EP0620739A1 - Method of treating tnf-dependent inflammation using tumor necrosis factor antagonists - Google Patents

Method of treating tnf-dependent inflammation using tumor necrosis factor antagonists

Info

Publication number
EP0620739A1
EP0620739A1 EP93921562A EP93921562A EP0620739A1 EP 0620739 A1 EP0620739 A1 EP 0620739A1 EP 93921562 A EP93921562 A EP 93921562A EP 93921562 A EP93921562 A EP 93921562A EP 0620739 A1 EP0620739 A1 EP 0620739A1
Authority
EP
European Patent Office
Prior art keywords
tnfr
ser
pro
ala
thr
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Withdrawn
Application number
EP93921562A
Other languages
German (de)
French (fr)
Other versions
EP0620739A4 (en
Inventor
Craig A. Smith
Cindy A. Jacobs
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
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=25484181&utm_source=google_patent&utm_medium=platform_link&utm_campaign=public_patent_search&patent=EP0620739(A1) "Global patent litigation dataset” by Darts-ip is licensed under a Creative Commons Attribution 4.0 International License.
Application filed by Immunex Corp filed Critical Immunex Corp
Publication of EP0620739A1 publication Critical patent/EP0620739A1/en
Publication of EP0620739A4 publication Critical patent/EP0620739A4/en
Withdrawn legal-status Critical Current

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/705Receptors; Cell surface antigens; Cell surface determinants
    • C07K14/715Receptors; Cell surface antigens; Cell surface determinants for cytokines; for lymphokines; for interferons
    • C07K14/7151Receptors; Cell surface antigens; Cell surface determinants for cytokines; for lymphokines; for interferons for tumor necrosis factor [TNF], for lymphotoxin [LT]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P29/00Non-central analgesic, antipyretic or antiinflammatory agents, e.g. antirheumatic agents; Non-steroidal antiinflammatory drugs [NSAID]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides

Abstract

Procédé de traitement de maladies inflammatoires dépendantes du TNF chez un mammifère, consistant à administrer un antagoniste du TNF, tel qu'un récepteur de TNF soluble.A method of treating TNF-dependent inflammatory diseases in a mammal, comprising administering a TNF antagonist, such as a soluble TNF receptor.

