EP2195337A1 - Trimeres il-1ra - Google Patents

Trimeres il-1ra

Info

Publication number
EP2195337A1
EP2195337A1 EP08837224A EP08837224A EP2195337A1 EP 2195337 A1 EP2195337 A1 EP 2195337A1 EP 08837224 A EP08837224 A EP 08837224A EP 08837224 A EP08837224 A EP 08837224A EP 2195337 A1 EP2195337 A1 EP 2195337A1
Authority
EP
European Patent Office
Prior art keywords
seq
ira
fusion protein
lra
protein
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Withdrawn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
EP08837224A
Other languages
English (en)
French (fr)
Inventor
Thor Las Holtet
Mikkel Holmen Andersen
Josephus Dirk Nieland
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.)
Anaphore Inc
Original Assignee
Anaphore Inc
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Anaphore Inc filed Critical Anaphore Inc
Publication of EP2195337A1 publication Critical patent/EP2195337A1/de
Withdrawn legal-status Critical Current

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/52Cytokines; Lymphokines; Interferons
    • C07K14/54Interleukins [IL]
    • C07K14/545IL-1
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • A61P1/04Drugs for disorders of the alimentary tract or the digestive system for ulcers, gastritis or reflux esophagitis, e.g. antacids, inhibitors of acid secretion, mucosal protectants
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • A61P1/18Drugs for disorders of the alimentary tract or the digestive system for pancreatic disorders, e.g. pancreatic enzymes
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P11/00Drugs for disorders of the respiratory system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P11/00Drugs for disorders of the respiratory system
    • A61P11/06Antiasthmatics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P13/00Drugs for disorders of the urinary system
    • A61P13/12Drugs for disorders of the urinary system of the kidneys
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P17/00Drugs for dermatological disorders
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P17/00Drugs for dermatological disorders
    • A61P17/06Antipsoriatics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P19/00Drugs for skeletal disorders
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P19/00Drugs for skeletal disorders
    • A61P19/02Drugs for skeletal disorders for joint disorders, e.g. arthritis, arthrosis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P19/00Drugs for skeletal disorders
    • A61P19/06Antigout agents, e.g. antihyperuricemic or uricosuric agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P19/00Drugs for skeletal disorders
    • A61P19/08Drugs for skeletal disorders for bone diseases, e.g. rachitism, Paget's disease
    • A61P19/10Drugs for skeletal disorders for bone diseases, e.g. rachitism, Paget's disease for osteoporosis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P21/00Drugs for disorders of the muscular or neuromuscular system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/04Centrally acting analgesics, e.g. opioids
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/14Drugs for disorders of the nervous system for treating abnormal movements, e.g. chorea, dyskinesia
    • A61P25/16Anti-Parkinson drugs
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/28Drugs for disorders of the nervous system for treating neurodegenerative disorders of the central nervous system, e.g. nootropic agents, cognition enhancers, drugs for treating Alzheimer's disease or other forms of dementia
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P29/00Non-central analgesic, antipyretic or antiinflammatory agents, e.g. antirheumatic agents; Non-steroidal antiinflammatory drugs [NSAID]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P3/00Drugs for disorders of the metabolism
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P3/00Drugs for disorders of the metabolism
    • A61P3/08Drugs for disorders of the metabolism for glucose homeostasis
    • A61P3/10Drugs for disorders of the metabolism for glucose homeostasis for hyperglycaemia, e.g. antidiabetics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/04Antibacterial agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • A61P35/02Antineoplastic agents specific for leukemia
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • A61P35/04Antineoplastic agents specific for metastasis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/02Immunomodulators
    • A61P37/06Immunosuppressants, e.g. drugs for graft rejection
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • A61P9/10Drugs for disorders of the cardiovascular system for treating ischaemic or atherosclerotic diseases, e.g. antianginal drugs, coronary vasodilators, drugs for myocardial infarction, retinopathy, cerebrovascula insufficiency, renal arteriosclerosis
    • 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
    • C07K14/46Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates
    • C07K14/47Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates from mammals
    • C07K14/4701Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates from mammals not used
    • C07K14/4713Autoimmune diseases, e.g. Insulin-dependent diabetes mellitus, multiple sclerosis, rheumathoid arthritis, systemic lupus erythematosus; Autoantigens
    • 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
    • C07K14/46Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates
    • C07K14/47Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates from mammals
    • C07K14/4701Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates from mammals not used
    • C07K14/4726Lectins
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/11DNA or RNA fragments; Modified forms thereof; Non-coding nucleic acids having a biological activity
    • C12N15/62DNA sequences coding for fusion proteins
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2319/00Fusion polypeptide
    • C07K2319/31Fusion polypeptide fusions, other than Fc, for prolonged plasma life, e.g. albumin

Definitions

  • the invention relates to treatment of diseases that are mediated by interleukin 1. More particularly, the invention relates to interleukin 1 receptor antagonists (IL-IRa) that are useful for treating such diseases.
  • IL-IRa interleukin 1 receptor antagonists
  • the IL-I family is an important part of the innate immune system, which is a regulator of the adaptive immune system.
  • the balance between IL-I and IL-IRa in local tissues influences the possible development of inflammatory diseases and resulting structural damage.
  • inflammatory and autoimmune diseases may develop in the joints, lungs, gastrointestinal tract, central nervous system (CNS), or blood vessels.
  • CNS central nervous system
  • Treatment of human disease with IL-IRa has been carried out through injection of recombinant IL-IRa or through gene therapy approaches.
  • Treatment with recombinant IL-IRa has been approved for rheumatoid arthritis (RA) and phase 2 studies are ongoing for osteoarthritis (OA).
  • RA rheumatoid arthritis
  • OA osteoarthritis
  • IL-I An important pro-inflammatory role for IL-I in many human diseases has been described over the past 10 years.
  • RA rheumatoid arthritis
  • OA osteoarthritis
  • IBD inflammatory bowel disease
  • GVHD graft-versus-host disease
  • leukemia cancer
  • osteoporosis granulomatous and fibrotic lung disorders
  • diabetes diseases of the liver and pancreas
  • GVHD graft-versus-host disease
  • leukemia cancer
  • osteoporosis granul
  • the IL-I family consists of two agonists, IL-l ⁇ and IL-I ⁇ ; the specific receptor antagonist IL-IRa; and three different receptors, EL-IR type I (IL-IRI), IL-IR type II (IL- IRII) and IL-I receptor accessory protein (IL-IR AcP).
  • IL-IRI is an 80 kDa protein with a long cytoplasmic domain of 215 residues.
  • the biologically inert IL-IRII is a 60 kDa protein with a short cytoplasmic domain of 29 residues.
  • IL-IR AcP is recruited to the complex after binding of IL-l ⁇ or IL- l ⁇ to the single chain IL-IRI.
  • IL-IRII functions as a decoy receptor, binding IL-I both on the plasma membrane and as a soluble receptor in the fluid phase, thereby preventing IL-I from interacting with the functional IL-IRI.
  • IL-IRa The third ligand in the IL-I family, IL-IRa, is a structural variant of IL-I that binds to both IL-IR but fails to activate cells.
  • IL-IRa is a 17 kDa protein with 18% amino acid homology to IL-l ⁇ and 26% homology to IL-l ⁇ .
  • the originally described isoform of IL- IRa is secreted from monocytes, macrophages, neutrophils, and other cells and is now termed sIL-IRa. Three additional intracellular isoforms of IL-IRa have been described to date.
  • IL-IRa 18 kDa form of IL-IRa, created by an alternative transcriptional splice mechanism from an upstream exon is called icIL-lRal and is found inside keratinocytes and other epithelial cells, monocytes, tissue macrophages, fibroblasts, and endothelial cells.
  • IL-IRa cDNA cloned from human leukocytes contains an additional 63 bp sequence as an insert in the 5' region of the cDNA.
