATION ANTAGONIST, PHARMACEUTICAL COMPOSITION UNDERSTANDING
US8460647B2 (en) 2007-04-20 2013-06-11 Amgen Inc. Pre-ligand assembly domain of the IL-17 receptor
EP2182943B1 (en) * 2007-07-23 2016-10-26 Janssen Biotech, Inc. Methods and compositions for treating fibrosis related disorders using il-17 antagonists
EP2666783B1 (en) * 2007-09-25 2019-06-26 Genzyme Corporation Compositions for inhibiting interleukin-23 pathways
JP5704677B2 (en) * 2007-11-05 2015-04-22 独立行政法人理化学研究所 Method for producing membrane protein
JP2011514335A (en) * 2008-02-21 2011-05-06 アムジェン インコーポレイテッド IL-17RA-IL-17RB antagonist and use of the antagonist
CA2721713C (en) 2008-05-05 2019-07-09 Novimmune Sa Anti-il-17a/il-17f cross-reactive antibodies and methods of use thereof
EP2166358A1 (en) * 2008-09-17 2010-03-24 Fundacio Institut de Recerca de l'Hospital Universitari Vall d'Hebron Differential diagnostic biomarkers of stroke mimicking conditions and methods of use thereof
US20100233270A1 (en) 2009-01-08 2010-09-16 Northwestern University Delivery of Oligonucleotide-Functionalized Nanoparticles
MX2011009797A (en) 2009-03-20 2012-01-12 Amgen Inc Selective and potent peptide inhibitors of kv1.3.
JP6053517B2 (en) 2009-05-05 2016-12-27 ノヴィミュンヌ エスア Anti-IL-17F antibodies and methods for their use
KR20120093932A (en) * 2009-10-10 2012-08-23 일레븐 바이오테라피틱스, 아이엔씨. Il-17 family cytokine compositions and uses
TW201117824A (en) 2009-10-12 2011-06-01 Amgen Inc Use of IL-17 receptor a antigen binding proteins
EP2725034B1 (en) 2010-09-22 2019-04-03 Amgen Inc. Carrier immunoglobulins with no specificity for human tissues and uses thereof
US10092706B2 (en) 2011-04-20 2018-10-09 Amgen Inc. Autoinjector apparatus
WO2013016220A1 (en) 2011-07-22 2013-01-31 Amgen Inc. Il-17 receptor a is required for il-17c biology
WO2013116682A1 (en) 2012-02-02 2013-08-08 Ensemble Therapeutics Corporation Macrocyclic compounds for modulating il-17
EP3593839A1 (en) 2013-03-15 2020-01-15 Amgen Inc. Drug cassette
US10092703B2 (en) 2013-03-15 2018-10-09 Amgen Inc. Drug cassette, autoinjector, and autoinjector system
ES2760002T3 (en) 2014-03-31 2020-05-12 Amgen K A Inc Methods of treating nail and scalp psoriasis
WO2017022696A1 (en) 2015-07-31 2017-02-09 国立研究開発法人理化学研究所 Method of manufacturing membrane protein and utilization thereof
US11866700B2 (en) 2016-05-06 2024-01-09 Exicure Operating Company Liposomal spherical nucleic acid (SNA) constructs presenting antisense oligonucleotides (ASO) for specific knockdown of interleukin 17 receptor mRNA
US11285191B2 (en) 2016-07-26 2022-03-29 The Board Of Supervisors Of Louisiana State University And Agricultural And Mechanical College Immunostimulatory compositions and uses therefor
WO2018201090A1 (en) 2017-04-28 2018-11-01 Exicure, Inc. Synthesis of spherical nucleic acids using lipophilic moieties
CA3084486A1 (en) 2017-11-16 2019-05-23 Amgen Inc. Autoinjector with stall and end point detection
CN114107385A (en) 2019-01-17 2022-03-01 百奥赛图(北京)医药科技股份有限公司 Humanized transgenic animal
CA3150951A1 (en) 2019-09-11 2021-03-18 Bausch Health Ireland Limited Methods of treating nonalcoholic fatty liver disease (nafld) using il-17ra antibody
JP2022176672A (en) * 2021-05-17 2022-11-30 国立大学法人北海道大学 Peptide and pharmaceutical composition

Citations (8)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5116964A (en) * 1989-02-23 1992-05-26 Genentech, Inc. Hybrid immunoglobulins
US5703088A (en) * 1989-08-21 1997-12-30 Beth Israel Deaconess Medical Center, Inc. Topical application of spiperone or derivatives thereof for treatment of pathological conditions associated with immune responses
US5716805A (en) * 1991-10-25 1998-02-10 Immunex Corporation Methods of preparing soluble, oligomeric proteins
US6043344A (en) * 1995-07-19 2000-03-28 Genetics Institute, Inc. Human CTLA-8 and uses of CTLA-8-related proteins
US6083906A (en) * 1990-04-09 2000-07-04 Immunex Corporation Method of regulating nitric oxide production or arthritis with soluble IL-17 receptor
US6100235A (en) * 1995-03-23 2000-08-08 Immunex Corporation Methods of treating immune and inflammatory response with IL-17 receptors
US6274711B1 (en) * 1993-06-14 2001-08-14 Inserm, Institut National De La Sante Et De La Recherche Medicale Purified mammalian CTLA-8 antigens and related reagents
US6793919B2 (en) * 2000-10-18 2004-09-21 Immunex Corporation Methods for treating rheumatoid arthritis using IL-17 antagonists