Description

TITLE
Method of Treating TNF-Dependent Inflammation Using Tumor Necrosis Factor
Antagonists
CROSS-REFERENCE TO RELATED APPLICATIONS
This application is a continuation-in-part of U.S. application Serial No. 523,635, filed May 10, 1990, now pending, which is a continuation-in-part of U.S. application Serial No. 421,417, filed October 13, 1989, now abandoned, which is a continuation-in-part of U.S. application Serial No. 405,370, filed September 11, 1989, now abandoned, which is a continuation-in-part of U.S. application Serial No. 403,241, filed September 5, 1989, now abandoned.
BACKGROUND OF THE INVENTION The present invention relates generally to cytokine receptors and more specifically to a method of using tumor necrosis factor antagonists to suppress TNF- dependent inflammatory diseases.
Tumor necrosis factor-α (TNFα, also known as cachectin) and tumor necrosis factor-β (TNFβ, also known as lymphotoxin) are homologous mammalian endogenous secretory proteins capable of inducing a wide variety of effects on a large number of cell types. The great similarities in the structural and functional characteristics of these two cytokines have resulted in their collective description as "TNF." Complementary cDNA clones encoding TNFα (Pennica et al., Nature 312:124, 1984) and TNFβ (Gray et al., Nature 312:121, 1984) have been isolated, permitting further structural and biological characterization of TNF.
TNF proteins initiate their biological effect on cells by binding to specific TNF receptor (TNFR^ proteins expressed on the plasma membrane of a TNF-responsive cell. Two distinct forms of TNFR are known to exist: Type I TNFR (TNFRI), having a molecular weight of approximately 75 kilodaltons, and Type II TNFR (TNFRII), having a molecular weight of approximately 55 kilodaltons. TNFRI and TNFRII each bind to both TNFα and TNFβ. TNFRI and TNFRII have both been molecularly cloned (Smith et al., Science 248:1019, 1990; Loetscher et al., Cell 67:351, 1990 and Schall et al., Cell 61:361, 1990), permitting recombinant expression and purification of soluble TNFR proteins. Soluble TNF binding proteins from human urine have also been identified
(Peetre et al., Ewr. J. Haematol. 41:414, 1988; Seckinger et al., J. Exp. Med. 767:1511, 1988; Seckinger et al., J. Biol. Chem. 264:11966, 1989; UK Patent Application, Publ. No. 2 218 101 A to Seckinger et al.; Engelmann et al., J. Biol. Chem. 264:1191 , 1989).
TNF antagonists, such as soluble TNFR and TNF binding proteins, bind to TNF and prevent TNF from binding to cell membrane bound TNF receptors. Such proteins may therefore be useful to suppress biological activities caused by TNF.
The role of TNF in mediated inflammatory diseases and the in vivo biological effects of such soluble TNFR and TNF binding protein proteins in suppressing such TNF-dependent inflammatory diseases have not been fully elucidated and potential therapeutic uses for TNF antagonists have yet to be identified.
SUMMARY OF THE INVENTION The present invention provides a method of using TNF antagonists to suppress TNF-dependent inflammatory diseases. Specifically, the present invention provides a method of treating a human having arthritis comprising the step of administering a TNF antagonist, such as soluble human TNFR, to a human.
These and other aspects of the present invention will become evident upon reference to the following detailed description.
BRIEF DESCRIPTION OF THE DRAWINGS FIGURE 1 shows the dimeric structure of the recombinant human TNFR/Fc fusion protein. The primary translation product of the plasmid coding for rhu TNFR/Fc is a single molecule of soluble TNFR linked to single chain of Fc derived from human IgGl. Following translation, but prior to secretion, this fusion molecule dimerizes via 3 cysteine residues in the Fc region to form dimeric rhu TNFR/Fc. Boxes denote structural domains of TNFR.
FIGURE 2 shows the construction of plasmid pCAVDHFR rhu TNFR/Fc. Abbreviations are as follows: ADH2, yeast alcohol dehydrogenase gene and regulatory region; CMV, cytomegalovirus immediate early enhancer; TPL, adenovirus-2 tripartite leader; VA, adenovirus-2 virus-associated RNA genes I and II; DHFR, hamster dihydrofolate reductase gene.
FIGURES 3 and 4 are graphs showing the effect of intra-articular administration of recombinant human TNFR/Fc, monomeric TNFR, recombinant murine IL-IR and TNFR monomer combined with rmuIL-lR on antigen-induced arthritis in rats. The data indicate that TNFR/Fc, TNFR monomer, rmu IL-IR and TNFR combined with IL-IR suppress inflammation associated with antigen-induced arthritis. FIGURE 5 shows the effect of intraperitoneal administration of recombinant human TNFR/Fc and PBS (vehicle control) on the development of collagen induced arthritis (CIA) in B10.RIII mice. TNFR Fc significantly delayed the onset of CIA.
FIGURE 6 shows the effect of intraperitoneal administration of recombinant human TNFR/Fc and PBS (vehicle control) on the development of collagen induced arthritis (CIA) in DBA/1 mice. TNFR Fc significantly delayed the onset of CIA.
FIGURE 7 shows that administration of TNFR Fc in mice reduced the arthritis index and the number of joints showing signs of arthritis.
DETAILED DESCRIPTION OF THE INVENTION
Definitions
As used herein, the terms "TNF receptor" and "TNFR" refer to proteins having amino acid sequences which are substantially similar to the native mammalian TNF receptor or TNF binding protein amino acid sequences, and which are capable of binding TNF molecules and inhibiting TNF from binding to cell membrane bound TNFR. Two distinct types of TNFR are known to exist: Type I TNFR (TNFRI) and Type II TNFR (TNFRII). The mature full-length human TNFRI is a glycoprotein having a molecular weight of about 75-80 kilodaltons (kDa). The mature full-length human TNFRII is a glycoprotein having a molecular weight of about 55-60 kilodaltons (kDa). The preferred TNFRs of the present invention are soluble forms of TNFRI and TNFRII, as well as soluble TNF binding proteins. Soluble TNFR molecules include, for example, analogs or subunits of native proteins having at least 20 amino acids and which exhibit at least some biological activity in common with TNFRI, TNFRII or TNF binding proteins. Soluble TNFR constructs are devoid of a transmembrane region (and are secreted from the cell) but retain the ability to bind TNF. Various bioequivalent protein and amino acid analogs have an amino acid sequence corresponding to all or part of the extracellular region of a native TNFR, for example, huTNFRIΔ235, huTNFRIΔ185 and huTNFRIΔ163, or amino acid sequences substantially similar to the sequences of amino acids 1-163, amino acids 1-185, or amino acids 1-235 of SEQ ID NO:l, and which are biologically active in that they bind to TNF ligand. Equivalent soluble TNFRs include polypeptides which vary from these sequences by one or more substitutions, deletions, or additions, and which retain the ability to bind TNF or inhibit TNF signal transduction activity via cell surface bound TNF receptor proteins, for example huTNFRIΔx, wherein x is selected from the group consisting of any one of amino acids 163-235 of SEQ ID NO: 1. Analogous deletions may be made to muTNFR. Inhibition of TNF signal transduction activity can be determined by transfecting cells with recombinant TNFR DNAs to obtain recombinant receptor expression. The cells are then contacted with TNF and the resulting metabolic effects examined. If an effect results which is attributable to the action of the ligand, then the recombinant receptor has signal transduction activity. Exemplary procedures for determining whether a polypeptide has signal transduction activity are disclosed by Idzerda et al., J. Exp. Med. 777:861 (1990); Curtis et al., Proc. Natl. Acad. Sci. USA 86:3045 (1989); Prywes et al., EMBO J. 5:2119 (1986) and Chou et al., J. Biol. Chem. 262:1842 (1987). Alternatively, primary cells or cell lines which express an endogenous TNF receptor and have a detectable biological response to TNF could also be utilized.
The nomenclature for TNFR analogs as used herein follows the convention of naming the protein (e.g., TNFR) preceded by either hu (for human) or u (for murine) and followed by a Δ (to designate a deletion) and the number of the C-terminal amino acid. For example, huTNFRΔ235 refers to human TNFR having Asp235 as the C- terminal amino acid (i.e., a polypeptide having the sequence of amino acids 1-235 of SEQ ID NO:l). In the absence of any human or murine species designation, TNFR refers genetically to mammalian TNFR. Similarly, in the absence of any specific designation for deletion mutants, the term TNFR means all forms of TNFR, including mutants and analogs which possess TNFR biological activity.
The term "isolated" or "purified", as used in the context of this specification to define the purity of TNFR protein or protein compositions, means that the protein or protein composition is substantially free of other proteins of natural or endogenous origin and contains less than about 1% by mass of protein contaminants residual of production processes. Such compositions, however, can contain other proteins added as stabilizers, carriers, excipients or co-therapeutics. TNFR is isolated if it is detectable as a single protein band in a polyacrylamide gel by silver staining. "Recombinant," as used herein, means that a protein is derived from recombinant (e.g., microbial or mammalian) expression systems. "Microbial" refers to recombinant proteins made in bacterial or fungal (e.g., yeast) expression systems. As a product, "recombinant microbial" defines a protein produced in a microbial expression system which is essentially free of native endogenous substances. Protein expressed in most bacterial cultures, e.g., E. coli, will be free of glycan. Protein expressed in yeast may have a glycosylation pattern different from that expressed in mammalian cells.
"Biologically active," as used throughout the specification as a characteristic of TNF receptors, means that a particular molecule shares sufficient amino acid sequence similarity with the embodiments of the present invention disclosed herein to be capable of binding detectable quantities of TNF, transmitting a TNF stimulus to a cell, for example, as a component of a hybrid receptor construct, or cross-reacting with anti- TNFR antibodies raised against TNFR from natural (i.e., nonrecombinant) sources. Preferably, biologically active TNF receptors within the scope of the present invention are capable of binding greater than 0.1 nmoles TNF per nmole receptor, and most preferably, greater than 0.5 nmole TNF per nmole receptor in standard binding assays (see below).
Soluble TNF Antagonists and Analogs
The present invention utilizes isolated and purified TNF antagonist polypeptides. The isolated and purified TNF antagonist polypeptides used in this invention are substantially free of other contaminating materials of natural or endogenous origin and contain less than about 1% by mass of protein contaminants residual of production processes. The TNF antagonist polypeptides used in this invention are optibnally without associated native-pattern glycosylation.
In preferred aspects of the present invention, the TNF antagonists are selected from the group consisting of soluble human TNFRI and TNFR II. The pCAV/NOT- TNFR vector, containing the human TNFRI cDNA clone 1, was used to express and purify soluble human TNFRI. pCAV/NOT-TNFR has been deposited with the American Type Culture Collection, 12301 Parklawn Drive, Rockville, MD 20852, USA (Accession No. 68088) under the name pCAV/NOT-TNFR.
Like most mammalian genes, mammalian TNF receptors are presumably encoded by multi-exon genes. Alternative mRNA constructs which can be attributed to different mRNA splicing events following transcription, and which share large regions of identity or similarity with the cDNAs claimed herein may also be used.
Other mammalian TNFR cDNAs may be isolated by using an appropriate human TNFR DNA sequence as a probe for screening a particular mammalian cDNA library by cross-species hybridization. Mammalian TNFR used in the present invention includes, by way of example, primate, human, murine, canine, feline, bovine, ovine, equine and porcine TNFR. Mammalian TNFRs can be obtained by cross species hybridization, using a single stranded cDNA derived from the human TNFR DNA sequence as a hybridization probe to isolate TNFR cDNAs from mammalian cDNA libraries. Derivatives of TNFR which may be used in the present 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 TNFR 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 TNFR amino acid side chains or at the N- or C-termini. Other derivatives of TNFR include covalent or aggregative conjugates of TNFR 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 oc-factor leader). TNFR protein fusions can comprise peptides added to facilitate purification or identification of TNFR (e.g., poly-His). The amino acid sequence of TNF receptor can also be linked to the peptide Asp-Tyr-Lys-Asp-Asp-Asp-Asp-Lys (DYKDDDDK) (Hopp et al., Bio/Technology 6:1204,1988.) The latter sequence is highly antigenic and provides an epitope reversibly bound by a specific monoclonal antibody, enabling rapid assay and facile purification of expressed recombinant protein. This sequence is also specifically cleaved by bovine mucosal enterokinase at the residue immediately following the Asp-Lys pairing. Fusion proteins capped with this peptide may also be resistant to intracellular degradation in E. coli.
TNFR with or without associated native-pattern glycosylation may also be used. TNFR 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 TNFR DNAs in bacteria such as E. coli provides non-glycosylated molecules. Functional mutant analogs of mammalian TNFR 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-Aj-Z, where Ai is any amino acid except Pro, and Z is Ser or Thr. In this sequence, Asn 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 Ai and Z, or an amino acid other than Asn between Asn and A].