  • a 15 kDa isoform of IL-IRa termed icIL-lRa3, is found in monocytes, macrophages, neutrophils, and hepatocytes, and may be created both by an alternative transcriptional splice as well as by alternative translational initiation.
  • IL-IRa functions as a specific receptor antagonist by binding to IL-IRI but preventing IL-IR AcP from associating with the IL-IRI, which results failure of initiation of signal transduction pathways.
  • the decoy receptor IL-IRII binds IL-I both on the plasma membrane and as a soluble receptor in the fluid phase, preventing IL-I from interacting with the functional IL- IRI. Therefore, soluble IL-IRII and IL-IRa can inhibit IL-I in co-operation. Soluble IL-RI can bind to IL-I as well as IL-IRa, but due to the balance between IL-I and IL-IRa, soluble IL-IRI seems to act as a pro-inflammatory agent.
  • BQNERET ® is an E. coli produced IL-IRa from Amgen, which has been shown to benefit patients with active rheumatoid arthritis.
  • KINERET has to be injected subcutaneously once per day. With subcutaneous administration, KINERET ® has a half-life ranged from 4 to 6 hours; for i.v. administration the half life is approximately 2/4 hours.
  • IL- IRa is cleared by renal clearance.
  • KINERET is a specific receptor antagonist of IL-I that differs from naturally occurring IL-I receptor antagonist by the presence of a methionine group.
  • KINERET ® When given alone or in combination with methotrexate, KINERET ® has been shown to benefit patients as assessed by improvement in clinical signs and symptoms, decreased radiographic progression and improvement in patient function, pain and fatigue. KINERET ® has a favorable safety profile as demonstrated in clinical trials.
  • the inventors have identified a need in the art for an improved delivery method for ILl-Ra, which provides for a longer half-life of the molecule and provides a favorable safety profile.
  • Figure 1 shows alignment of the amino acid sequences of the trimerising structural element of the tetranectin protein family.
  • Amino acid sequences (one letter code) corresponding to residue Vl 7 to K52 comprising exon 2 and the first three residues of exon 3 of human tetranectin (SEQ ID NO: 59); murine tetranectin (SEQ ID NO: 60); tetranectin homologous protein isolated from reefshark cartilage (SEQ ID NO: 61) and tetranectin homologous protein isolated from bovine cartilage (SEQ ID NO: 62. Residues at a and d positions in the heptad repeats are listed in boldface.
  • the listed consensus sequence of the tetranectin protein family trimerising structural element comprise the residues present at a and d positions in the heptad repeats shown in the figure in addition to the other conserved residues of the region, "hy” denotes an aliphatic hydrophobic residue.
  • Figure 2 shows the results of CII-H6-GrB-TripK-IL-lRa refolding by dialysis.
  • Figure 3 displays the capturing CII-H6-GrB-TripK-IL- 1 Ra on NiNTA.
  • Figure 4 is a graph showing the ability of GG-TripV-IL-lRa (trip V-IL-IRa), GG- TripK-IL-lRa (trip K-IL-IRa), GG-TripT-IL-lRa (trip T-IL-IRa) and GG-TripT-IL-lRa (trip T-IL-IRa) to inhibit IL-I induction of IL-8 in U937 cells.
  • Figure 5 is a graph showing the ability of pegylated TripT and TripV to inhibit IL- 1 induction of IL-8 in U937 cells as compared to non-pegylated forms and KINERET ® .
  • Figure 6 is a graph showing the ability of TripT-IL-lRa, I10-TripT-IL-lRa, V17- TripT-IL-lRa used in the PK study to inhibit IL-I induction of IL-8 in U937 cells
  • Figure 7 is a graph showing the blood concentrations of TripT-IL- 1 Ra, 110-TripT- IL-IRa, and V17-TripT-IL-lRa after lOOmg/kg i.v. injection in rats.
  • Figure 8 shows an SDS-PAGE analysis of multiple batches of Met-110-TrpT-IL- IRa (LM022 and LM023) and GG-V 17-TrpT-IL- IRa (CF019 and CF020) protein yields.
  • Figure 9 shows analytical SEC results of Met-I10-TrpT-IL-lRa and GG-V17- TrpT-IL-lRa protein yields.
  • Figure 10 shows the results of the rat CIA study. Ankle diameters of female Lewis rats with type II collagen arthritis were measured following treatment with Vehicle (10 mM phosphate buffer pH 7.4), or equimolar amounts of IL- Ira administering either monomeric IL-lra (100 mg/kg KINERET ® ), or trimerized ILlra (120 mg/lcg Met-IlO-TripT- ILlra, or 120 mg/kg GG-V 17-TripT-IL Ira).
  • Vehicle 10 mM phosphate buffer pH 7.4
  • IL- Ira administering either monomeric IL-lra (100 mg/kg KINERET ® ), or trimerized ILlra (120 mg/lcg Met-IlO-TripT- ILlra, or 120 mg/kg GG-V 17-TripT-IL Ira).
  • Figure 11 shows study reduction of final paw weight when rats treated with KINERET ® , Met-IlO-TripT-ILlra QD, or GG-V 17-TripT-IL Ira QD, as compared to vehicle treated disease control animals.
  • Figure 12 shows reduction of blood glucose levels observed after daily i.p. dosing of either I10-TripT-ILl-Ra or KINERET ®
  • the present invention provides a fusion protein comprising a trimerizing domain and an IL-IRa polypeptide sequence that inhibits IL-I activity.
  • the fusion protein comprises an IL-IRa sequence that comprises a variant or fragment of SEQ ID NO: 38 that inhibits IL-I activity.
  • the fusion protein comprises an IL-IRa polypeptide sequence that is at least 85% identical to SEQ ID NO: 38.
  • the fusion proteins may include polyethylene glycol.
  • the trimerizing domain of the fusion protein may be derived from tetranectin.
  • the present invention also provides a trimeric complex comprising three fusion proteins of the invention.
  • the trimeric complex comprises a trimerizing domain that is a tetranectin trimerizing structural element (TTSE).
  • TTSE tetranectin trimerizing structural element
  • the trimeric complex comprises a trimerizing domain is at least 66% identical to SEQ ID NO: 1.
  • the trimeric complex comprises at least one of the fusion proteins selected from the group consisting of TripK-IL-lra (SEQ ID NO: 39); TripV-IL-lra (SEQ ID NO: 40); TripT-IL-lra (SEQ ID NO: 41); TripQ-IL-lra (SEQ ID NO: 42); I10-TripK-IL-lra (SEQ ID NO: 43); I10-TripV-IL-lra (SEQ ID NO: 44); I10-TripT-IL-lra (SEQ ID NO: 45); I10-TripQ- ⁇ L-lra (SEQ ID NO: 46); V17-TripT-ILlRa (SEQ ID NO: 55); V17-TripK-IL- IRa (SEQ ID NO: 56); V17-TripV-IL-lRA (SEQ ID NO: 57); and V17-TripQ-ILlRA (SEQ ID NO: 58).
  • TripK-IL-lra
  • the present invention provides a pharmaceutical composition comprising a trimeric and at least one pharmaceutically acceptable excipient.
  • the invention is directed to a method for treating a disease mediated by interleukin 1.
  • the method includes administering to a patient in need thereof of the pharmaceutical composition of the invention.
  • the disease may be an inflammatory disease such as rheumatoid arthritis or diabetes.
  • the method also includes administering to the patient, either simultaneously or sequentially, an anti-inflammatory agent.
  • the invention also provides a fusion protein further comprising an antiinflammatory agent covalently linked to the fusion protein.
  • the invention is directed to compounds and methods for treating diseases mediated by IL-I .
  • the invention is directed to a fusion protein of an IL-IRa polypeptide sequence fused to a trimerizing or multimerizing domain.