Family Cites Families (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6562333B1 (en) * 1993-06-14 2003-05-13 Schering Corporation Purified mammalian CTLA-8 antigens and related reagents

Patent Citations (9)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5116964A (en) * 1989-02-23 1992-05-26 Genentech, Inc. Hybrid immunoglobulins
US5703088A (en) * 1989-08-21 1997-12-30 Beth Israel Deaconess Medical Center, Inc. Topical application of spiperone or derivatives thereof for treatment of pathological conditions associated with immune responses
US6083906A (en) * 1990-04-09 2000-07-04 Immunex Corporation Method of regulating nitric oxide production or arthritis with soluble IL-17 receptor
US5716805A (en) * 1991-10-25 1998-02-10 Immunex Corporation Methods of preparing soluble, oligomeric proteins
US6274711B1 (en) * 1993-06-14 2001-08-14 Inserm, Institut National De La Sante Et De La Recherche Medicale Purified mammalian CTLA-8 antigens and related reagents
US6100235A (en) * 1995-03-23 2000-08-08 Immunex Corporation Methods of treating immune and inflammatory response with IL-17 receptors
US6191104B1 (en) * 1995-03-23 2001-02-20 Immunex Corporation Methods of treating graft rejection with the IL-17 receptor
US6043344A (en) * 1995-07-19 2000-03-28 Genetics Institute, Inc. Human CTLA-8 and uses of CTLA-8-related proteins
US6793919B2 (en) * 2000-10-18 2004-09-21 Immunex Corporation Methods for treating rheumatoid arthritis using IL-17 antagonists

Cited By (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US10208122B2 (en) 2006-10-02 2019-02-19 Amgen K-A, Inc. IL-17 receptor A antigen binding proteins
US11180564B2 (en) 2006-10-02 2021-11-23 Amgen K-A, Inc. IL-17 Receptor A antigen binding proteins
US11858999B2 (en) 2006-10-02 2024-01-02 Amgen K-A, Inc. IL-17 receptor A antigen binding proteins
US10072085B2 (en) 2010-01-15 2018-09-11 Kirin-Amgen, Inc. Method of treating psoriasis using an IL-17 receptor antibody formulation
US10808033B2 (en) 2010-01-15 2020-10-20 Amgen K-A, Inc. IL-17 receptor antibody formulation
US11505612B2 (en) 2010-01-15 2022-11-22 Amgen K-A, Inc. Method of treating diseases using an IL-17 receptor antibody formulation

Also Published As

Publication number Publication date
US6072033A (en) 2000-06-06
CA2215394C (en) 2011-04-26
JPH11503309A (en) 1999-03-26
EP0817847A1 (en) 1998-01-14
US6191104B1 (en) 2001-02-20
NO974258D0 (en) 1997-09-15
NZ306653A (en) 1999-03-29
US5869286A (en) 1999-02-09
DE69633617T2 (en) 2005-11-03
AU5526396A (en) 1996-10-08
KR100467998B1 (en) 2005-11-25
MX9707021A (en) 1997-11-29
DK0817847T4 (en) 2009-10-05
US6096305A (en) 2000-08-01
DK0817847T3 (en) 2005-02-14
NO974258L (en) 1997-11-24
DE69633617T3 (en) 2010-05-20
DE69633617D1 (en) 2004-11-18
CA2215394A1 (en) 1996-09-26
EP0817847B2 (en) 2009-09-09
WO1996029408A1 (en) 1996-09-26
US20090068737A1 (en) 2009-03-12
ATE279517T1 (en) 2004-10-15
AU696775B2 (en) 1998-09-17
KR19980703061A (en) 1998-09-05
EP0817847B1 (en) 2004-10-13
US6072037A (en) 2000-06-06
US6197525B1 (en) 2001-03-06
ES2229264T5 (en) 2009-12-17
ES2229264T3 (en) 2005-04-16
JP4373495B2 (en) 2009-11-25
US6100235A (en) 2000-08-08
PT817847E (en) 2005-02-28

Similar Documents

Publication Publication Date Title
US20100233186A1 (en) Novel receptor that binds ctla-8
US5464937A (en) Type II Interleukin-1 receptors
EP0460846B1 (en) Type II interleukin-1 receptors
US7317090B2 (en) Antibodies to Interleukin-4 receptors and uses thereof
US6410711B1 (en) DNA encoding CD40 ligand, a cytokine that binds CD40
US7309489B1 (en) Mouse CD40 ligand-specific antibodies and hybridomas
EP0959897B1 (en) Method of regulating nitric oxide production
AU8656991A (en) Isolated viral protein cytokine antagonists
US7405270B2 (en) CD40-Ligand lacking native-pattern glycosylation
US5925734A (en) Isolated Epstein-Barr virus BZLF2 proteins that bind MHC class II βchains
US7273843B2 (en) Methods of treating allergy by administering interleukin-17 receptor
US5716623A (en) Isolated Herpesvirus saimiri proteins that bind MHC Class II molecules
US7338930B2 (en) Methods of treating osteoarthritis using IL-17 receptor proteins in combination with TNF-receptor proteins

Legal Events

Date Code Title Description
STCB Information on status: application discontinuation

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

AS Assignment

Owner name: KIRIN-AMGEN, INC., CALIFORNIA

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNOR:AMGEN INC.;REEL/FRAME:030045/0876

Effective date: 20101029