TNFR derivatives may also be obtained by mutations of TNFR or its subunits. A TNFR mutant, as referred to herein, is a polypeptide homologous to TNFR but which has an amino acid sequence different from native TNFR because of a deletion, insertion or substitution.
Bioequivalent analogs of TNFR 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 (e.g., Cys178) or replaced with other amino acids to prevent formation of unnecessary or 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 physiochemical 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. Substantially similar polypeptide sequences, as defined above, generally comprise a like number of amino acids sequences, although C-terminal truncations for the purpose of constructing soluble TNFRs will contain fewer amino acid sequences. In order to preserve the biological activity of TNFRs, deletions and substitutions will preferably result in homologous or conservatively substituted sequences, meaning that a given residue is replaced by a biologically similar residue. Examples of conservative substitutions include substitution of one aliphatic residue for another, such as He, Val, Leu, or Ala for one another, or substitutions of one polar residue for another, such as between Lys and Arg; Glu and Asp; or Gin and Asn. Other such conservative substitutions, for example, substitutions of entire regions having similar hydrophobicity characteristics, are well known. Moreover, particular amino acid differences between human, murine and other mammalian TNFRs is suggestive of additional conservative substitutions that may be made without altering the essential biological characteristics of TNFR.
Subunits of TNFR may be constructed by deleting terminal or internal residues or sequences. Particularly preferred sequences include those in which the transmembrane region and intracellular domain of TNFR are deleted or substituted with hydrophilic residues to facilitate secretion of the receptor into the cell culture medium. The resulting protein is referred to as a soluble TNFR molecule which retains its ability to bind TNF. A particularly preferred soluble TNFR construct is TNFRIΔ235 (the sequence of amino acids 1-235 of SEQ ID NO:l), which comprises the entire extracellular region of TNFRI, terminating with Asp235 immediately adjacent the transmembrane region. Additional amino acids may be deleted from the transmembrane region while retaining TNF binding activity. For example, huTNFRIΔ183 which comprises the sequence of amino acids 1-183 of SEQ ID NO:l, and TNFRIΔ163 which comprises the sequence of amino acids 1-163 of SEQ ID NO:l, retain the ability to bind TNF ligand. TNFRIΔ142, however, does not retain the ability to bind TNF ligand. This suggests that one or both of Cys157 and Cys163 is required for formation of an intramolecular disulfide bridge for the proper folding of TNFRI. Cys178, which was deleted without any apparent adverse effect on the ability of the soluble TNFRI to bind TNF, does not appear to be essential for proper folding of TNFRI. Thus, any deletion C-terminal to Cys163 would be expected to result in a biologically active soluble TNFRI. The present invention contemplates use of such soluble TNFR constructs corresponding to all or part of the extracellular region of TNFR terminating with any amino acid after Cys163. Other C-terminal deletions, such as TNFRIΔ157, may be made as a matter of convenience by cutting TNFR cDNA with appropriate restriction enzymes and, if necessary, reconstructing specific sequences with synthetic oligonucleotide linkers. Soluble TNFR with N-terminal deletions may also be used in the present invention. For example, the N- terminus of TNFRI may begin with Leu1, Pro2 or Ala3 without significantly affecting the ability of TNFRI to effectively act as a TNF antagonist. The resulting soluble TNFR constructs are then inserted and expressed in appropriate expression vectors and assayed for the ability to bind TNF.
Mutations in nucleotide sequences constructed for expression of analog TNFR 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 TNFR mutants screened for the desired activity.
Not all mutations in the nucleotide sequence which encodes TNFR 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 37:13, 1985); Craik (BioTechniques, January 1985, 12-19); Smith et al. (Genetic Engineering: Principles and Methods, Plenum Press, 1981); and U.S. Patent Nos. 4,518,584 and 4,737,462 disclose suitable techniques, and are incorporated by reference herein.
Both monovalent forms and polyvalent forms of TNFR may also be used in the present invention. Polyvalent forms possess multiple TNFR binding sites for TNF ligand. For example, a bivalent soluble TNFR may consist of two tandem repeats of amino acids 1-235 of SEQ ID NO:l, separated by a linker region. Alternate polyvalent forms may also be constructed, for example, by chemically coupling TNFR to any clinically acceptable carrier molecule, a polymer selected from the group consisting of Ficoll, polyethylene glycol or dextran using conventional coupling techniques. Alternatively, TNFR may be chemically coupled to biotin, and the biotin-TNFR conjugate then allowed to bind to avidin, resulting in tetravalent avidin/biotin/TNFR molecules. TNFR may also be covalently coupled to dinitrophenol (DNP) or trinitrophenol (TNP) and the resulting conjugate precipitated with anti-DNP or anti- TNP-IgM, to form decameric conjugates with a valency of 10 for TNFR binding sites. A recombinant chimeric antibody molecule may also be produced having TNFR sequences substituted for the variable domains of either or both of the immunoglobulin molecule heavy and light chains and having unmodified constant region domains. For example, chimeric TNFR/IgGi may be produced from two chimeric genes ~ a TNFR/human K light chain chimera (TNFR/CK) and a TNFR/human γi heavy chain chimera (TNFR/Cγ.i). Following transcription and translation of the two chimeric genes, the gene products assemble into a single chimeric antibody molecule having TNFR displayed bivalently. Such polyvalent forms of TNFR may have enhanced binding affinity for TNF ligand. One specific example of a TNFR/Fc fusion protein is disclosed in SEQ ID NO:3 and SEQ ID NO:4. Additional details relating to the construction of such chimeric antibody molecules are disclosed in WO 89/09622 and EP 315062.
Expression of Recombinant TNFR
Recombinant expression vectors are preferably used to amplify or express DNA encoding TNFR to obtain purified TNFR. Recombinant expression vectors are replicable DNA constructs which have synthetic or cDNA-derived DNA fragments encoding mammalian TI "R or bioequivalent analogs operably linked to suitable transcriptional or translational regulatory elements derived from mammalia;; nicrobial, viral or insect genes. A transcriptional unit generally comprises an assemoiy of (1) a genetic element or elements having a regulatory role in gene expression, for example, transcriptional promoters or enhancers, (2) a structural or coding sequence which is transcribed into mRNA and translated into protein, and (3) appropriate transcription and translation initiation and termination sequences, as described in detail below. Such regulatory elements may include an operator sequence to control transcription, a sequence encoding suitable mRNA ribosomal binding sites. 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. Structural elements intended for use in yeast expression systems preferably include a leader sequence enabling extracellular secretion of translated protein by a host cell. Alternatively, where recombinant protein is expressed without a leader or transport sequence, it may include an N-terminal methionine residue. This residue may optionally be subsequently cleaved from the expressed recombinant protein to provide a final product.
DNA sequences encoding mammalian TNF receptors which are to be expressed in a microorganism will preferably contain no introns that could prematurely terminate transcription of DNA into mRNA; however, premature termination of transcription may be desirable, for example, where it would result in mutants having advantageous C-terminal truncations, for example, deletion of a transmembrane region to yield a soluble receptor not bound to the cell membrane. 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 to the sequences of the provided cDNA under moderately stringent conditions (50°C, 2x SSC) and other sequences hybridizing or degenerate to those which encode biologically active TNF receptor polypeptides.
Recombinant TNFR DNA is expressed or amplified in a recombinant expression system comprising a substantially homogeneous monoculture of suitable host microorganisms, for example, bacteria such as E. coli or yeast such as S. cerevisiae, which have stably integrated (by transformation or transfection) a recombinant transcriptional unit into chromosomal DNA or carry the recombinant transcriptional unit as a component of a resident plasmid. Generally, cells constituting the system are the progeny of a single ancestral transformant. Recombinant expression systems as defined herein will express heterologous protein upon induction of the regulatory elements linked to the DNA sequence or synthetic gene to be expressed. Transformed host cells are cells which have been transformed or transfected with TNFR vectors constructed using recombinant DNA techniques. Transformed host cells ordinarily express TNFR, but host cells transformed for purposes of cloning or amplifying TNFR DNA do not need to express TNFR. Expressed TNFR will be deposited in the cell membrane or secreted into the culture supernatant, depending on the TNFR DNA selected. Suitable host cells for expression of mammalian TNFR 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 bacilli. Higher eukaryotic cells include established cell lines of mammalian origin as described below. Cell-free translation systems could also be employed to produce mammalian TNFR using RNAs derived from the DNA constructs of the present invention. 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, New York, 1985), the relevant disclosure of which is hereby incorporated by reference.
Prokaryotic expression hosts may be used for expression of TNFR 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 Staphyolococcus, although others may also be employed as a matter of choice. 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 pGEMl (Promega Biotec, Madison, WI, 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. 8:4051, 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 λ PL promoter and cI857ts thermolabile repressor. Plasmid vectors available from the American Type Culture Collection which incorporate derivatives of the λ P promoter include plasmid pHUB2, resident in E. coli strain JMB9 (ATCC 37092) and pPLc28, resident in E. coli RR1 (ATCC 53082).
Recombinant TNFR proteins 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 TNFR, 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 or URA3 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 or URA3 lesion in the yeast host cell genome then provides an effective environment for detecting transformation by growth in the absence of tryptophan or uracil. Suitable promoter sequences in yeast vectors include the promoters for metallothionein, 3-phosphoglycerate kinase (Hitzeman et al., J. Biol. Chem. 255:2013, 1980) or other glycolytic enzymes (Hess et al., /. Adv. Enzyme Reg. 7:149, 1968; and Holland et al., Biochem. 77:4900, 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., ΕPA 73,657.
Preferred yeast vectors can be assembled using DNA sequences from pUC18 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. 258:2614, 1982) and Beier et al. (Nature 300:124, 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. 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 or URA+ tranformants in medium consisting of 0.67% YNB, with amino acids and bases as described by Sherman et al., Laboratory Course Manual for Methods in Yeast Genetics, Cold Spring Harbor Laboratory, Cold Spring Harbor, New York, 1986.
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% or 4% 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 are also advantageously employed to express recombinant protein. Expression of recombinant proteins in mammalian cells is particularly preferred because such proteins are generally correctly folded, appropriately modified and completely functional. Examples of suitable mammalian host cell lines include the COS-7 lines of monkey kidney cells, described by Gluzman (Cell 23:115, 1981), and other cell lines capable of expressing an appropriate vector including, for example, L cells, C127, 3T3, Chinese hamster ovary (CHO), HeLa and BHK cell lines. Mammalian expression vectors may comprise non transcribed 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 non transcribed 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. Baculovirus systems for production of heterologous proteins in insect cells are reviewed by Luckow and Summers, Bio/Technology 6:41 (1988).
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 re obtained easily from the virus as a fragment which also contains the S V40 viral origin of replication (Fiers et al., Nature 273:113, 1978). Smaller or larger SV40 fragments may also be used, provided the approximately 250 bp sequence extending from the Hind 3 site toward the Bgll site located in the viral origin of replication is included. Further, mammalian genomic TNFR promoter, control and/or signal sequences may be utilized, provided such control sequences are compatible with the host cell chosen. Additional details regarding the use of a mammalian high expression vector to produce a recombinant mammalian TNF receptor are provided in Examples 2 and 7 below. Exemplary vectors can be constructed as disclosed by Okayama and Berg (Mol. Cell. Biol. 5:280, 1983).