  • Three or more of fusion proteins may trimerize or multimerize to provide compositions providing for greater stability and improved pharmacokinetic properties than IL-IRa alone, and provide a favorable safety profile.
  • the invention provides a nucleic acid which encodes any one of the polypeptides defined above, as well as methods of preparing these polypeptides under conditions that allow for specific expression and recovery.
  • polypeptides of the invention may be used for the preparation of pharmaceutical compositions for use in the treatment of a subject having a pathology mediated by IL-I, such as a method of treatment of inflammatory diseases, by administering to the subject an effective amount of pharmaceutical composition.
  • a disease or medical condition is considered to be an "interleukin- 1 mediated disease” or "a disease mediated by interleukin-1” if the spontaneous or experimental disease or medical condition is associated with elevated levels of IL-I in bodily fluids or tissue or if cells or tissues taken from the body produce elevated levels of IL-I in culture.
  • interleukin-1 mediated diseases are also recognized by the following additional two conditions: (1) pathological findings associated with the disease or medical condition can be mimicked experimentally in animals by the administration of IL-I; and (2) the pathology induced in experimental animal models of the disease or medical condition can be inhibited or abolished by treatment with agents which inhibit the action of IL-I.
  • IL-I mediated diseases at least two of the three conditions are met, and in many IL-I mediated diseases all three conditions are met.
  • a non-exclusive list of acute and chronic IL-I -mediated inflammatory diseases includes but is not limited to the following: gout, acute pancreatitis; ALS; Alzheimer's disease; cachexia/anorexia; asthma; atherosclerosis; chronic fatigue syndrome, fever; diabetes (e.g., insulin diabetes); glomerulonephritis; graft versus host rejection; hemohorragic shock; hyperalgesia, inflammatory bowel disease; inflammatory conditions of a joint, including osteoarthritis, psoriatic arthritis, juvenile arthritis, and rheumatoid arthritis; ischemic injury, including cerebral ischemia (e.g., brain injury as a result of trauma, epilepsy, hemorrhage or stroke, each of which may lead to neurodegeneration); lung diseases (e.g., ARDS); multiple myeloma; multiple s
  • osteoporosis in sepsis); osteoporosis; Parkinson's disease; pain; pre-term labor; psoriasis; reperfusion injury; septic shock; side effects from radiation therapy, temporal mandibular joint disease, tumor metastasis; or an inflammatory condition resulting from strain, sprain, cartilage damage, trauma, orthopedic surgery, infection or other disease processes, and Cryopyrin-associated periodic syndromes, including Muckle Wells syndrome, familial cold autoinflammatory syndrome and neonatal-onset multisystem inflammatory disease.
  • the term "multimerizing domain” means an amino acid sequence that comprises the functionality that can associate with two or more other amino acid sequences to form trimers or other multimerized complexes.
  • the fusion protein contains an amino acid sequence — a trimerizing domain — which forms a trimeric complex with two other trimerizing domains.
  • a trimerizing domain can associate with other trimerizing domains of identical amino acid sequence (a homotrimer), or with trimerizing domains of different amino acid sequence (a heterotrimer). Such an interaction may be caused by covalent bonds between the components of the trimerizing domains as well as by hydrogen bond forces, hydrophobic forces, van der Waals forces and salt bridges.
  • the multimerizing domain is a dimerizing, domain, a trimerizing domain, a tetramerizing domain, a pentamerizing domain, etc. These domains are capaple of forming polypeptide complexes of two, three, four, five or more fusion proteins of the invention.
  • the trimerizing domain of a fusion protein of the invention may be derived from tetranectin as described in U.S. Patent Application Publication No. 2007/0154901 ('901 Application), which is incorporated by reference in its entirety.
  • the full length human tetranectin polypeptide sequence is provided herein as SEQ ID NO: 63.
  • Examples of a tetranectin trimerizing domain includes the amino acids 17 to 49, 17 to 50, 17 to 51 and 17- 52 of SEQ ID NO: 63, which represent the amino acids encoded by exon 2 of the human tetranectin gene, and optionally the first one, two or three amino acids encoded by exon 3 of the gene.
  • amino acids 1 to 49, 1 to 50, 1 to 51 and 1 to 52 which represents all of exons 1 and 2, and optionally the first one, two or three amino acids encoded by exon 3 of the gene.
  • the N-terminus of the trimerizing domain may begin at any of residues 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16 and 17 of SEQ ID NO: 63.
  • the N terminus is 110 or V17 and the C- terminus is Q47, T48, V49, C(S)50, L51 or K52 (numbering according to SEQ ID NO: 63).
  • the trimerizing domain is a tetranectin trimerizing structural element ("TTSE") having a amino acid sequence of SEQ ID NO: 1 which a consensus sequence of the tetranectin family trimerizing structural element as more fully described in US 2007/00154901.
  • TTSE tetranectin trimerizing structural element
  • the TTSE embraces variants of a naturally occurring member of the tetranectin family of proteins, and in particular variants that have been modified in the amino acid sequence without adversely affecting, to any substantial degree, the ability of the TTSE to form alpha helical coiled coil trimers.
  • the trimeric polypeptide according to the invention includes a TTSE as a trimerizing domain having at least 66% amino acid sequence identity to the consensus sequence of SEQ ID NO: 1; for example at least 73%, at least 80%, at least 86% or at least 92% sequence identity to the consensus sequence of SEQ ID NO: 1 (counting only the defined (not Xaa) residues).
  • TTSE trimerizing domain having at least 66% amino acid sequence identity to the consensus sequence of SEQ ID NO: 1; for example at least 73%, at least 80%, at least 86% or at least 92% sequence identity to the consensus sequence of SEQ ID NO: 1 (counting only the defined (not Xaa) residues).
  • at least one, at least two, at least three, at least four, or at least five of the defined amino acids in SEQ ID NO: 1 may be substituted.
  • the cysteine at position 50 (C50) of SEQ ID NO: 63 can be advantageously be mutagenized to serine, threonine, methionine or to any other amino acid residue in order to avoid formation of an unwanted inter-chain disulphide bridge, which can lead to unwanted multimerization.
  • Other known variants include at least one amino acid residue selected from amino acid residue nos. 6, 21, 22, 24, 25, 27, 28, 31, 32, 35, 39, 41, and 42 (numbering according to SEQ ID NO:63), which may be substituted by any non-helix breaking amino acid residue. These residues have been shown not to be directly involved in the intermolecular interactions that stabilize the trimeric complex between three TTSEs of native tetranectin monomers.
  • the TTSE has a repeated heptad having the formula a-b-c-d-e-f-g (N to C), wherein residues a and d (i.e., positions 26, 33, 37, 40, 44, 47, and 51 may be any hydrophobic amino acid (numbering according to claim 63).
  • the TTSE trimerization domain may be modified by the incorporation of polyhistidine sequence and/or a protease cleavage site, e.g, Blood Coagulating Factor Xa or Granzyme B (see US 2005/0199251, which is incorporated herein by reference), and by including a C-terminal KG or KGS sequence.
  • a protease cleavage site e.g, Blood Coagulating Factor Xa or Granzyme B (see US 2005/0199251, which is incorporated herein by reference
  • Proline at position 2 may be substituted with Glycine to assist in purification.
  • TTSE truncations and variants are shown in Table 1 below.
  • trimerizing domain is disclosed in US 6, 190,886 (incorporated herein in its entirety), which describes polypeptides comprising a collectin neck region. Trimers can then be made under appropriate conditions with three polypeptides comprising the collectin neck region amino acid sequence.
  • trimerizing domain is an MBP trimerizing domain, as described in U.S. Provisional Patent Application Serial No. 60/996,288, filed by the assignee of the present application on November 9, 2007, which is incorporated by reference in its entirety.
  • This trimerizing domain can oligomerize even further and create higher order multimeric complexes.
  • the IL-I Ra polypeptide of the invention may either be linked to the N- or the C- terminal amino acid residue of the trimerization domain.
  • a flexible molecular linker optionally may be interposed between, and covalently join, the polypeptide representing the IL-I Ra and the trimerization domain.
  • the linker is a polypeptide sequence of about 1 to 20, 2 to 10, or 3 to 7 amino acid residues.
  • the linker is non-immunogenic, not prone to proteolytic cleavage, and does not comprise amino acid residues which are known to interact with other residues (e.g. cystein residues).
  • IL-I Ra refers to a polypeptide having the amino acid sequence shown below:
  • IL-IRa also included in the "IL-IRa" definition are variants and fragments of SEQ ID NO: 38 that provide for IL-IRa binding to IL-IR, and preferably IL-IR inhibitory activity.
  • Such fragments may be truncated at the N-terminus or C-terminus of the IL-IRa, or may lack internal residues, when compared with the full length native IL-IRa protein.
  • Certain fragments may lack amino acid residues that are not essential for a desired biological activity of the trimeric IL-I Ra protein according to the invention.
  • Evans, et al. J. Biol. Chem.
  • IL-IRa variants exist, any of which may be used.
  • An 18 kDa form of IL-IRa created by an alternative transcriptional splice mechanism from an upstream exon is called icIL-lRal and is found inside keratinocytes and other epithelial cells, monocytes, tissue macrophages, fibroblasts, and endothelial cells.
  • IL-IRa cDNA cloned from human leukocytes contains an additional 63 bp sequence as an insert in the 5' region of the cDNA.
  • IL-IRa A 15 kDa isoform of IL-IRa, termed icIL-lRa3, is found in monocytes, macrophages, neutrophils, and hepatocytes, and may be created both by an alternative transcriptional splice as well as by alternative translational initiation.
  • IL-IRa peptides that are useful for fusion proteins of the invention include polypeptides that are at least 65%, at least 70%, at least 75%, at least 80%, at least 85%, at least 90%, or at least 95% identical to SEQ ID NO: 38.
  • the fusion proteins include an IL-IRa peptide sequence that is 85% identical to SEQ ID NO: 38 and has IL-IR binding activity, and preferably IL-IRa inhibitory activity.
  • the fusion proteins include an IL-IRa peptide sequence that is 95% identical to SEQ ID NO: 38 and has IL-IR binding activity, and preferably IL-IRa inhibitory activity.
  • polypeptides comprise Trpl6, Gln20, Tyr34, Gln36 and Tyrl47 according to the numbering of SEQ ID NO: 38
  • polypeptides may further include one or more amino acids substitutions D47N, E52R, E90Y, P38Y, H54R, Q129L and M136N (numbering according to SEQ ID NO: 38).
  • variations of the IL-IRa polypeptides can be accomplished by replacing one or more amino acids with another amino acid having similar structural or chemical properties, for example, conservative amino acid substitutions.
  • the fusion protein according to the invention is selected from an IL-I receptor antagonist selected from the following:
  • the trimeric IL-I Ra protein of the invention may be chemically synthesized or expressed in any suitable standard protein expression system.
  • the protein expression systems are systems from which the desired protein may readily be isolated and refolded in vitro.
  • Prokaryotic expression systems are preferred since high yields of protein can be obtained and efficient purification and refolding strategies are available.
  • Eukaryotic expression systems may also be used. Thus, it is well within the abilities and discretion of the skilled artisan to choose an appropriate expression system.
  • recombinant DNA constructs which will encode the desired proteins, taking into consideration such factors as codon biases in the chosen host, the need for secretion signal sequences in the host, the introduction of proteinase cleavage sites within the signal sequence, and the like.
  • These recombinant DNA constructs may be inserted in-frame into any of a number of expression vectors appropriate to the chosen host.
  • the expression vector will include a strong promoter to drive expression of the recombinant constructs.
  • the fusion protein of the invention can be expressed in any suitable standard protein expression system by culturing a host transformed with a vector encoding the fusion protein under such conditions that the fusion protein is expressed.
  • the expression system is a system from which the desired protein may readily be isolated and refolded in vitro.
  • prokaryotic expression systems are preferred since high yields of protein can be obtained and efficient purification and refolding strategies are available.
  • selection of appropriate expression systems is within the knowledge of one skilled in the art.
  • the isolated polynucleotide encodes a fusion protein of the invention.
  • an IL-IRa polypeptide and the trimerizing domain are encoded by non-contiguous polynucleotide sequences. Accordingly, in some embodiments an IL-IRa polypeptide and the trimerizing domain are expressed, isolated, and purified as separate polypeptides and fused together to form the fusion protein of the invention.
  • These recombinant DNA constructs may be inserted in-frame into any of a number of expression vectors appropriate to the chosen host.
  • the expression vector comprises a strong promoter that controls expression of the recombinant fusion protein constructs.
  • the resulting fusion protein can be isolated and purified using suitable standard procedures well known in the art, and optionally subjected to further processing such as e.g. lyophilization.
  • Standard techniques may be used for recombinant DNA molecule, protein, and fusion protein production, as well as for tissue culture and cell transformation. See, e.g., Sambrook, et al. (below) or Current Protocols in Molecular Biology (Ausubel et al. , eds., Green Publishers Inc. and Wiley and Sons 1994). Purification techniques are typically performed according to the manufacturer's specifications or as commonly accomplished in the art using conventional procedures such as those set forth in Sambrook et al. (Molecular Cloning: A Laboratory Manual. Cold Spring Harbor Laboratory Press, Cold Spring Harbor, NY (1989), or as described herein.
  • a flexible molecular linker optionally may be interposed between, and covalently join, the IL-IRa polypeptide and the trimerizing domain.
  • the linker is a polypeptide sequence of about 1 to 20 amino acid residues.
  • the linker may be less than 10 amino acids, most preferably, five, four, three, two, or one amino acid. It may be in certain cases that nine, eight, seven, or six amino acids are suitable.
  • the linker is essentially non-immunogenic, not prone to proteolytic cleavage and does not comprise amino acid residues which are known to interact with other residues (e.g. cysteine residues).
  • conjugates are covalently attached (hereinafter "conjugated") to one or more chemical groups.
  • Chemical groups suitable for use in such conjugates are preferably not significantly toxic or immunogenic.
  • the chemical group is optionally selected to produce a conjugate that can be stored and used under conditions suitable for storage.
  • a variety of exemplary chemical groups that can be conjugated to polypeptides are known in the art and include for example carbohydrates, such as those carbohydrates that occur naturally on glycoproteins, polyglutamate, and non-proteinaceous polymers, such as polyols (see, e.g., U.S. Pat. No. 6,245,901).
  • a polyol for example, can be conjugated to fusion proteins of the invention at one or more amino acid residues, including lysine residues, as is disclosed in WO 93/00109, supra.
  • the polyol employed can be any water-soluble poly(alkylene oxide) polymer and can have a linear or branched chain. Suitable polyols include those substituted at one or more hydroxyl positions with a chemical group, such as an alkyl group having between one and four carbons.
  • the polyol is a poly(alkylene glycol), such as poly(ethylene glycol) (PEG), and thus, for ease of description, the remainder of the discussion relates to an exemplary embodiment wherein the polyol employed is PEG and the process of conjugating the polyol to a polypeptide is termed "pegylation.”
  • PEG poly(ethylene glycol)
  • pegylation the process of conjugating the polyol to a polypeptide
  • other polyols such as, for example, poly(propylene glycol) and polyethylene- polypropylene glycol copolymers, can be employed using the techniques for conjugation described herein for PEG.
  • the average molecular weight of the PEG employed in the pegylation of IL-IRa can vary, and typically may range from about 500 to about 30,000 daltons (D).