A useful system for stable high level expression of mammalian receptor cDNAs in C 127 murine mammary epithelial cells can be constructed substantially as described by Cosman et al. (Mol. Immunol. 23:935, 1986).
Recombinant expression vectors comprising TNFR cDNAs are stably integrated into a host cell's DNA. Elevated levels of expression product is achieved by selecting for cell lines having amplified numbers of vector DNA. Cell lines having amplified numbers of vector DNA are selected, for example, by transforming a host cell with a vector comprising a DNA sequence which encodes an enzyme which is inhibited by a known drug. The vector may also comprise a DNA sequence which encodes a desired protein. Alternatively, the host cell may be co-transformed with a second vector which comprises the DNA sequence which encodes the desired protein. The transformed or co-transformed host cells are then cultured in increasing concentrations of the known drug, thereby selecting for drug-resistant cells. Such drug-resistant cells survive in increased concentrations of the toxic drug by over-production of the enzyme which is inhibited by the drug, frequently as a result of amplification of the gene encoding the enzyme. Where drug resistance is caused by an increase in the copy number of the vector DNA encoding the inhibitable enzyme, there is a concomitant co-amplification of the vector DNA encoding the desired protein (TNFR) in the host cell's DNA.
A preferred system for such co-amplification uses the gene for dihydrofolate reductase (DHFR), which can be inhibited by the drug methotrexate (MTX). To achieve co-amplification, a host cell which lacks an active gene encoding DHFR is either transformed with a vector which comprises DNA sequence encoding DHFR and a desired protein, or is co- transformed with a vector comprising a DNA sequence encoding DHFR and a vector comprising a DNA sequence encoding the desired protein. The transformed or co-transformed host cells are cultured in media containing increasing levels of MTX, and those cells lines which survive are selected.
A particularly preferred co-amplification system uses the gene for glutamine synthetase (GS), which is responsible for the synthesis of glutamate and ammonia using the hydrolysis of ATP to ADP and phosphate to drive the reaction. GS is subject to inhibition by a variety of inhibitors, for example methionine sulphoximine (MSX). Thus, TNFR can be expressed in high concentrations by co-amplifying cells transformed with a vector comprising the DNA sequence for GS and a desired protein, or co-transformed with a vector comprising a DNA sequence encoding GS and a vector comprising a DNA sequence encoding the desired protein, culturing the host cells in media containing increasing levels of MSX and selecting for surviving cells. The GS co-amplification system, appropriate recombinant expression vectors and cells lines, are described in the following PCT applications: WO 87/04462, WO 89/01036, WO 89/10404 and WO 86/05807.
Recombinant proteins are preferably expressed by co-amplification of DHFR or
GS in a mammalian host cell, such as Chinese Hamster Ovary (CHO) cells, or alternatively in a murine myeloma cell line, such as SP2/0-Agl4 or NSO or a rat myeloma cell line, such as YB2/3.0-Ag20, disclosed in PCT applications
WO/89/10404 and WO 86/05807.
A preferred eukaryotic vector for expression of TNFR DNA is disclosed below in Example 1. This vector, referred to as pCAV/NOT, was derived from the mammalian high expression vector pDC201 and contains regulatory sequences from SV40, adenovirus-2, and human cytomegalovirus.
Purification of Recombinant TNFR
Purified mammalian TNF receptors 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 TNF 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. 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 TNFR 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 mammalian TNFR 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 mammalian TNFR 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. 296:111, 1984). This reference describes two sequential, reversed- phase HPLC steps for purification of recombinant human GM-CSF on a preparative HPLC column.
Human TNFR synthesized in recombinant culture is characterized by the presence of non-human cell components, including proteins, in amounts and of a character which depend upon the purification steps taken to recover human TNFR 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 TNFR free of proteins which may be normally associated with TNFR as it is found in nature in its species of origin, e.g. in cells, cell exudates or body fluids.
Therapeutic Administration of Recombinant Soluble TNFR
The present invention provides methods of suppressing TNF-dependent inflammatory responses in humans comprising administering an effective amount of a TNF antagonist, such as TNFR, and a suitable diluent and carrier. For therapeutic use, purified soluble TNFR protein is administered to a patient, preferably a human, for treatment of arthritis. Thus, for example, soluble TNFR protein compositions can be administered, for example, via intra-articular, intraperitoneal or subcutaneous routes by bolus injection, continuous infusion, sustained release from implants, or other suitable techniques. Typically, a soluble TNFR therapeutic agent will be administered in the form of a composition comprising purified protein 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 compositions entails combining the TNFR 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 con specific 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. In accordance with appropriate industry standards, preservatives may also be added, such as benzyl alcohol. 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.
TNF antagonist proteins are administered to a mammal, preferably a human, for the purpose treating TNF-dependent inflammatory diseases, such as arthritis. For example, TNFRI proteins inhibit TNF-dependent arthritic responses. Because of the primary roles IL-1 and IL-2 play in the production of TNF, combination therapy using TNFR in combination with IL-IR and/or IL-2R may be preferred in the treatment of TNF-associated clinical indications. In the treatment of humans, soluble human TNFR is preferred. Either Type I IL-IR or Type II IL-IR, or a combination thereof, may be used in accordance with the present invention to treat TNF-dependent inflammatory diseases, such as arthritis. Other types of TNF binding proteins may be similarly used.
For treatment of arthritis, TNFR is administered in systemic amounts ranging from about 0.1 mg kg/week to about 100 mg/kg/week. In preferred embodiments of the present invention, TNFR is administered in amounts ranging from about 0.5 mg kg/week to about 50 mg/kg/week. For local intra-articular administration, dosages preferably range from about 0.01 mg/kg to about 1.0 mg/kg per injection.
The following examples are offered by way of illustration, and not by way of limitation.
EXAMPLES
Example 1
Expression and Purification of Soluble Human TNFRI
The cloning of the cDNA for the 80 kD form of the human TNF receptor has been described in detail (Smith et al., Science 248:1019, 1990). The expression vector pCAV/NOT-TNFR (ATCC 68088) containing the TNFR cDNA clone 1 was used to prepare and express a soluble human TNFRI as follows. A cDNA encoding a soluble human TNFRIΔ235 (the primary translation product of which had the sequence of amino acids -22-235 of SEQ ID NO: l) was constructed by excising an 840 bp fragment from pCAV/NOT-TNFR with the restriction enzymes Notl and Pvu2. Notl cuts at the multiple cloning site of pCAV/NOT-TNFR and Pvu2 cuts within the TNFR coding region 20 nucleotides 5' of the transmembrane region. In order to reconstruct the 3' end of the TNFR sequences, two oligonucleotides were synthesized and annealed to create the following oligonucleotide linker encoding amino acids corresponding to amino acids 229-235 of SEQ ID NO: 1:
Pvu2 BamHl Bgl2
CTGAAGGGAGCACTGGCGACX GGATCCA GACTTCCCTCGTGACCGCTGATTCCTAGGTCTAG AlaGluGlySerThrGlyAspEn≤L
This oligonucleotide linker has terminal Pvu2 and Bgl2 restriction sites, regenerates 20 nucleotides of the TNFR, followed by a termination codon (underlined) and a BamHl restriction site (for convenience in isolating the entire soluble TNFR by Notl/BamHl digestion). This oligonucleotide was then ligated with the 840 bp Notl/Pvu2 TNFR insert into Bgl2/Notl cut pCAV/NOT to yield psolhuTNFRΔ235/CAVNOT, which was transfected into COS-7 cells as described above. The host cells expressed a mature a soluble human TNFRI protein having the sequence of amino acids 1-235 which was capable of binding TNF.
Example 2
Construction and Expression of Soluble Human TNFR/Fc Fusion Protein
A schematic diagram showing the construction of a recombinant soluble human TNFR:Fc expression vector is shown in Figure 1. The rhu TNFR:Fc fusion gene was created by ligating the following fragments into Bluescript®, a commercially available cloning vector (Stratagene):
1) An 867 bp Asp718-Pvu2 fragment from pCAV/NOT-TNFR (ATCC 68088) containing the cDNA encoding the truncated TNFR.
2) A 700 bp Styl-Spel fragment from plasmid pIXY498 coding for 232 amino acids of the Fc portion of human IgGl. Plasmid pIXY498 is a yeast expression vector containing the Fc fragment of human IgGl (see Figure 2). 3) An oligonucleotide linker, to fuse the truncated TNFR with the human IgGl Fc fragment. This linker was created by PCR (polymerase chain reaction) amplification using two primers, one having the sequence CCCCAGCTGAAGGGAGCACTGGCG ACGAGCCCAAATCTTGTGACAAAACTC (nucleotides 833-883 of SEQ ID
NO: 3) which encodes the 3' end of the truncated TNF receptor and the 5' end of human IgGl, and the other having the sequence
CGGTACGTGCTGTTGTTACTGC (SEQ ID NO:5), an antisense sequence encoding nucleotides 257-237 of human IgGl. The template for this reaction was pIXY498. The reaction product was digested with Pvu2 and Styl, and a
115 bp fragment was isolated.
This construct was then digested with Notl and the resulting 1.4 kilobase fragment containing the rhu TNFR:Fc fusion DNA sequence was ligated into the Notl site of plasmid CA V NOT/DHFR. Plasmid pCA V/NOT/DHFR was derived from plasmid pCAV/NOT by inserting the hamster dihydrofolate reductase DNA sequence (DHFR) into the Hpal site of pCAV/NOT (Figure 2). This construct was designated plasmid pCAVDHFRhuTNFRFc. The entire coding region sequence was confirmed by DNA sequencing and is depicted in Figure 2. To prepare the host strain, DXB-11 CHO cells deficient in the expression of dihydrofolate reductase (DHFR) were obtained from Dr. Lawren Chasin at Columbia University. A bank of 100 vials of these cells was established, and representative vials were sent to Microbiological Associates for examination via the following procedures:
Test Result
1. Transmission Electron Microscopy (TEM) Type A only,
2. Sterility - Bacterial and Fungal negative
3. Mycoplasma negative
4. Mouse Antibody Production (MAP) negative
All transfections and amplification steps were performed in a separate laboratory set aside for this purpose. Only mycoplasma-free cell lines were allowed into this facility.
Transfections were performed by mixing pCAVDHFRhuTNFRFc plasmid DNA with LipofectinTM reagent from Gibco BRL. Approximately 10 >g of DNA was added to 10 cm petri dishes containing CHO DXB-11 cells. After the initial transfection, cells were selected for the expression of DHFR by subculturing in selective medium lacking glycine, hypoxanthine and thymidine. The resulting colonies were then transferred to 24 well plates and analyzed for rhu TNFR:Fc expression. The highest expressing cultures were subjected to amplification by exposure to increasing concentrations of methotrexate (MTX). Cells able to grow at 25 nM MTX were cloned by limiting dilution in 96 well plates. The highest expressing clones were transferred to suspension culture and the final selection of clone 4-4FC102A5-3 was made based on its high level of rhu TNFR:Fc expression under these conditions.
Example 3
Expression of Monomeric Soluble TNF Receptors in CHO Cells
Soluble TNF receptor was expressed in Chinese Hamster Ovary (CHO) cells using the glutamine-synthetase (GS) gene amplification system, substantially as described in PCT patent application Nos. WO87/04462 and WO89/01036. Briefly, CHO cells are transfected with an expression vector containing genes for both TNFR and GS. CHO cells are selected for GS gene expression based on the ability of the transfected DNA to confer resistance to low levels of methionine sulphoximine (MSX). GS sequence amplification events in such cells are selected using elevated MSX concentrations. In this way, contiguous TNFR sequences are also amplified and enhanced TNFR expression is achieved. The vector used in the GS expression system was psolTNFR/P6/PSVLGS, which was constructed as follows. First, the vector pSVLGS.l (described in PCT Application Nos. WO87/04462 and WO89/01036, and available from Celltech, Ltd., Berkshire, UK) was cut with the BamHl restriction enzyme and dephosphorylated with calf intestinal alkaline phosphatase (CIAP) to prevent the vector from religating to itself. The BamHl cut pSVLGS.l fragment was then ligated to a 2.4 kb BamHl to Bgl2 fragment of pEEόhCMV (described in PCT Application No. WO89/01036, also available from Celltech) which was cut with Bgl2, BamHl and Fspl to avoid two fragments of similar size, to yield an 11.2 kb vector designated p6/PSVLGS.l. pSVLGS.l contains the glutamine synthetase selectable marker gene under control of the SV40 later promoter. The BamHl to Bgl2 fragment of pEEόhCMV contains the human cytomegalovirus major immediate early promoter (hCMV), a polylinker, and the SV40 early polyadenylation signal. The coding sequences for soluble TNFR were added to p6 PSVLGS.l by excising a Notl to BamHl fragment from the expression vector psolTNFR/CAVNOT (made according to Example 3 above), blunt ending with Klenow and ligating with Smal cut dephosphorylated p6/PSVLGS.l, thereby placing the solTNFR coding sequences under the control of the hCMV promoter. This resulted in a single plasmid vector in which the SV40/GS and hCMB/solTNFR transcription units are transcribed in opposite directions. This vector was designated psolTNFR/P6/PSVLGS. psolTNFR/P6/PSVLGS was used to transfect CHO-K1 cells (available from ATCC, Rochville, MD, under accession number CCL 61) as follows. A monolayer of CHO-K1 cells were grown to subconfluency in Minimum Essential Medium (MEM) 10X (Gibco: 330-1581AJ) without glutamine and supplemented with 10% dialysed fetal bovine serum (Gibco: 220-6300AJ), 1 mM sodium pyruvate (Sigma), MEM non- essential amino acids (Gibco: 320-1140AG), 500 μM asparagine and glutamate (Sigma) and nucleosides (30 μM adenosine, guanosine, cytidine and uridine and 10 μM thymidine)(Sigma).