  • the average molecular weight of the PEG is from about 1,000 to about 25,000 D, and more preferably from about 1,000 to about 5,000 D.
  • pegylation is carried out with PEG having an average molecular weight of about 1,000 D.
  • the PEG homopolymer is unsubstituted, but it may also be substituted at one end with an alkyl group.
  • the alkyl group is a C1-C4 alkyl group, and most preferably a methyl group.
  • PEG preparations are commercially available, and typically, those PEG preparations suitable for use in the present invention are non-homogeneous preparations sold according to average molecular weight.
  • commercially available PEG(5000) preparations typically contain molecules that vary slightly in molecular weight, usually ⁇ 500 D.
  • the fusion protein of the invention can be further modified using techniques known in the art, such as, conjugated to a small molecule compounds (e.g., a chemotherapeutic); conjugated to a signal molecule (e.g., a fluorophore); conjugated to a molecule of a specific binding pair (e.g,. biotin/streptavidin, antibody/antigen); or stabilized by glycosylation, PEGylation, or further fusions to a stabilizing domain (e.g., Fc domains).
  • a small molecule compounds e.g., a chemotherapeutic
  • a signal molecule e.g., a fluorophore
  • a variety of methods for pegylating proteins are known in the art. Specific methods of producing proteins conjugated to PEG include the methods described in U.S. Pat. Nos. 4,179,337, 4,935,465 and 5,849,535.
  • the protein is covalently bonded via one or more of the amino acid residues of the protein to a terminal reactive group on the polymer, depending mainly on the reaction conditions, the molecular weight of the polymer, etc.
  • the polymer with the reactive group(s) is designated herein as activated polymer.
  • the reactive group selectively reacts with free amino or other reactive groups on the protein.
  • the PEG polymer can be coupled to the amino or other reactive group on the protein in either a random or a site specific manner.
  • the type and amount of the reactive group chosen, as well as the type of polymer employed, to obtain optimum results will depend on the particular protein or protein variant employed to avoid having the reactive group react with too many particularly active groups on the protein. As this may not be possible to avoid completely, it is recommended that generally from about 0.1 to 1000 moles, preferably 2 to 200 moles, of activated polymer per mole of protein, depending on protein concentration, is employed. The final amount of activated polymer per mole of protein is a balance to maintain optimum activity, while at the same time optimizing, if possible, the circulatory half-life of the protein.
  • polyol when used herein refers broadly to polyhydric alcohol compounds.
  • Polyols can be any water-soluble poly(alkylene oxide) polymer for example, and can have a linear or branched chain.
  • Preferred polyols include those substituted at one or more hydroxyl positions with a chemical group, such as an alkyl group having between one and four carbons.
  • the polyol is a poly(alkylene glycol), preferably poly(ethylene glycol) (PEG).
  • PEG poly(ethylene glycol)
  • those skilled in the art recognize that other polyols, such as, for example, poly(propylene glycol) and polyethylene-polypropylene glycol copolymers, can be employed using the techniques for conjugation described herein for PEG.
  • polyols of the invention include those well known in the art and those publicly available, such as from commercially available sources.
  • other half-life extending molecules can be attached to the N-or C- terminus of the trimerization domain including serum albumin-binding peptides, FcRn- binding peptides or IgG-binding peptides.
  • the trimeric IL-I Ra protein of the invention is expressed in a prokaryotic host cell such as E.coli and is additionally linked to a third polypeptide, i.e. a third fusion partner.
  • a third polypeptide i.e. a third fusion partner.
  • the third fusion partner may be any suitable peptide, oligopeptide, polypeptide or protein, including a di-peptide, a tri-peptide, tetra-peptide, penta-peptide or hexa-peptide.
  • the fusion partner may in certain instances be a single amino acid. It may be selected such that it renders the fusion protein more resistant to proteolytic degradation, facilitates enhanced expression and secretion of the fusion protein, improves solubility, and/or allows for subsequent affinity purification of the fusion protein.
  • the junction region between the fusion protein of the invention i.e. the IL-IRa portion and the trimerization domain
  • the third fusion partner such as ubiquitin
  • a Granzyme B protease cleavage site such as human Granzyme B (E.C. 3.4.21.79) as described in US 2005/0199251.
  • the third fusion partner may in further embodiments be coupled to an affinity-tag.
  • an affinity-tag may be an affinity domain which allows for the purification of the fusion protein on an affinity resin.
  • the affinity-tag may be a polyhistidine-tag such as a hexabis-tag, polyarginine-tag, FLAG-tag, Strep-tag, c-myc-tag, S-tag, calmodulin-binding peptide, cellulose-binding peptide, chitin-binding domain, glutathione S-transferase-tag, or maltose binding protein.
  • the method of the invention may be in an isolation step for isolating the trimeric IL-I Ra protein that is formed by the enzymatic cleavage of the fusion protein that has been immobilized by the use of the above mentioned affinity-tag systems.
  • This isolation step can be performed by any suitable means known in the art for protein isolation, including the use of ion exchange and fractionation by size, the choice of which depends on the character of the fusion protein.
  • the region between the third fusion partner and the region comprising the trimerization domain and IL-I Ra is contacted with the human serine protease Granzyme B to cleave off the fusion protein at a Granzyme B protease cleavage site which yields the fusion protein of the invention.
  • the present invention also provides plasmids, vectors, transcription or expression cassettes which comprise at least one nucleic acid as described above.
  • Suitable vectors can be chosen or constructed containing appropriate regulatory sequences, including promoter sequences, terminator sequences, polyadenylation sequences, enhancer sequences, marker genes and other sequences as appropriate.
  • Vectors may be plasmids, viral, phage, or phagemid, as appropriate. (Molecular Cloning: a Laboratory Manual: 2nd edition, Sambrook et ah, 1989, Cold Spring Harbor Laboratory Press).
  • the present invention also provides a recombinant host cell which comprises one or more constructs of the invention.
  • Suitable host cells include bacteria, mammalian cells, yeast and baculovirus systems.
  • Mammalian cell lines available for expression of a heterologous polypeptide include Chinese hamster ovary cells, HeLa cells, baby hamster kidney cells, NSO mouse melanoma cells and many others.
  • a preferred bacterial host is E. coli.
  • the invention relates to a pharmaceutical composition
  • a pharmaceutical composition comprising a therapeutically effective amount of the fusion protein of the invention along with a pharmaceutically acceptable carrier or excipient.
  • pharmaceutically acceptable carrier or “pharmaceutically acceptable excipient” includes any and all solvents, dispersion media, coating, antibacterial and antifungal agents, isotonic and absorption delaying agents, and the like that are physiologically compatible.
  • pharmaceutically acceptable carriers or excipients include one or more of water, saline, phosphate buffered saline, dextrose, glycerol, ethanol and the like as well as combinations thereof.
  • isotonic agents for example, sugars, polyalcohols such as mannitol, sorbitol, or sodium chloride in the composition.
  • Pharmaceutically acceptable substances such as wetting or minor amounts of auxiliary substances such as wetting or emulsifying agents, preservatives or buffers, which enhance the shelf life or effectiveness of the of the antibody or antibody portion also may be included.
  • disintegrating agents can be included, such as cross-linked polyvinyl pyrrolidone, agar, alginic acid or a salt thereof, such as sodium alginate and the like.
  • the pharmaceutical composition can include one or more of the following, carrier proteins such as serum albumin, buffers, binding agents, sweeteners and other flavoring agents; coloring agents and polyethylene glycol.
  • compositions can be in a variety of forms including, for example, liquid, semi-solid and solid dosage forms, such as liquid solutions (e.g. injectable and infusible solutions), dispersions or suspensions, tablets, pills, powders, liposomes and suppositories.
  • liquid solutions e.g. injectable and infusible solutions
  • dispersions or suspensions tablets, pills, powders, liposomes and suppositories.
  • the preferred form will depend on the intended route of administration and therapeutic application.
  • the compositions are in the form of injectable or infusible solutions, such as compositions similar to those used for passive immunization of humans with antibodies.
  • the mode of administration is parenteral (e.g., intravenous, subcutaneous, intraperitoneal, intramuscular).
  • the fusion protein (or trimeric complex) is administered by intravenous infusion or injection.
  • the fusion protein or trimeric complex is administered by intramuscular or subcutaneous injection.
  • compositions include, but are not limited to, oral, rectal, transdermal, vaginal, transmucosal or intestinal administration.
  • compositions are typically sterile and stable under the conditions of manufacture and storage.
  • the composition can be formulated as a solution, microemulsion, dispersion, liposome, or other ordered structure suitable to high drug concentration.
  • Sterile injectable solutions can be prepared by incorporating the active compound (i.e. fusion protein or trimeric complex) in the required amount in an appropriate solvent with one or a combination of ingredients enumerated above, as required, followed by filtered sterilization.
  • dispersions are prepared by incorporating the active compound into a sterile vehicle that contains a basic dispersion medium and the required other ingredients from those enumerated above.
  • the preferred methods of preparation are vacuum drying and freeze-drying that yields a powder of the active ingredient plus any additional desired ingredient from a previously sterile-filtered solution thereof.
  • the proper fluidity of a solution can be maintained, for example, by the use of a coating such as lecithin, by the maintenance of the required particle size in the case of dispersion and by the use of surfactants.
  • Prolonged absorption of injectable compositions can be brought about by including in the composition an agent that delays absorption, for example, monostearate salts and gelatin.
  • An article of manufacture such as a kit containing therapeutic agents useful in the treatment of the disorders described herein comprises at least a container and a label.
  • Suitable containers include, for example, bottles, vials, syringes, and test tubes.
  • the containers may be formed from a variety of materials such as glass or plastic.
  • the label on or associated with the container indicates that the formulation is used for treating the condition of choice.
  • the article of manufacture may further comprise a container comprising a pharmaceutically- acceptable buffer, such as phosphate-buffered saline, Ringer's solution, and dextrose solution.
  • the article of manufacture may also comprise a container with another active agent as described above.
  • an appropriate amount of a pharmaceutically-acceptable salt is used in the formulation to render the formulation isotonic.
  • pharmaceutically-acceptable carriers include saline, Ringer's solution and dextrose solution.
  • the pH of the formulation is preferably from about 6 to about 9, and more preferably from about 7 to about 7.5. It will be apparent to those persons skilled in the art that certain carriers may be more preferable depending upon, for instance, the route of administration and concentrations of therapeutic agent.
  • compositions can be prepared by mixing the desired molecules having the appropriate degree of purity with optional pharmaceutically acceptable carriers, excipients, or stabilizers (Remington's Pharmaceutical Sciences, 16th edition, Osol, A. ed. (1980)), in the form of lyophilized formulations, aqueous solutions or aqueous suspensions.
  • Acceptable carriers, excipients, or stabilizers are preferably nontoxic to recipients at the dosages and concentrations employed, and include buffers such as Tris, HEPES, PIPES, phosphate, citrate, and other organic acids; antioxidants including ascorbic acid and methionine; preservatives (such as octadecyldimethylbenzyl ammonium chloride; hexamethonium chloride; benzalkonium chloride, benzethonium chloride; phenol, butyl or benzyl alcohol; alkyl parabens such as methyl or propyl paraben; catechol; resorcinol; cyclohexanol; 3-pentanol; and m-cresol); low molecular weight (less than about 10 residues) polypeptides; proteins, such as serum albumin, gelatin, or immunoglobulins; hydrophilic polymers such as polyvinylpyrrolidone; amino acids such as glycine,
  • Such carriers include ion exchangers, alumina, aluminum stearate, lecithin, serum proteins, such as human serum albumin, buffer substances such as glycine, sorbic acid, potassium sorbate, partial glyceride mixtures of saturated vegetable fatty acids, water, salts, or electrolytes such as protamine sulfate, disodium hydrogen phosphate, potassium hydrogen phosphate, sodium chloride, colloidal silica, magnesium trisilicate, polyvinyl pyrrolidone, and cellulose-based substances.
  • ion exchangers alumina, aluminum stearate, lecithin
  • serum proteins such as human serum albumin
  • buffer substances such as glycine, sorbic acid, potassium sorbate, partial glyceride mixtures of saturated vegetable fatty acids, water, salts, or electrolytes
  • protamine sulfate disodium hydrogen phosphate
  • potassium hydrogen phosphate sodium chloride
  • colloidal silica magnesium trisilicate
  • Carriers for topical or gel-based forms include polysaccharides such as sodium carboxymethylcellulose or methylcellulose, polyvinylpyrrolidone, polyacrylates, polyoxyethylene-polyoxypropylene-block polymers, polyethylene glycol, and wood wax alcohols.
  • conventional depot forms are suitably used.
  • Such forms include, for example, microcapsules, nano-capsules, liposomes, plasters, inhalation forms, nose sprays, sublingual tablets, and sustained-release preparations.
  • Formulations to be used for in vivo administration should be sterile. This is readily accomplished by filtration through sterile filtration membranes, prior to or following lyophilization and reconstitution.
  • the formulation may be stored in lyophilized form or in solution if administered systemically. If in lyophilized form, it is typically formulated in combination with other ingredients for reconstitution with an appropriate diluent at the time for use.
  • An example of a liquid formulation is a sterile, clear, colorless unpreserved solution filled in a single-dose vial for subcutaneous injection.
  • Therapeutic formulations generally are placed into a container having a sterile access port, for example, an intravenous solution bag or vial having a stopper pierceable by a hypodermic injection needle.
  • the formulations are preferably administered as repeated intravenous (i.v.), subcutaneous (s.c), intramuscular (i.m.) injections or infusions, or as aerosol formulations suitable for intranasal or intrapulmonary delivery (for intrapulmonary delivery see, e.g., EP 257,956).
  • sustained-release preparations include semipermeable matrices of solid hydrophobic polymers containing the protein, which matrices are in the form of shaped articles, e.g., films, or microcapsules.
  • sustained-release matrices include polyesters, hydrogels (e.g., poly(2-hydroxyethyl- methacrylate) as described by Langer et al., J. Biomed. Mater. Res., 15: 167-277 (1981) and Langer, Chem. Tech., 12: 98-105 (1982) or poly(vinylalcohol)), polylactides (U.S. Pat. No.
  • Another aspect the invention relates to a method of treating diseases that are mediated by IL-IRa.
  • the method includes treating a subject suffering from such as disease with a therapeutically effective amount of the pharmaceutical compositions of the invention.
  • formulations comprising therapeutic agents are also provided by the present invention. It is believed that such formulations will be particularly suitable for storage as well as for therapeutic administration.
  • the formulations may be prepared by known techniques. For instance, the formulations may be prepared by buffer exchange on a gel filtration column.
  • compositions can be administered in accord with known methods, such as intravenous administration as a bolus or by continuous infusion over a period of time, by intramuscular, intraperitoneal, intracerobrospinal, subcutaneous, intraarticular, intrasynovial, intrathecal, oral, topical, or inhalation routes.
  • administration may be performed through mini-pump infusion using various commercially available devices.
  • Effective dosages and schedules for administering the trimeric IL-IRa may be determined empirically, and making such determinations is within the skill in the art. Single or multiple dosages may be employed.
  • an effective dosage or amount of the trimeric IL-IRa used alone may range from about 1 ⁇ g/kg to about 100 mg/kg of body weight or more per day.
  • Interspecies scaling of dosages can be performed in a manner known in the art, e.g., as disclosed in Mordenti, et al, Pharmaceut. Res., 8:1351 (1991).