Approximately 1 x 106 cells per 10 cm petri dish were transfected with 10 ug of psolTNFR/P6/PSVLGS by standard calcium phosphate precipitation, substantially as described by Graham & van der Eb, Virology 52:456 (1983). Cells were subjected to glycerol shock (15% glycerol in serum-free culture medium for approximately 1.5 minutes) approximately 4 hours after transfection, substantially as described by Frost & Williams, Virology 91:39 (1978), and then washed with serum-free medium. One day later, transfected cells were fed with fresh selective medium containing MSX at a final concentration of 25 uM. Colonies of MSX-resistant surviving cells were visible within 3-4 weeks. Surviving colonies were transferred to 24-well plates and allowed to grow to confluency in selective medium. Conditioned medium from confluent wells were then assayed for soluble TNFR activity using standard binding assays. These assays indicated that the colonies expressed biologically active soluble TNFR.
In order to select for GS gene amplification, several MSX-resistant cell lines are transfected with psolTNFR/P6/PSVLGS and grown in various concentrations of MSX. For each cell line, approximately lxlO6 cells are plated in gradually increasing concentrations of 100 uM, 250 uM, 500 uM and 1 mM MSX and incubated for 10-14 days. After 12 days, colonies resistant to the higher levels of MSX appear. The surviving colonies are assayed for TNFR activity. Each of these highly resistant cell lines contains cells which arise from multiple independent amplification events. From these cells lines, one or more of the most highly resistant cells lines are isolated. The amplified cells with high production rates are then cloned by limiting dilution cloning. Mass cell cultures of the transfectants secrete active soluble TNFR.
Example 4 Effect of Soluble TNFR on Antigen-Induced Arthritis in Rats
Lewis rats previously immunized with methylated bovine serum albumin (mBSA) in complete Freund's adjuvant develop antigen-induced arthritis (ALA) when challenged with mBSA in knee joints. Administration of rhu TNFR:Fc, TNFR monomer, recombinant murine soluble IL-1 receptor (rm IL-IR) or a combination of TNFR monomer plus rm IL-IR was shown to be effective in suppressing the effects of antigen-induced arthritis in rats. Lewis rats were immunized in the hind flank with 0.5 mg mBSA in complete
Freund's adjuvant. Twenty-one days later (day 0), the animals were injected in both hind knee joints with 50 μg mBSA in pyrogen-free saline. Groups of six rats were injected intra-aiticularly in both knee joints on that day and on the following 2 days (days 0, 1 and 2) as indicated below in Table A:
Table A Treatment and Dosage Schedule
Group Treatment Dose
Knee joint width was measured daily on days 0-6 relative to treatment. TNFR monomer was produced in CHO cells according to Example 2. The rhu TNFR:Fc used in this experiment was produced in BHK (hamster kidney) cells. This material is similar to the CHO cell-derived TNFR.
Figures 3 and 4 demonstrate that treatment with BHK-derived rhu TNFR:Fc at the time of mBSA challenge and for two days following challenge resulted in a reduction of knee-joint swelling in comparison to diluent-treated control rats. A reduction in joint swelling and inflammation was observed in rats treated with 5 or 10 μg BHK-derived rhu TNFR:Fc or 5 μg TNFR monomer or 1 μg of rmuIL-lR. Reduction in joint swelling was even more pronounced when rmuIL-lR and TNFR monomer treatment was combined.
Histopathological examination of the joints harvested on day 6 was performed to confirm the degree of swelling. Histopathology scores were derived by evaluating knee joints and scoring their condition as follows: Grade 1, minimal, <10% of area affected; Grade 2, moderate, 10-50% of area affected; Grade 3, marked, at least 50%, but less than all, of area affected; Grade 4, maximal, total area severely affected. A variety of lesions/alterations involving five knee joint structures were evaluated: joint capsule, joint space, synovial membrane, articular cartilage, and subchondral bone. Each structural alteration was scored from 1 to 4, and the scores were added and means were calculated. Histopathology results are expressed as the mean score in each treatment group.
The following Table B shows histopathology results, which also indicate that rhu TNFR:Fc, TNFR monomer and rmu IL-IR were effective in reducing the severity of antigen-induced arthritis, and that a combination of rm IL-IR and TNFR monomer was more effective than either receptor alone.
Table B Effect of rhu TNFRrFc on Antigen Induced Arthritis in Rats
Treatment Histopathology Score Number (Mean ± SD (SE)) Of
Animals
Saline 18.4 ± 4.9 (1.5) 10
LO μg rmu IL-lR 13.1 1 4.7 (1.7) 8
10.0 μg TNFR monomer 12.8 ± 3.1 (1.1) 8
1.0 μg rmu IL-1R/10.0 μg TNFR monomer 7.9 ± 5.2 (2.0) 5
5.0 μg TNFR monomer 13.4 ± 2.8 (1.0) 9
5.0 μg rhu TNFR:Fc (BHK) 13.4 ± 3.6 (1.3) 8
In summary, treatment with rhu TNFR Fc, TNFR monomer, or rmu IL-IR at the time of mBSA challenge and for two days following challenge resulted in a reduction of knee-joint swelling in comparison to diluent-treated control rats. A combination of both rmu IL-IR and TNFR monomer resulted in greater reduction of swelling than either receptor molecule alone. Histopathology results also indicated that rhu TNFR Fc, TNFR and rmu IL-IR were effective in reducing the severity of antigen- induced arthritis, and that a combination of rmu IL-IR and TNFR monomer was more effective than either receptor alone. Example 5 Effect of Soluble TNFR on Collagen-Induced Arthritis in BlO.Riπ Mice
BIO.RIII mice previously immunized with porcine type II collagen (CII) in complete Freund's adjuvant consistently develop collagen-induced arthritis (CIA).
Administration of rhu TNFR:Fc was shown to be effective in suppressing the symptoms of CIA in mice.
BIO.RIII mice were immunized intradermally with 100 μg porcine type II collagen (CII) in complete Freund's adjuvant to induced arthritic symptoms. Approximately 14-17 days post-immunization, symptoms of clinical arthritis began to appear in the mice, with 90-100% of the mice displaying severe arthritis by day 28.
Mice were injected intraperitoneally with TNFR Fc or PBS to determine the effect of soluble TNFR/Fc on CIA. Mice were assessed for symptoms of arthritis at 12 weeks post-immunization . In a first experiment, TNFR Fc was administered over the entire period of CIA development. Twelve mice were injected with 10 μg TNFR/Fc, 3 days per week, from days 0 to 35. Twelve control mice were injected with PBS. Figure 5 shows that
TNFR/Fc significantly reduced the incidence of arthritis when compared to controls.
Upon cessation of treatment with TNFR/Fc, the mice developed arthritis. In a second experiment, TNFR/Fc was administered during only the developmental stages of CIA on days -1-17 relative to immunization, as set forth in the following Table C.
Table C Effect of rhu TNFR:Fc Administered During Inductive Stage of CIA
Treatment Incidence Onset Severity (Positive/Total) (Mean Day ± SE) (Mean ± SE)
30 μg TNFR/Fc 10/10 24 ± 2 10.5 ± 0.5 Days -1, 3
10 μg TNFR/Fc 8/10 21± 2 8.6 ± 0.6 Days -1 to 17 (alternate days)
100 μl PBS 10/10 18 + 1 10.6 ± 0.4
Days -1 to 17 (alternate days) These data show that TNFR/Fc delayed the onset of arthritis, but that CIA was unaltered in mice receiving 30 μg TNFR/Fc the day before and 3 days after immunization with type II collagen. Mice given 10 μg TNFR/Fc, every other day, from day -1 to day 17 displayed a slight decrease in CIA incidence and severity versus controls injected with PBS.
In a third experiment, TNFR/Fc was administered during only the progressive stages of CIA every other day on days 14-28 post-immunization as set forth in the following Table D.
Table D
Effect of rhu TNFR:Fc Administered During Progressive Stage of CIA
Treatment Incidence Onset Severity
(Positive Total) (Mean Day ± SE) (Mean ± SE)
10 μg TNFR/Fc 8/9 27 ± 6 8.6 ± 1.3
Days 14-28
(alternate days)
100 3 μμll PPBBSS 9/9 21 ± 1 8.7 ± 0.6 DDaayyss 1144--2288 (alternate days)
These data show that mice given 10 μg TNFR Fc, every other day, from days 14-28 showed a slight delay in CIA onset when compared to control animals. However, the incidence and severity of arthritis appears to be unaltered.
In summary, these experiments indicate that TNFR/Fc was effective in delaying the onset of CIA when administered over the entire course of CIA development.
Example 6
Effect of Soluble TNFR on Collagen-Induced Arthritis in DBA/1 Mice
The effect of soluble TNFR Fc on CIA in DBA/1 mice previously immunized with porcine type II collagen (CII) in complete Freund's adjuvant was also tested.
Administration of rhu TNFR:Fc was shown to be effective in suppressing the symptoms of CIA.
In this experiment, DBA/1 mice were immunized with 100 μg of CII and then injected intraperitoneally with 50 μg recombinant soluble human TNFR/Fc in sterile saline from day 21 to day 28. Control mice received sterile saline (vehicle) injections. This treatment period was prior to the development of the clinical signs of CIA, but during the development of DTH responses to type II collagen and rapid IgG anti-CII production.
Both groups of mice were assessed for the development of CIA for 70 days, and onset of CIA for 44-55 days post-immunization. Figures 6 and 7 show that
TNFR/Fc significantly reduced the incidence of CIA compared with controls (28% vs. 86%; p<0.03), and reduced both arthritis index (a subjective measure of severity) and the number of involved joints. The antibody response to CII was significantly lower immediately post treatment with TNFR/Fc (day 28), but antibody levels were equivalent at the conclusion of the experiment (day 70).
These results indicate that TNFR Fc is effective in reducing the incidence of CIA in mice and may therefore be useful in the treatment arthritis.
SEQUENCE LISTING
(1) GENERAL INFORMATION:
(i) APPLICANT: Jacobs, Cindy A.
(ii) TITLE OF INVENTION: Method of Treating TNF-Dependent
Inflammation Using Tumor Necrosis Factor Antagonists
(iii) NUMBER OF SEQUENCES: 5
(iv) CORRESPONDENCE ADDRESS:
(A) ADDRESSEE: Immunex Corporation (B) STREET: 51 University Street
(C) CITY: Seattle
(D) STATE: Washington
(E) COUNTRY: U.S.A.
(F) ZIP: 98101
(v) COMPUTER READABLE FORM:
(A) MEDIUM TYPE: Floppy disk
(B) COMPUTER: IBM PC compatible
(C) OPERATING SYSTEM: PC-DOS/MS-DOS (D) SOFTWARE: PatentIn Release #1.0, Version #1.25
(vi) CURRENT APPLICATION DATA:
(A) APPLICATION NUMBER:
(B) FILING DATE: (C) CLASSIFICATION:
(viii) ATTORNEY/AGENT INFORMATION:
(A) NAME: Wight, Christopher L.
(B) REGISTRATION NUMBER: 31,680 (C) REFERENCE/DOCKET NUMBER: 2503
(ix) TELECOMMUNICATION INFORMATION:
(A) TELEPHONE: (206) 587-0430
(B) TELEFAX: (206) 587-0606
(2) INFORMATION FOR SEQ ID NO:l:
(i) SEQUENCE CHARACTERISTICS: (A) LENGTH: 1641 base pairs
(B) TYPE: nucleic acid
(C) STRANDEDNESS: single
(D) TOPOLOGY: linear (ii) MOLECULE TYPE: cDNA (iii) HYPOTHETICAL: NO (iv) ANTI-SENSE: NO
(vi) ORIGINAL SOURCE:
(A) ORGANISM: Homo sapiens (G) CELL TYPE: Fibroblast (H) CELL LINE: WI-26 VA4 28
(vii) IMMEDIATE SOURCE:
(A) LIBRARY: WI-26 VA4
(B) CLONE: Clone 1 (ix) FEATURE:
(A) NAME/KEY: CDS
(B) LOCATION: 88..1473
(ix) FEATURE: (A) NAME/KEY: mat_peptide
(B) LOCATION: 154..1470
(ix) FEATURE:
(A) NAME/KEY: sig_peptide (B) LOCATION: 88..153
(xi) SEQUENCE DESCRIPTION: SEQ ID NO: 1 : GCGAGGCAGG CAGCCTGGAG AGAAGGCGCT GGGCTGCGAG GGCGCGAGGG CGCGAGGGCA
GGGGGCAACC GGACCCCGCC CGCATCC ATG GCG CCC GTC GCC GTC TGG GCC
Met Ala Pro Val Ala Val Trp Ala -22 -20 -15
GCG CTG GCC GTC GGA CTG GAG CTC TGG GCT GCG GCG CAC GCC TTG CCC Ala Leu Ala Val Gly Leu Glu Leu Trp Ala Ala Ala His Ala Leu Pro -10 -5 1 GCC CAG GTG GCA TTT ACA CCC TAC GCC CCG GAG CCC GGG AGC ACA TGC Ala Gin Val Ala Phe Thr Pro Tyr Ala Pro Glu Pro Gly Ser Thr Cys 5 10 15
CGG CTC AGA GAA TAC TAT GAC CAG ACA GCT CAG ATG TGC TGC AGC AAA Arg Leu Arg Glu Tyr Tyr Asp Gin Thr Ala Gin Met Cys Cys Ser Lys 20 25 30
TGC TCG CCG GGC CAA CAT GCA AAA GTC TTC TGT ACC AAG ACC TCG GAC Cys Ser Pro Gly Gin His Ala Lys Val Phe Cys Thr Lys Thr Ser Asp 35 40 45 50
ACC GTG TGT GAC TCC TGT GAG GAC AGC ACA TAC ACC CAG CTC TGG AAC Thr Val Cys Asp Ser Cys Glu Asp Ser Thr Tyr Thr Gin Leu Trp Asn 55 60 65
TGG GTT CCC GAG TGC TTG AGC TGT GGC TCC CGC TGT AGC TCT GAC CAG Trp Val Pro Glu Cys Leu Ser Cys Gly Ser Arg Cys Ser Ser Asp Gin 70 75 80 GTG GAA ACT CAA GCC TGC ACT CGG GAA CAG AAC CGC ATC TGC ACC TGC Val Glu Thr Gin Ala Cys Thr Arg Glu Gin Asn Arg lie Cys Thr Cys 85 90 95