  • normal dosage amounts may vary from about 10 ng/kg to up to 100 mg/kg of mammal body weight or more per day, preferably about 1 ⁇ g/kg/day to 50 mg/kg/day, depending upon the route of administration.
  • Guidance as to particular dosages and methods of delivery is provided in the literature (see, for example, U.S. Pat. Nos. 4,657,760; 5,206,344; or 5,225,212).
  • U.S. Pat. Nos. 4,657,760; 5,206,344; or 5,225,212 See, for example, U.S. Pat. Nos. 4,657,760; 5,206,344; or 5,225,212).
  • administration targeting one organ or tissue for example, may necessitate delivery in a manner different from that to another organ or tissue.
  • the dosage of the trimeric IL-IRa that must be administered will vary depending on, for example, the mammal which will receive trimeric IL-IRa, the route of administration, and other drugs or therapies being administered to the mammal.
  • the trimeric complexes and other therapeutic agents may be administered concurrently (simultaneously) or sequentially.
  • a fusion protein or trimeric complex and a therapeutic agent are administered concurrently.
  • a fusion protein or trimeric complex is administered prior to administration of a therapeutic agent.
  • a therapeutic agent is administered prior to a fusion protein or trimeric complex.
  • treated cells in vitro can be analyzed. Where there has been in vivo treatment, a treated mammal can be monitored in various ways well known to the skilled practitioner. For instance, serum cytokine responses can be analyzed.
  • the IL-IRa fusion proteins described herein may be used in combination (pre- treatment, post-treatment, or concurrent treatment) with any of one or more TNF inhibitors for the treatment or prevention of the diseases and disorders recited herein, such as but not limited to, all forms of soluble TNF receptors including Etanercept (such as ENBREL ), as well as all forms of monomelic or multimeric p75 and/or p55 TNF receptor molecules and fragments thereof; anti-human TNF antibodies, such as but not limited to, Infliximab (such as REMICADE ® ), and D2E7 (such as HUMIRA ® ), and the like.
  • TNF inhibitors for the treatment or prevention of the diseases and disorders recited herein, such as but not limited to, all forms of soluble TNF receptors including Etanercept (such as ENBREL ), as well as all forms of monomelic or multimeric p75 and/or p55 TNF receptor molecules and fragments thereof; anti-human TNF antibodies, such as
  • TNF inhibitors include compounds and proteins which block in vivo synthesis or extracellular release of TNF.
  • the present invention is directed to the use of an IL- 17RA IL-IRa fusion proteins in combination (pre-treatment, post-treatment, or concurrent treatment) with any of one or more of the following TNF inhibitors: TNF binding proteins (soluble TNF receptor type-I and soluble TNF receptor type-II ("sTNFRs"), as defined herein), anti-TNF antibodies, granulocyte colony stimulating factor; thalidomide; BN 50730; tenidap; E 5531; tiapafant PCA 4248; nimesulide; panavir; rolipram; RP 73401; peptide T; MDL 201,449A; (lR,3S)-Cis-l-[9-(2,6-diaminopurinyl)]-3-hydroxy-4-cyclopentene hydrochloride; (1R,3R)- trans
  • TNF binding proteins are disclosed in the art (EP 308 378, EP 422 339, GB 2 218 101, EP 393 438, WO 90/13575, EP 398 327, EP 412 486, WO 91/03553, EP 418 014, JP 127,800/1991, EP 433 900, U.S. Pat. No. 5,136,021, GB 2 246 569, EP 464 533, WO 92/01002, WO 92/13095, WO 92/16221, EP 512 528, EP 526 905, WO 93/07863, EP 568 928, WO 93/21946, WO 93/19777, EP 417 563, WO 94/06476, and PCT International Application No. PCT/US97/12244).
  • EP 393 438 and EP 422 339 teach the amino acid and nucleic acid sequences of a soluble TNF receptor type I (also known as “sTNFR-I” or “30 kDa TNF inhibitor”) and a soluble TNF receptor type II (also known as “sTNFR-II” or “40 kDa TNF inhibitor”), collectively termed "sTNFRs", as well as modified forms thereof (e.g., fragments, functional derivatives and variants).
  • sTNFRs also disclose methods for isolating the genes responsible for coding the inhibitors, cloning the gene in suitable vectors and cell types and expressing the gene to produce the inhibitors.
  • polyvalent forms i.e., molecules comprising more than one active moiety
  • the polyvalent form may be constructed by chemically coupling at least one TNF inhibitor and another moiety with any clinically acceptable linker, for example polyethylene glycol (WO 92/16221 and WO 95/34326), by a peptide linker (Neve et al. (1996), Cytokine, 8(5):365-370, by chemically coupling to biotin and then binding to avidin (WO 91/03553) and, finally, by combining chimeric antibody molecules (U.S. Pat. No. 5,116,964, WO 89/09622, WO 91/16437 and EP 315062.
  • Anti-TNF antibodies include the MAK 195F Fab antibody (Holler et al. (1993), 1st International Symposium on Cytokines in Bone Marrow Transplantation, 147); CDP 571 anti-TNF monoclonal antibody (Rankin et al. (1995), British Journal of Rheumatology, 34:334-342); BAY X 1351 murine anti-tumor necrosis factor monoclonal antibody (Kieft et al. (1995), 7th European Congress of Clinical Microbiology and Infectious Diseases, page 9); CenTNF cA2 anti-TNF monoclonal antibody (Elliott et al. (1994), Lancet, 344:1125-1127 and Elliott et al (1994), Lancet, 344: 1105-1110).
  • IL-IRa fusion proteins described herein may be used in combination with all forms of IL-17 inhibitors (e.g. anti-IL17 receptor antibody, Amgen; anti-IL-17A, anti- ILl 7F), RORc inhibitors.
  • IL-17 inhibitors e.g. anti-IL17 receptor antibody, Amgen; anti-IL-17A, anti- ILl 7F, RORc inhibitors.
  • IL-IRa fusion proteins described herein may be used in combination with all forms of CD28 inhibitors, such as but not limited to, abatacept (for example ORENCIA ® ).
  • IL-IRa fusion proteins described herein may be used in combination with all forms of IL-6 and/or IL-6 receptor inhibitors, such as but not limited to, Tocilizumab (for example ACTEMRA ® ).
  • IL-IRa fusion proteins described herein may be used in combination with all forms of anti-IL-18 compounds, such as IL-18BP or a derivative, an IL-18 trap, anti-IL-18, anti-IL- 18Rl , or anti-IL- 18RAcP.
  • IL-IRa fusion proteins described herein may be used in combination with all forms of anti-IL22, such as anti-IL22 or anti-IL22R.
  • IL-IRa fusion proteins described herein may be used in combination with all forms of anti-IL-23 and or IL- 12 such as anti-pl9, anti-p40 (Ustekinumab), anti-IL-23R.
  • the IL-IRa fusion proteins described herein may be used in combination with all forms of anti-IL21, such as anti-IL21 or anti-IL21R. [0094] The IL-IRa fusion proteins described herein may be used in combination with all forms of anti-TGF-beta.
  • the IL-IRa fusion proteins may be used in combination with one or more cytokines, lymphokines, hematopoietic factor(s), and/or an anti-inflammatory agent.
  • Treatment of the diseases and disorders recited herein can include the use of first line drags for control of pain and inflammation in combination (pretreatment, post-treatment, or concurrent treatment) with treatment with one or more of the IL-IRa fusion proteins provided herein.
  • These drugs are classified as non-steroidal, anti-inflammatory drugs (NSAIDs).
  • Secondary treatments include corticosteroids, slow acting antirheumatic drags (SAARDs), or disease modifying (DM) drags.