AGG CCC GGC TGG TAC TGC GCG CTG AGC AAG CAG GAG GGG TGC CGG CTG Arg Pro Gly Trp Tyr Cys Ala Leu Ser Lys Gin Glu Gly Cys Arg Leu 100 105 110
TGC GCG CCG CTG CGC AAG TGC CGC CCG GGC TTC GGC GTG GCC AGA CCA Cys Ala Pro Leu Arg Lys Cys Arg Pro Gly Phe Gly Val Ala Arg Pro 115 120 125 130 GGA ACT GAA ACA TCA GAC GTG GTG TGC AAG CCC TGT GCC CCG GGG ACG 591 Gly Thr Glu Thr Ser Asp Val Val Cys Lys Pro Cys Ala Pro Gly Thr 135 140 145 TTC TCC AAC ACG ACT TCA TCC ACG GAT ATT TGC AGG CCC CAC CAG ATC 639 Phe Ser Asn Thr Thr Ser Ser Thr Asp lie Cys Arg Pro His Gin lie 150 155 160
TGT AAC GTG GTG GCC ATC CCT GGG AAT GCA AGC ATG GAT GCA GTC TGC 687 Cys Asn Val Val Ala lie Pro Gly Asn Ala Ser Met Asp Ala Val Cys 165 170 175
ACG TCC ACG TCC CCC ACC CGG AGT ATG GCC CCA GGG GCA GTA CAC TTA 735 Thr Ser Thr Ser Pro Thr Arg Ser Met Ala Pro Gly Ala Val His Leu 180 185 190
CCC CAG CCA GTG TCC ACA CGA TCC CAA CAC ACG CAG CCA ACT CCA GAA 783 Pro Gin Pro Val Ser Thr Arg Ser Gin His Thr Gin Pro Thr Pro Glu 195 200 205 210
CCC AGC ACT GCT CCA AGC ACC TCC TTC CTG CTC CCA ATG GGC CCC AGC 831 Pro Ser Thr Ala Pro Ser Thr Ser Phe Leu Leu Pro Met Gly Pro Ser 215 220 225 CCC CCA GCT GAA GGG AGC ACT GGC GAC TTC GCT CTT CCA GTT GGA CTG 879 Pro Pro Ala Glu Gly Ser Thr Gly Asp Phe Ala Leu Pro Val Gly Leu 230 235 240
ATT GTG GGT GTG ACA GCC TTG GGT CTA CTA ATA ATA GGA GTG GTG AAC 927 lie Val Gly Val Thr Ala Leu Gly Leu Leu lie lie Gly Val Val Asn 245 250 255
TGT GTC ATC ATG ACC CAG GTG AAA AAG AAG CCC TTG TGC CTG CAG AGA 975 Cys Val lie Met Thr Gin Val Lys Lys Lys Pro Leu Cys Leu Gin Arg 260 265 270
GAA GCC AAG GTG CCT CAC TTG CCT GCC GAT AAG GCC CGG GGT ACA CAG 1023 Glu Ala Lys Val Pro His Leu Pro Ala Asp Lys Ala Arg Gly Thr Gin 275 280 285 290
GGC CCC GAG CAG CAG CAC CTG CTG ATC ACA GCG CCG AGC TCC AGC AGC 1071 Gly Pro Glu Gin Gin His Leu Leu lie Thr Ala Pro Ser Ser Ser Ser 295 300 305 AGC TCC CTG GAG AGC TCG GCC AGT GCG TTG GAC AGA AGG GCG CCC ACT 1119 Ser Ser Leu Glu Ser Ser Ala Ser Ala Leu Asp Arg Arg Ala Pro Thr 310 315 320
CGG AAC CAG CCA CAG GCA CCA GGC GTG GAG GCC AGT GGG GCC GGG GAG 1167 Arg Asn Gin Pro Gin Ala Pro Gly Val Glu Ala Ser Gly Ala Gly Glu 325 330 335
GCC CGG GCC AGC ACC GGG AGC TCA GAT TCT TCC CCT GGT GGC CAT GGG 1215 Ala Arg Ala Ser Thr Gly Ser Ser Asp Ser Ser Pro Gly Gly His Gly 340 345 350
ACC CAG GTC AAT GTC ACC TGC ATC GTG AAC GTC TGT AGC AGC TCT GAC 1263 Thr Gin Val Asn Val Thr Cys lie Val Asn Val Cys Ser Ser Ser Asp 355 360 365 370 CAC AGC TCA CAG TGC TCC TCC CAA GCC AGC TCC ACA ATG GGA GAC ACA 131 His Ser Ser Gin Cys Ser Ser Gin Ala Ser Ser Thr Met Gly Asp Thr 375 380 385 GAT TCC AGC CCC TCG GAG TCC CCG AAG GAC GAG CAG GTC CCC TTC TCC 135 Asp Ser Ser Pro Ser Glu Ser Pro Lys Asp Glu Gin Val Pro Phe Ser 390 395 400
AAG GAG GAA TGT GCC TTT CGG TCA CAG CTG GAG ACG CCA GAG ACC CTG 1407 Lys Glu Glu Cys Ala Phe Arg Ser Gin Leu Glu Thr Pro Glu Thr Leu 405 410 415
CTG GGG AGC ACC GAA GAG AAG CCC CTG CCC CTT GGA GTG CCT GAT GCT 145 Leu Gly Ser Thr Glu Glu Lys Pro Leu Pro Leu Gly Val Pro Asp Ala 420 425 430
GGG ATG AAG CCC AGT TAACCAGGCC GGTGTGGGCT GTGTCGTAGC CAAGGTGGGC 151 Gly Met Lys Pro Ser 435 440
TGAGCCCTGG CAGGATGACC CTGCGAAGGG GCCCTGGTCC TTCCAGGCCC CCACCACTAG 157
GACTCTGAGG CTCTTTCTGG GCCAAGTTCC TCTAGTGCCC TCCACAGCCG CAGCCTCCCT 163 CTGACCTGCA G 164
(2) INFORMATION FOR SEQ ID NO:2 : (i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 461 amino acids
(B) TYPE: amino acid (D) TOPOLOGY: linear (ii) MOLECULE TYPE: protein
(xi) SEQUENCE DESCRIPTION: SEQ ID NO:2 :
Met Ala Pro Val Ala Val Trp Ala Ala Leu Ala Val Gly Leu Glu Leu -22 -20 -15 -10
Trp Ala Ala Ala His Ala Leu Pro Ala Gin Val Ala Phe Thr Pro Tyr -5 1 5 10 Ala Pro Glu Pro Gly Ser Thr Cys Arg Leu Arg Glu Tyr Tyr Asp Gin
15 20 25
Thr Ala Gin Met Cys Cys Ser Lys Cys Ser Pro Gly Gin His Ala Lys 30 35 40
Val Phe Cys Thr Lys Thr Ser Asp Thr Val Cys Asp Ser Cys Glu Asp 45 50 55
Ser Thr Tyr Thr Gin Leu Trp Asn Trp Val Pro Glu Cys Leu Ser Cys 60 65 70
Gly Ser Arg Cys Ser Ser Asp Gin Val Glu Thr Gin Ala Cys Thr Arg
75 80 85 90 Glu Gin Asn Arg lie Cys Thr Cys Arg Pro Gly Trp Tyr Cys Ala Leu
95 100 105 Ser Lys Gin Glu Gly Cys Arg Leu Cys Ala Pro Leu Arg Lys Cys Arg 110 115 120
Pro Gly Phe Gly Val Ala Arg Pro Gly Thr Glu Thr Ser Asp Val Val 125 130 135
Cys Lys Pro Cys Ala Pro Gly Thr Phe Ser Asn Thr Thr Ser Ser Thr 140 145 150
Asp lie Cys Arg Pro His Gin lie Cys Asn Val Val Ala lie Pro Gly 155 160 165 170
Asn Ala Ser Met Asp Ala Val Cys Thr Ser Thr Ser Pro Thr Arg Ser 175 180 185
Met Ala Pro Gly Ala Val His Leu Pro Gin Pro Val Ser Thr Arg Ser
190 195 200 Gin His Thr Gin Pro Thr Pro Glu Pro Ser Thr Ala Pro Ser Thr Ser 205 210 215
Phe Leu Leu Pro Met Gly Pro Ser Pro Pro Ala Glu Gly Ser Thr Gly 220 225 230
Asp Phe Ala Leu Pro Val Gly Leu lie Val Gly Val Thr Ala Leu Gly 235 240 245 250
Leu Leu lie lie Gly Val Val Asn Cys Val He Met Thr Gin Val Lys 255 260 265
Lys Lys Pro Leu Cys Leu Gin Arg Glu Ala Lys Val Pro His Leu Pro
270 275 280 Ala Asp Lys Ala Arg Gly Thr Gin Gly Pro Glu Gin Gin His Leu Leu 285 290 295
He Thr Ala Pro Ser Ser Ser Ser Ser Ser Leu Glu Ser Ser Ala Ser 300 305 310
Ala Leu Asp Arg Arg Ala Pro Thr Arg Asn Gin Pro Gin Ala Pro Gly
315 320 325 330
Val Glu Ala Ser Gly Ala Gly Glu Ala Arg Ala Ser Thr Gly Ser Ser 335 340 345
Asp Ser Ser Pro Gly Gly His Gly Thr Gin Val Asn Val Thr Cys He 350 355 360 Val Asn Val Cys Ser Ser Ser Asp His Ser Ser Gin Cys Ser Ser Gin 365 370 375
Ala Ser Ser Thr Met Gly Asp Thr Asp Ser Ser Pro Ser Glu Ser Pro 380 385 390
Lys Asp Glu Gin Val Pro Phe Ser Lys Glu Glu Cys Ala Phe Arg Ser
395 400 405 410
Gin Leu Glu Thr Pro Glu Thr Leu Leu Gly Ser Thr Glu Glu Lys Pro 415 420 425 Leu Pro Leu Gly Val Pro Asp Ala Gly Met Lys Pro Ser 430 435
(2) INFORMATION FOR SEQ ID NO:3:
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 1557 base pairs
(B) TYPE: nucleic acid (C) STRANDEDNESS : single
(D) TOPOLOGY: linear
(ii) MOLECULE TYPE: cDNA (iii) HYPOTHETICAL: NO
(iv) ANTI-SENSE: NO
(vii) IMMEDIATE SOURCE: (B) CLONE: TNFR/Fc Fusion Protein
(ix) FEATURE:
(A) NAME/KEY: CDS
(B) LOCATION: 1..1557
(ix) FEATURE:
(A) NAME/KEY: mat_peρtide
(B) LOCATION: 1..1554
(xi) SEQUENCE DESCRIPTION: SEQ ID NO:3:
GCG AGG CAG GCA GCC TGG AGA GAA GGC GCT GGG CTG CGA GGG CGC GAG 48 Ala Arg Gin Ala Ala Trp Arg Glu Gly Ala Gly Leu Arg Gly Arg Glu 1 5 10 15
GGC GCG AGG GCA GGG GGC AAC CGG ACC CCG CCC GCA TCC ATG GCG CCC 96 Gly Ala Arg Ala Gly Gly Asn Arg Thr Pro Pro Ala Ser Met Ala Pro 20 25 30
GTC GCC GTC TGG GCC GCG CTG GCC GTC GGA CTG GAG CTC TGG GCT GCG 144 Val Ala Val Trp Ala Ala Leu Ala Val Gly Leu Glu Leu Trp Ala Ala 35 40 45 GCG CAC GCC TTG CCC GCC CAG GTG GCA TTT ACA CCC TAC GCC CCG GAG 192 Ala His Ala Leu Pro Ala Gin Val Ala Phe Thr Pro Tyr Ala Pro Glu 50 55 60
CCC GGG AGC ACA TGC CGG CTC AGA GAA TAC TAT GAC CAG ACA GCT CAG 240 Pro Gly Ser Thr Cys Arg Leu Arg Glu Tyr Tyr Asp Gin Thr Ala Gin 65 70 75 80
ATG TGC TGC AGC AAA TGC TCG CCG GGC CAA CAT GCA AAA GTC TTC TGT 288 Met Cys Cys Ser Lys Cys Ser Pro Gly Gin His Ala Lys Val Phe Cys 85 90 95
ACC AAG ACC TCG GAC ACC GTG TGT GAC TCC TGT GAG GAC AGC ACA TAC 336 Thr Lys Thr Ser Asp Thr Val Cys Asp Ser Cys Glu Asp Ser -Thr Tyr 100 105 110 ACC CAG CTC TGG AAC TGG GTT CCC GAG TGC TTG AGC TGT GGC TCC CGC 384 Thr Gin Leu Trp Asn Trp Val Pro Glu Cys Leu Ser Cys Gly Ser Arg 115 120 125 TGT AGC TCT GAC CAG GTG GAA ACT CAA GCC TGC ACT CGG GAA CAG AAC 432 Cys Ser Ser Asp Gin Val Glu Thr Gin Ala Cys Thr Arg Glu Gin Asn 130 135 140
CGC ATC TGC ACC TGC AGG CCC GGC TGG TAC TGC GCG CTG AGC AAG CAG 480 Arg He Cys Thr Cys Arg Pro Gly Trp Tyr Cys Ala Leu Ser Lys Gin 145 150 155 160
GAG GGG TGC CGG CTG TGC GCG CCG CTG CGC AAG TGC CGC CCG GGC TTC 528 Glu Gly Cys Arg Leu Cys Ala Pro Leu Arg Lys Cys Arg Pro Gly Phe 165 170 175
GGC GTG GCC AGA CCA GGA ACT GAA ACA TCA GAC GTG GTG TGC AAG CCC 576 Gly Val Ala Arg Pro Gly Thr Glu Thr Ser Asp Val Val Cys Lys Pro 180 185 190 TGT GCC CCG GGG ACG TTC TCC AAC ACG ACT TCA TCC ACG GAT ATT TGC 624 Cys Ala Pro Gly Thr Phe Ser Asn Thr Thr Ser Ser Thr Asp He Cys 195 200 205
AGG CCC CAC CAG ATC TGT AAC GTG GTG GCC ATC CCT GGG AAT GCA AGC 672 Arg Pro His Gin He Cys Asn Val Val Ala He Pro Gly Asn Ala Ser 210 215 220
ATG GAT GCA GTC TGC ACG TCC ACG TCC CCC ACC CGG AGT ATG GCC CCA 720 Met Asp Ala Val Cys Thr Ser Thr Ser Pro Thr Arg Ser Met Ala Pro 225 230 235 240
GGG GCA GTA CAC TTA CCC CAG CCA GTG TCC ACA CGA TCC CAA CAC ACG 768 Gly Ala Val His Leu Pro Gin Pro Val Ser Thr Arg Ser Gin His Thr 245 250 255
CAG CCA ACT CCA GAA CCC AGC ACT GCT CCA AGC ACC TCC TTC CTG CTC 816 Gin Pro Thr Pro Glu Pro Ser Thr Ala Pro Ser Thr Ser Phe Leu Leu 260 265 270 CCA ATG GGC CCC AGC CCC CCA GCT GAA GGG AGC ACT GGC GAC GAG CCC 864 Pro Met Gly Pro Ser Pro Pro Ala Glu Gly Ser Thr Gly Asp Glu Pro 275 280 285
AAA TCT TGT GAC AAA ACT CAC ACA TGC CCA CCG TGC CCA GCA CCT GAA 912 Lys Ser Cys Asp Lys Thr His Thr Cys Pro Pro Cys Pro Ala Pro Glu 290 295 300
CTC CTG GGG GGA CCG TCA GTC TTC CTC TTC CCC CCA AAA CCC AAG GAC 960 Leu Leu Gly Gly Pro Ser Val Phe Leu Phe Pro Pro Lys Pro Lys Asp 305 310 315 320
ACC CTC ATG ATC TCC CGG ACC CCT GAG GTC ACA TGC GTG GTG GTG GAC 1008- Thr Leu Met He Ser Arg Thr Pro Glu Val Thr Cys Val Val Val Asp 325 330 335
GTG AGC CAC GAA GAC, CCT GAG GTC AAG TTC AAC TGG TAC GTG GAC GGC 1056 Val Ser His Glu Asp Pro Glu Val Lys Phe Asn Trp Tyr Val Asp Gly 340 345 350 - GTG GAG GTG CAT AAT GCC AAG ACA AAG CCG CGG GAG GAG CAG TAC AAC 1104 Val Glu Val His Asn Ala Lys Thr Lys Pro Arg Glu Glu Gin Tyr Asn 355 360 365 AGC ACG TAC CGG GTG GTC AGC GTC CTC ACC GTC CTG CAC CAG GAC TGG 1152 Ser Thr Tyr Arg Val Val Ser Val Leu Thr Val Leu His Gin Asp Trp 370 375 380
CTG AAT GGC AAG GAC TAC AAG TGC AAG GTC TCC AAC AAA GCC CTC CCA 1200 Leu Asn Gly Lys Asp Tyr Lys Cys Lys Val Ser Asn Lys Ala Leu Pro 385 390 395 400
GCC CCC ATG CAG AAA ACC ATC TCC AAA GCC AAA GGG CAG CCC CGA GAA 1248 Ala Pro Met Gin Lys Thr He Ser Lys Ala Lys Gly Gin Pro Arg Glu 405 410 415
CCA CAG GTG TAC ACC CTG CCC CCA TCC CGG GAT GAG CTG ACC AAG AAC 1296 Pro Gin Val Tyr Thr Leu Pro Pro Ser Arg Asp Glu Leu Thr Lys Asn 420 425 430
CAG GTC AGC CTG ACC TGC CTG GTC AAA GGC TTC TAT CCC AGG CAC ATC 1344 Gin Val Ser Leu Thr Cys Leu Val Lys Gly Phe Tyr Pro Arg His He 435 440 445 GCC GTG GAG TGG GAG AGC AAT GGG CAG CCG GAG AAC AAC TAC AAG ACC 1392 Ala Val Glu Trp Glu Ser Asn Gly Gin Pro Glu Asn Asn Tyr Lys Thr 450 455 460
ACG CCT CCC GTG CTG GAC TCC GAC GGC TCC TTC TTC CTC TAC AGC AAG 1440 Thr Pro Pro Val Leu Asp Ser Asp Gly Ser Phe Phe Leu Tyr Ser Lys 465 470 475 480
CTC ACC GTG GAC AAG AGC AGG TGG CAG CAG GGG AAC GTC TTC TCA TGC 1488 Leu Thr Val Asp Lys Ser Arg Trp Gin Gin Gly Asn Val Phe Ser Cys 485 490 495
TCC GTG ATG CAT GAG GCT CTG CAC AAC CAC TAC ACG CAG AAG AGC CTC 153 Ser Val Met His Glu Ala Leu His Asn His Tyr Thr Gin Lys Ser Leu 500 505 510
TCC CTG TCT CCG GGT AAA TGA 1557
Ser Leu Ser Pro Gly Lys 515
(2) INFORMATION FOR SEQ ID NO:4:
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 518 amino acids (B) TYPE: amino acid
(D) TOPOLOGY: linear
(ii) MOLECULE TYPE: protein (xi) SEQUENCE DESCRIPTION: SEQ ID NO:4:
Ala Arg Gin Ala Ala Trp Arg Glu Gly Ala Gly Leu Arg Gly Arg Glu 1 5 10 15 Gly Ala Arg Ala Gly Gly Asn Arg Thr Pro Pro Ala Ser Met Ala Pro
20 25 30 Val Ala Val Trp Ala Ala Leu Ala Val Gly Leu Glu Leu Trp Ala Ala 35 40 45 Ala His Ala Leu Pro Ala Gin Val Ala Phe Thr Pro Tyr Ala Pro Glu 50 55 60
Pro Gly Ser Thr Cys Arg Leu Arg Glu Tyr Tyr Asp Gin Thr Ala Gin 65 70 75 80
Met Cys Cys Ser Lys Cys Ser Pro Gly Gin His Ala Lys Val Phe Cys 85 90 95
Thr Lys Thr Ser Asp Thr Val Cys Asp Ser Cys Glu Asp Ser Thr Tyr 100 105 110
Thr Gin Leu Trp Asn Trp Val Pro Glu Cys Leu Ser Cys Gly Ser Arg
115 120 125 Cys Ser Ser Asp Gin Val Glu Thr Gin Ala Cys Thr Arg Glu Gin Asn 130 135 140
Arg He Cys Thr Cys Arg Pro Gly Trp Tyr Cys Ala Leu Ser Lys Gin 145 150 155 160
Glu Gly Cys Arg Leu Cys Ala Pro Leu Arg Lys Cys Arg Pro Gly Phe 165 170 175
Gly Val Ala Arg Pro Gly Thr Glu Thr Ser Asp Val Val Cys Lys Pro 180 185 190.
Cys Ala Pro Gly Thr Phe Ser Asn Thr Thr Ser Ser Thr Asp He Cys
195 200 205 Arg Pro His Gin He Cys Asn Val Val Ala He Pro Gly Asn Ala Ser 210 215 220
Met Asp Ala Val Cys Thr Ser Thr Ser Pro Thr Arg Ser Met Ala Pro 225 230 235 240
Gly Ala Val His Leu Pro Gin Pro Val Ser Thr Arg Ser Gin His Thr 245 250 255
Gin Pro Thr Pro Glu Pro Ser Thr Ala Pro Ser Thr Ser Phe Leu Leu 260 265 270
Pro Met Gly Pro Ser Pro Pro Ala Glu Gly Ser Thr Gly Asp Glu Pro
275 280 285 Lys Ser Cys Asp Lys Thr His Thr Cys Pro Pro Cys Pro Ala Pro Glu 290 295 300
Leu Leu Gly Gly Pro Ser Val Phe Leu Phe Pro Pro Lys Pro Lys Asp
305 310 315 320
Thr Leu Met He Ser Arg Thr Pro Glu Val Thr Cys Val Val Val Asp
325 330 335
Val Ser His Glu Asp Pro Glu Val Lys Phe Asn Trp Tyr Val Asp Gly 340 345 350 Val Glu Val His Asn Ala Lys Thr Lys Pro Arg Glu Glu Gin Tyr Asn 355 360 365
Ser Thr Tyr Arg Val Val Ser Val Leu Thr Val Leu His Gin Asp Trp 370 375 380
Leu Asn Gly Lys Asp Tyr Lys Cys Lys Val Ser Asn Lys Ala Leu Pro 385 390 395 400 Ala Pro Met Gin Lys Thr He Ser Lys Ala Lys Gly Gin Pro Arg Glu
405 410 415
Pro Gin Val Tyr Thr Leu Pro Pro Ser Arg Asp Glu Leu Thr Lys Asn 420 425 430
Gin Val Ser Leu Thr Cys Leu Val Lys Gly Phe Tyr Pro Arg His He 435 440 445
Ala Val Glu Trp Glu Ser Asn Gly Gin Pro Glu Asn Asn Tyr Lys Thr 450 455 460
Thr Pro Pro Val Leu Asp Ser Asp Gly Ser Phe Phe Leu Tyr Ser Lys 465 470 475 480 Leu Thr Val Asp Lys Ser Arg Trp Gin Gin Gly Asn Val Phe Ser Cys
485 490 495
Ser Val Met His Glu Ala Leu His Asn His Tyr Thr Gin Lys Ser Leu 500 505 510
Ser Leu Ser Pro Gly Lys 515
(2) INFORMATION FOR SEQ ID NO:5:
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 22 base pairs
(B) TYPE: nucleic acid (C) STRANDEDNESS: single
(D) TOPOLOGY: linear
(ii) MOLECULE TYPE: DNA (genomic) (iii) HYPOTHETICAL: NO (iv) ANTI-SENSE: YES
(vii) IMMEDIATE SOURCE:
(B) CLONE: oligonucleotide
(xi) SEQUENCE DESCRIPTION: SEQ ID NO:5 : CGGTACGTGC TGTTGTTACT GC 22

Claims

CLAIMSWe claim:
1. A method for treating TNF-mediated inflammatory diseases which comprises administering to a mammal in need thereof a therapeutically effective amount of a TNF antagonist.
2. A method according to claim 1, wherein the TNF-mediated inflammatory disease is arthritis.
3. A method according to claim 2, wherein the mammal is a human.
4. A method according to claim 3, wherein the TNF antagonist is soluble human TNFR.
5. A method according to claim 4, wherein the soluble human TNFR is selected from the group consisting of soluble human Type I TNFR and soluble human Type π TNFR.
6. A method according to claim 4, wherein the soluble human TNFR is fused to the Fc region of a human immunoglobulin molecule.
7. A method according to claim 2, wherein TNFR is administered in combination with IL-IR.
8. A method for treating arthritis in a mammal, comprising the step of administering to a mammal having arthritis an amount of soluble human TNFR ranging from about 0.1 mg/kg/week to about 100 mg/kg/week.
9. A method according to claim 8, wherein the amount of soluble human TNFR ranges from about 0.5 mg/kg/week to about 50 mg/kg/week.
EP93921562A 1992-09-15 1993-09-14 Method of treating tnf-dependent inflammation using tumor necrosis factor antagonists. Withdrawn EP0620739A4 (en)

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
US94623692A 1992-09-15 1992-09-15
US946236 1992-09-15
PCT/US1993/008666 WO1994006476A1 (en) 1992-09-15 1993-09-14 Method of treating tnf-dependent inflammation using tumor necrosis factor antagonists

Publications (2)

Publication Number Publication Date
EP0620739A1 true EP0620739A1 (en) 1994-10-26
EP0620739A4 EP0620739A4 (en) 1997-01-15

Family

ID=25484181

Family Applications (1)

Application Number Title Priority Date Filing Date
EP93921562A Withdrawn EP0620739A4 (en) 1992-09-15 1993-09-14 Method of treating tnf-dependent inflammation using tumor necrosis factor antagonists.

Country Status (8)

Country Link
EP (1) EP0620739A4 (en)
JP (1) JPH07504203A (en)
KR (1) KR100232688B1 (en)
AU (1) AU670125B2 (en)
CA (1) CA2123593C (en)
NO (1) NO941780L (en)
NZ (1) NZ256293A (en)
WO (1) WO1994006476A1 (en)

Families Citing this family (118)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
ATE289350T1 (en) 1989-04-21 2005-03-15 Amgen Inc TNF RECEPTOR, TNF BINDING PROTEINS AND DNA CODING THEREOF
US7264944B1 (en) 1989-04-21 2007-09-04 Amgen Inc. TNF receptors, TNF binding proteins and DNAs coding for them
US6221675B1 (en) 1989-04-21 2001-04-24 Amgen, Inc. TNF receptors, TNF binding proteins and DNAs coding for them
IL95031A (en) 1989-07-18 2007-03-08 Amgen Inc Method for the production of a human recombinant tumor necrosis factor inhibitor
US6143866A (en) * 1989-07-18 2000-11-07 Amgen, Inc. Tumor necrosis factor (TNF) inhibitor and method for obtaining the same
AU671116B2 (en) * 1992-03-30 1996-08-15 Immunex Corporation Fusion proteins comprising tumor necrosis factor receptor
US6270766B1 (en) 1992-10-08 2001-08-07 The Kennedy Institute Of Rheumatology Anti-TNF antibodies and methotrexate in the treatment of arthritis and crohn's disease
WO1996003141A1 (en) * 1994-07-22 1996-02-08 F. Hoffmann-La Roche Ag Pharmaceutical compositions comprising a chimaeric tnf binding protein
US7091181B2 (en) 1994-12-12 2006-08-15 Omeros Corporation Method of inhibition of pain and inflammation during surgery comprising administration of soluble TNF receptors
AU4658596A (en) * 1995-02-03 1996-08-21 G.D. Searle & Co. Novel c-mpl ligands
IL112834A (en) * 1995-03-01 2000-12-06 Yeda Res & Dev Pharmaceutical compositions for controlled release of soluble receptors
US5705364A (en) * 1995-06-06 1998-01-06 Genentech, Inc. Mammalian cell culture process
US6656466B1 (en) 1995-06-06 2003-12-02 Genetech, Inc. Human tumor necrosis factor—immunoglobulin(TNFR1-IgG1) chimera composition
US5721121A (en) * 1995-06-06 1998-02-24 Genentech, Inc. Mammalian cell culture process for producing a tumor necrosis factor receptor immunoglobulin chimeric protein
ES2171547T3 (en) 1995-07-14 2002-09-16 Applied Research Systems RECEPTOR OF TNF AND DHEA STEROID HORMONE IN COMBINED THERAPY.
US7012060B1 (en) 1995-07-14 2006-03-14 Applied Research Systems Ars Holding N.V. TNF receptor and steroid hormone in a combined therapy
US6936439B2 (en) 1995-11-22 2005-08-30 Amgen Inc. OB fusion protein compositions and methods
US20030040467A1 (en) 1998-06-15 2003-02-27 Mary Ann Pelleymounter Ig/ob fusions and uses thereof.
US6090382A (en) 1996-02-09 2000-07-18 Basf Aktiengesellschaft Human antibodies that bind human TNFα
AU725408B2 (en) * 1996-05-08 2000-10-12 F. Hoffmann-La Roche Ag Treatment of asthma with TNFR-Ig
TW555765B (en) 1996-07-09 2003-10-01 Amgen Inc Low molecular weight soluble tumor necrosis factor type-I and type-II proteins
AU5696198A (en) * 1996-12-06 1998-06-29 Amgen, Inc. Combination therapy using a tnf binding protein for treating tnf-mediated diseases
EP1352656A3 (en) * 1996-12-06 2004-02-04 Amgen Inc. Combination therapy using a TNF binding protein for treating TNF-mediated diseases
US6294170B1 (en) 1997-08-08 2001-09-25 Amgen Inc. Composition and method for treating inflammatory diseases
EP0980258A1 (en) * 1997-05-12 2000-02-23 The Kennedy Institute Of Rheumatology Suppression of tumor necrosis factor alpha and vascular endothelial growth factor in therapy
AU8492598A (en) * 1997-07-17 1999-02-10 Regents Of The University Of Michigan, The Methods and compositions for tumor reduction
AU772778B2 (en) * 1998-07-13 2004-05-06 University Of Southern California Novel inhibitors of angiogenesis and tumor growth
US7067144B2 (en) 1998-10-20 2006-06-27 Omeros Corporation Compositions and methods for systemic inhibition of cartilage degradation
US6660843B1 (en) * 1998-10-23 2003-12-09 Amgen Inc. Modified peptides as therapeutic agents
DE69937070D1 (en) * 1998-11-05 2007-10-18 Omeros Corp RINSE SOLUTION AND METHOD FOR PAIN LIFTING AND INFLAMMATION INHIBITION
TR200504220T2 (en) 1998-12-17 2007-04-24 Biogen Idec Ma Inc. Active lymphotoxin-beta receptor immunoglobulin chime A method for high level expression and purification of purified protein proteins and a method for purification of active lymphotoxin-beta receptor immunoglobulin chimeric proteins.
IL127851A0 (en) * 1998-12-30 1999-10-28 Applied Research Systems Inhibition of TNF activity
EP1022027A1 (en) * 1999-01-22 2000-07-26 Applied Research Systems ARS Holding N.V. Tumor necrosis factor antagonists and their use in endometriosis
EP1165116A1 (en) * 1999-04-02 2002-01-02 Immunex Corporation Use of soluble tumor necrosis factor receptor for treating heart failure
EP1939300A1 (en) * 1999-05-28 2008-07-02 Targeted Genetics Corporation Methods and compositions for lowering the level of tumor necrosis factor (TNF) in TNF-associated disorders
DK1180159T3 (en) * 1999-05-28 2008-11-17 Targeted Genetics Corp Methods and Compositions to Lower the Level of Tumor Necrosis Factor (TNF) in TNF-Associated Disorders
US6627199B1 (en) 1999-07-09 2003-09-30 Amgen Inc Isolation, identification and characterization of tmst2, a novel member of the TNF-receptor supergene family
ES2308985T3 (en) * 1999-07-21 2008-12-16 Omeros Corporation DISSOLUTIONS AND PROCEDURES FOR THE INHIBITION OF PAIN, INFLAMMATION AND DEGRADATION OF CARTILAGO.
WO2001010908A1 (en) 1999-08-04 2001-02-15 Amgen Inc. Ntr3, a member of the tnf-receptor supergene family
EP1210435A1 (en) 1999-08-04 2002-06-05 Amgen Inc. Fhm, A NOVEL MEMBER OF THE TNF LIGAND SUPERGENE FAMILY
GB9927681D0 (en) * 1999-11-23 2000-01-19 Glaxo Group Ltd Protein
KR20020097241A (en) 2000-05-04 2002-12-31 에이브이아이 바이오파마 인코포레이티드 Splice-region antisense composition and method
CA2868614A1 (en) 2001-06-08 2002-12-08 Abbott Laboratories (Bermuda) Ltd. Methods of administering anti-tnf.alpha. antibodies
KR100453877B1 (en) 2001-07-26 2004-10-20 메덱스젠 주식회사 METHOD OF MANUFACTURING Ig-FUSION PROTEINS BY CONCATAMERIZATION, TNFR/Fc FUSION PROTEINS MANUFACTURED BY THE METHOD, DNA CODING THE PROTEINS, VECTORS INCLUDING THE DNA, AND CELLS TRANSFORMED BY THE VECTOR
US20030206898A1 (en) 2002-04-26 2003-11-06 Steven Fischkoff Use of anti-TNFalpha antibodies and another drug
AU2003267999B2 (en) 2002-07-19 2010-03-11 Abbvie Biotechnology Ltd Treatment of TNFalpha related disorders
BR0313491A (en) * 2002-08-16 2007-08-14 Wyeth Corp compositions and methods for treating rage-associated disorders
WO2004024915A1 (en) * 2002-09-13 2004-03-25 The University Of Queensland Gene expression system based on codon translation efficiency
MY150740A (en) 2002-10-24 2014-02-28 Abbvie Biotechnology Ltd Low dose methods for treating disorders in which tnf? activity is detrimental
BRPI0407649A (en) 2003-02-28 2006-02-21 Ares Trading Sa liquid formulation of tumor necrosis factor binding proteins, process for preparing liquid formulation of tumor necrosis factor binding proteins and formulation presentation form
US20080241884A1 (en) * 2003-10-08 2008-10-02 Kenya Shitara Fused Protein Composition
TWI439284B (en) 2004-04-09 2014-06-01 Abbvie Biotechnology Ltd Multiple-variable dose regimen for treating tnfα-related disorders
GB0414054D0 (en) 2004-06-23 2004-07-28 Owen Mumford Ltd Improvements relating to automatic injection devices
CN102746404B (en) * 2004-11-12 2016-01-20 赞科股份有限公司 To FcRn in conjunction with reformed Fc variant
AU2006230419A1 (en) * 2005-03-31 2006-10-05 Targeted Genetics Corporation Methods for lowering the level of tumor necrosis factor (TNF) in TNF-associated disorders
NZ591701A (en) 2005-05-16 2012-11-30 Abbott Biotech Ltd Use of tnf inhibitor for treatment of erosive polyarthritis
US20070041905A1 (en) 2005-08-19 2007-02-22 Hoffman Rebecca S Method of treating depression using a TNF-alpha antibody
EP1948235B1 (en) 2005-11-01 2013-08-28 AbbVie Biotechnology Ltd Methods for determining efficacy of adalimumab in subjects having ankylosing spondylitis using ctx-ii and mmp3 as biomarkers
US7785834B2 (en) 2005-11-10 2010-08-31 Ercole Biotech, Inc. Soluble TNF receptors and their use in treatment of disease
EP2089521A1 (en) * 2006-02-08 2009-08-19 Ercole Biotech, Inc. Soluble tnf receptors and their use in treatment of disease
WO2007109686A2 (en) 2006-03-20 2007-09-27 University Of Pittsburgh Immunomodulation of inflammatory conditions utilizing follistatin-like protein-1 and agents that bind thereto
US8007790B2 (en) 2006-04-03 2011-08-30 Stowers Institute For Medical Research Methods for treating polycystic kidney disease (PKD) or other cyst forming diseases
NZ611859A (en) 2006-04-05 2014-12-24 Abbvie Biotechnology Ltd Antibody purification
EP2666478A3 (en) 2006-04-10 2014-10-22 AbbVie Biotechnology Ltd Uses and compositions for treatment of psoriasis
CA2564435A1 (en) 2006-04-10 2007-10-10 Abbott Biotechnology Ltd. Methods for monitoring and treating intestinal disorders
US9624295B2 (en) 2006-04-10 2017-04-18 Abbvie Biotechnology Ltd. Uses and compositions for treatment of psoriatic arthritis
TWI498137B (en) 2006-06-30 2015-09-01 Abbvie Biotechnology Ltd Automatic injection device
US8513393B2 (en) 2006-08-28 2013-08-20 Ares Trading S.A. Process for the purification of Fc-containing proteins
JP2010501622A (en) * 2006-08-28 2010-01-21 アレス トレーディング ソシエテ アノニム Purification method of Fc-fusion protein
WO2008025747A1 (en) * 2006-08-28 2008-03-06 Ares Trading S.A. Process for the purification of fc-fusion proteins
US7767206B2 (en) 2006-10-02 2010-08-03 Amgen Inc. Neutralizing determinants of IL-17 Receptor A and antibodies that bind thereto
US8338376B2 (en) 2006-10-20 2012-12-25 Biogen Idec Ma Inc. Compositions comprising variant LT-B-R-IG fusion proteins
EP2311481A3 (en) 2006-10-20 2013-10-16 Biogen Idec MA Inc. Treatment of demyelinating disorders with soluble lymphotoxin-beta-receptor
EP2679996A1 (en) 2007-05-31 2014-01-01 AbbVie Inc. Biomarkers predictive of the responsiveness to TNF-alfa inhibitors in autoimmune disorders
US8999337B2 (en) 2007-06-11 2015-04-07 Abbvie Biotechnology Ltd. Methods for treating juvenile idiopathic arthritis by inhibition of TNFα
US8398977B2 (en) 2007-06-14 2013-03-19 Galactica Pharmaceuticals, Inc. Rage fusion proteins
EP2072527A1 (en) 2007-12-21 2009-06-24 Altonabiotec AG Fusion polypeptides comprising a SHBG dimerization component and uses thereof
US8658379B2 (en) 2008-01-29 2014-02-25 University of Pittsburgh—of the Commonwealth System of Higher Education Follistatin-like protein-1 as a biomarker for sepsis
AU2009280913A1 (en) * 2008-08-12 2010-02-18 Avesthagen Limited An expression vector and a method thereof
JP5684724B2 (en) 2008-12-30 2015-03-18 ヤンセン バイオテツク,インコーポレーテツド Serum markers to predict clinical response to anti-TNFα antibodies in patients with ankylosing spondylitis
JP5677411B2 (en) 2009-04-29 2015-02-25 アッヴィ バイオテクノロジー リミテッド Automatic injection equipment
TW201117824A (en) 2009-10-12 2011-06-01 Amgen Inc Use of IL-17 receptor a antigen binding proteins
WO2011068993A1 (en) 2009-12-02 2011-06-09 Acceleron Pharma Inc. Compositions and methods for increasing serum half-life of fc fusion proteins.
AU2010330907A1 (en) 2009-12-16 2012-06-14 Bosch, Phillip Methods of treating interstitial cystitis
JO3417B1 (en) 2010-01-08 2019-10-20 Regeneron Pharma Stabilized formulations containing anti-interleukin-6 receptor (il-6r) antibodies
DK2523688T3 (en) 2010-01-15 2017-12-04 Kirin-Amgen Inc ANTIBODY FORMULATION AND THERAPEUTIC REGIMENS
US20120014956A1 (en) 2010-02-02 2012-01-19 Hartmut Kupper Methods and compositions for predicting responsiveness to treatment with tnf-alpha inhibitor
SG10201503130UA (en) 2010-04-21 2015-06-29 Abbvie Biotechnology Ltd Wearable automatic injection device for controlled delivery of therapeutic agents
WO2011146727A1 (en) 2010-05-19 2011-11-24 Philip Bosch Methods of treating interstitial cystitis
US8747854B2 (en) 2010-06-03 2014-06-10 Abbvie Biotechnology Ltd. Methods of treating moderate to severe hidradenitis suppurativa with anti-TNF-alpha antibodies
FR2962335B1 (en) 2010-07-12 2013-01-18 Cll Pharma USE OF PAT NONAPEPTIDE IN THE TREATMENT AND PREVENTION OF NEURODEGENERATIVE DISEASES
WO2012019099A2 (en) 2010-08-05 2012-02-09 University Of Pittsburgh - Of The Commonwealth System Of Higher Education Follistatin-like-protein-1 as a biomarker for inflammatory disorders
MX337208B (en) 2011-01-24 2016-02-17 Abbvie Biotechnology Ltd Automatic injection devices having overmolded gripping surfaces.
CA2829773A1 (en) 2011-03-18 2012-09-27 Abbvie Inc. Systems, devices and methods for assembling automatic injection devices and sub-assemblies thereof
JP2014515658A (en) 2011-03-29 2014-07-03 アッヴィ・インコーポレイテッド Improved shroud deployment in automatic injection devices
EP3020428A1 (en) 2011-04-21 2016-05-18 AbbVie Inc. Wearable automatic injection device for controlled administration of therapeutic agents
UY34105A (en) 2011-06-03 2012-07-31 Lg Life Sciences Ltd STABLE LIQUID FORMULATION OF ETANERCEPT
WO2012170938A1 (en) 2011-06-08 2012-12-13 Acceleron Pharma Inc. Compositions and methods for increasing serum half-life
US20140234330A1 (en) 2011-07-22 2014-08-21 Amgen Inc. Il-17 receptor a is required for il-17c biology
TWI589299B (en) 2011-10-11 2017-07-01 再生元醫藥公司 Compositions for the treatment of rheumatoid arthritis and methods of using same
US9943594B2 (en) 2011-10-11 2018-04-17 Sanofi Biotechnology Methods for the treatment of rheumatoid arthritis
JP6220789B2 (en) 2011-10-18 2017-10-25 コヒラス・バイオサイエンシズ・インコーポレイテッド Etanercept formulation stabilized by a combination of sugar and polyol
KR101525919B1 (en) * 2012-05-11 2015-06-03 가톨릭대학교 산학협력단 Bispecific antibody based on TNFR2 for preventing and traeating autoimmune disease
CN104902914B (en) 2012-09-11 2019-01-01 科荣生生物科学公司 The Etanercept of high-purity and excellent yield correctly folded
EP3126392B1 (en) 2014-03-31 2019-09-11 Kirin-Amgen, Inc. Methods of treating nail and scalp psoriasis
US20170183376A1 (en) 2014-06-24 2017-06-29 Insight Biopharmaceuticals Ltd. Methods of purifying antibodies
CA2970577A1 (en) 2014-12-22 2016-06-30 Ares Trading S.A. Liquid pharmaceutical composition
EP3078675A1 (en) 2015-04-10 2016-10-12 Ares Trading S.A. Induction dosing regimen for the treatment of tnf alpha mediated disorders
US20170306050A1 (en) 2016-04-08 2017-10-26 Gilead Sciences, Inc. Compositions and methods for treating cancer, inflammatory diseases and autoimmune diseases
EP3257866A1 (en) 2016-06-17 2017-12-20 Academisch Medisch Centrum Modified anti-tnf antibody and use thereof in the treatment of ibd
WO2018112240A1 (en) 2016-12-14 2018-06-21 Progenity Inc. Treatment of a disease of the gastrointestinal tract with a tnf inhibitor
JOP20190162A1 (en) 2016-12-30 2019-06-27 Biocad Joint Stock Co Aqueous Pharmaceutical Composition of a Recombinant Monoclonal Antibody to TNF?
EP3810095A1 (en) 2018-06-20 2021-04-28 Progenity, Inc. Treatment of a disease of the gastrointestinal tract with a tnf inhibitor
CN112654350A (en) 2018-07-03 2021-04-13 诺华股份有限公司 Methods of treating a subject resistant to a TNF inhibitor or selecting a treatment for said subject using an NLRP3 antagonist
CN112955222A (en) 2018-08-29 2021-06-11 里珍纳龙药品有限公司 Methods and compositions for treating individuals with rheumatoid arthritis
CN114206442A (en) 2019-01-31 2022-03-18 赛诺菲生物技术公司 anti-IL-6 receptor antibodies for the treatment of juvenile idiopathic arthritis
CN110964119A (en) * 2019-12-05 2020-04-07 沣潮医药科技(上海)有限公司 Anti-malarial dimeric immunoadhesin, pharmaceutical composition and use
JP2023554200A (en) 2020-12-09 2023-12-26 エイチケー イノ.エヌ コーポレーション Anti-OX40L antibody, anti-OX40L and anti-TNFα bispecific antibody, and their uses

Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1991003553A1 (en) * 1989-09-05 1991-03-21 Immunex Corporation TUMOR NECROSIS FACTOR-α AND -β RECEPTORS
WO1992013095A1 (en) * 1991-01-18 1992-08-06 Synergen, Inc. Methods for treating tumor necrosis factor mediated diseases

Family Cites Families (2)

* 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
WO1994006431A1 (en) * 1992-09-22 1994-03-31 Cell Therapeutics, Inc. Novel epoxide-containing compounds

Patent Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1991003553A1 (en) * 1989-09-05 1991-03-21 Immunex Corporation TUMOR NECROSIS FACTOR-α AND -β RECEPTORS
WO1992013095A1 (en) * 1991-01-18 1992-08-06 Synergen, Inc. Methods for treating tumor necrosis factor mediated diseases

Non-Patent Citations (3)

* Cited by examiner, † Cited by third party
Title
1992 MEETING OF THE FEDERATION OF AMERICAN SOCIETIES FOR EXPERIMENTAL BIOLOGY (FASEB), PART I, ANAHEIM, CALIFORNIA, USA, APRIL 5-9, 1992. FASEB (FED AM SOC EXP BIOL) J 6 (4). 1992. A1123, XP002015786 MOHLER K M ET AL: "IMMUNOTHERAPEUTIC POTENTIAL OF SOLUBLE CYTOKINE RECEPTORS IN INFLAMMATORY DISEASE." *
KEYSTONE SYMPOSIUM ON CYTOKINES AND CYTOKINE RECEPTORS: FROM CLONING TO THE CLINIC, KEYSTONE, COLORADO, USA, JANUARY 31-FEBRUARY 7, 1993. J CELL BIOCHEM SUPPL 0 (17 PART B). 1993. 56, XP002015787 MOHLER K M ET AL: "COUNTERACTING THE EFFECTS OF TNF WITH SOLUBLE TNF RECEPTORS." *
See also references of WO9406476A1 *

Also Published As

Publication number Publication date
CA2123593C (en) 2000-03-14
NO941780D0 (en) 1994-05-11
WO1994006476A1 (en) 1994-03-31
AU670125B2 (en) 1996-07-04
NZ256293A (en) 1997-06-24
NO941780L (en) 1994-07-15
CA2123593A1 (en) 1994-03-31
JPH07504203A (en) 1995-05-11
EP0620739A4 (en) 1997-01-15
KR100232688B1 (en) 1999-12-01
AU4920993A (en) 1994-04-12

Similar Documents

Publication Publication Date Title
AU670125B2 (en) Method of treating tnf-dependent inflammation using tumor necrosis factor antagonists
US5605690A (en) Methods of lowering active TNF-α levels in mammals using tumor necrosis factor receptor
US6201105B1 (en) Tumor necrosis factor receptor polypeptides recombinant P75 (Type II)
EP0418014B1 (en) Tumor necrosis factor-alpha and -beta receptors
CA2065346C (en) Tumor necrosis factor-.alpha. and-.beta. receptors
US5945397A (en) Purified p75 (type II) tumor necrosis factor receptor polypeptides
EP0460846B1 (en) Type II interleukin-1 receptors
AU646695B2 (en) Isolated viral protein cytokine antagonists
AU647566B2 (en) Leukemia inhibitory factor receptors
IE83679B1 (en) Type II interleukin-1 receptors
NZ251820A (en) Fusion proteins with tnf-r (tumour necrosis factor-receptor) peptide linked to another tnf-r or an il-1r (interleukin-1-receptor) peptide
US7700318B2 (en) Chimeric polypeptide and use thereof
IE911108A1 (en) Isolated Viral Protein Cytokine Antagonists

Legal Events

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

Free format text: ORIGINAL CODE: 0009012

17P Request for examination filed

Effective date: 19940518

AK Designated contracting states

Kind code of ref document: A1

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

A4 Supplementary search report drawn up and despatched

Effective date: 19961127

AK Designated contracting states

Kind code of ref document: A4

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

17Q First examination report despatched

Effective date: 19970228

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

Free format text: STATUS: THE APPLICATION IS DEEMED TO BE WITHDRAWN

18D Application deemed to be withdrawn

Effective date: 19980103