  • SAARDs slow acting antirheumatic drags
  • DM disease modifying
  • NSAIDs owe their anti-inflammatory action, at least in part, to the inhibition of prostaglandin synthesis (Goodman and Gilman in "The Pharmacological Basis of Therapeutics," MacMillan 7th Edition (1985)).
  • NSAIDs can be characterized into at least nine groups: (1) salicylic acid derivatives; (2) propionic acid derivatives; (3) acetic acid derivatives; (4) fenamic acid derivatives; (5) carboxylic acid derivatives; (6) butyric acid derivatives; (7) oxicams; (8) pyrazoles and (9) pyrazolones.
  • the IL-IRa fusion proteins described herein may be used in combination (pretreatment, post-treatment, or concurrent treatment) with any of one or more salicylic acid derivatives, prodrug esters or pharmaceutically acceptable salts thereof.
  • salicylic acid derivatives, prodrug esters and pharmaceutically acceptable salts thereof comprise: acetaminosalol, aloxiprin, aspirin, benorylate, bromosaligenin, calcium acetylsalicylate, choline magnesium trisalicylate, magnesium salicylate, choline salicylate, diflusinal, etersalate, fendosal, gentisic acid, glycol salicylate, imidazole salicylate, lysine acetylsalicylate, mesalamine, morpholine salicylate, 1-naphthyl salicylate, olsalazine, parsalmide, phenyl acetylsalicylate, phenyl salicyl
  • the present invention is directed to the use of an IL-IRa fusion proteins in combination (pretreatment, post-treatment, or concurrent treatment) with any of one or more propionic acid derivatives, prodrug esters or pharmaceutically acceptable salts thereof.
  • the propionic acid derivatives, prodrug esters, and pharmaceutically acceptable salts thereof comprise: alminoprofen, benoxaprofen, bucloxic acid, carprofen, dexindoprofen, fenoprofen, flunoxaprofen, fluprofen, flurbiprofen, furcloprofen, ibuprofen, ibuprofen aluminum, ibuproxam, indoprofen, isoprofen, ketoprofen, loxoprofen, miroprofen, naproxen, naproxen sodium, oxaprozin, piketoprofen, pimeprofen, pirprofen, pranoprofen, protizinic acid, pyridoxiprofen, suprofen, tiaprofenic acid and tioxaprofen. Structurally related propionic acid derivatives having similar analgesic and antiinflammatory properties are also intended to be encompassed by
  • the present invention is directed to the use of an IL-IRa fusion proteins in combination (pretreatment, post-treatment, or concurrent treatment) with any of one or more acetic acid derivatives, prodrug esters or pharmaceutically acceptable salts thereof.
  • acetic acid derivatives, prodrug esters, and pharmaceutically acceptable salts thereof comprise: acemetacin, alclofenac, amfenac, bufexamac, cinmetacin, clopirac, delmetacin, diclofenac potassium, diclofenac sodium, etodolac, felbinac, fenclofenac, fenclorac, fenclozic acid, fentiazac, furofenac, glucametacin, ibufenac, indomethacin, isofezolac, isoxepac, lonazolac, metiazinic acid, oxametacin, oxpinac, pimetacin, proglumetacin, sulindac, talmetacin, tiaramide, tiopinac, tolmetin, tolmetin sodium, zidometacin and zomepirac.
  • the present invention is directed to the use of an IL-IRa fusion proteins in combination (pretreatment, post-treatment, or concurrent treatment) with any of one or more fenamic acid derivatives, prodrug esters or pharmaceutically acceptable salts thereof.
  • the fenamic acid derivatives, prodrug esters and pharmaceutically acceptable salts thereof comprise: enfenamic acid, etofenamate, flufenamic acid, isonixin, meclofenamic acid, meclofenamate sodium, medofenamic acid, mefenamic acid, niflumic acid, talniflumate, terofenamate, tolfenamic acid and ufenamate.
  • Stracturally related fenamic acid derivatives having similar analgesic and anti-inflammatory properties are also intended to be encompassed by this group.
  • the present invention is directed to the use of an IL-IRa fusion proteins in combination (pretreatment, post-treatment, or concurrent treatment) with any of one or more carboxylic acid derivatives, prodrug esters or pharmaceutically acceptable salts thereof.
  • carboxylic acid derivatives, prodrug esters, and pharmaceutically acceptable salts thereof which can be used comprise: clidanac, diflunisal, flufenisal, inoridine, ketorolac and tinoridine.
  • Stracturally related carboxylic acid derivatives having similar analgesic and anti-inflammatory properties are also intended to be encompassed by this group.
  • the present invention is directed to the use of an IL-IRa fusion proteins in combination (pretreatment, post-treatment, or concurrent treatment) with any of one or more butyric acid derivatives, prodrug esters or pharmaceutically acceptable salts thereof.
  • the butyric acid derivatives, prodrug esters, and pharmaceutically acceptable salts thereof comprise: bumadizon, butibufen, fenbufen and xenbucin. Structurally related butyric acid derivatives having similar analgesic and antiinflammatory properties are also intended to be encompassed by this group.
  • the present invention is directed to the use of an IL-IRa fusion proteins in combination (pretreatment, post-treatment, or concurrent treatment) with any of one or more oxicams, prodrug esters, or pharmaceutically acceptable salts thereof.
  • the oxicams, prodrug esters, and pharmaceutically acceptable salts thereof comprise: droxicam, enolicam, isoxicam, piroxicam, sudoxicam, tenoxicam and 4-hydroxyl-l,2- benzotbiazine 1,1 -dioxide 4-(N-phenyl)-carboxamide.
  • Structurally related oxicams having similar analgesic and anti-inflammatory properties are also intended to be encompassed by this group.
  • the present invention is directed to the use of an IL-IRa fusion proteins in combination (pretreatment, post-treatment, or concurrent treatment) with any of one or more pyrazoles, prodrug esters, or pharmaceutically acceptable salts thereof.
  • the pyrazoles, prodrug esters, and pharmaceutically acceptable salts thereof which may be used comprise: difenamizole and epirizole. Structurally related pyrazoles having similar analgesic and anti-inflammatory properties are also intended to be encompassed by this group.
  • the present invention is directed to the use of an IL-IRa fusion proteins in combination (pretreatment, post-treatment or, concurrent treatment) with any of one or more pyrazolones, prodrug esters, or pharmaceutically acceptable salts thereof.
  • the pyrazolones, prodrug esters and pharmaceutically acceptable salts thereof which may be used comprise: apazone, azapropazone, benzpiperylon, feprazone, mofebutazone, morazone, oxyphenbutazone, phenylbutazone, pipebuzone, propylphenazone, ramifenazone, suxibuzone and thiazolinobutazone.
  • Structurally related pyrazalones having similar analgesic and anti-inflammatory properties are also intended to be encompassed by this group.
  • the present invention is directed to the use of an IL-IRa fusion proteins in combination (pretreatment, post-treatment, or concurrent treatment) with any of one or more of the following NSAIDs: .epsilon.-acetamidocaproic acid, S- adenosyl-methionine, 3-amino-4-hydroxybutyric acid, amixetrine, anitrazafen, antrafenine, bendazac, bendazac lysinate, benzydamine, beprozin, broperamole, bucolome, bufezolac, ciproquazone, cloximate, dazidamine, deboxamet, detomidine, difenpiramide, difenpyramide, difisalamine, ditazol, emorfazone, fanetizole mesylate, fenflumizole, floctafenine, flumizole, flunixin, fluproqua
  • NSAIDs .
  • the present invention is directed to the use of an IL-IRa fusion proteins in combination (pretreatment, post-treatment or concurrent treatment) with any of one or more corticosteroids, prodrug esters or pharmaceutically acceptable salts thereof for the treatment of the diseases and disorders recited herein, including acute and chronic inflammation such as rheumatic diseases, graft versus host disease and multiple sclerosis.
  • Corticosteroids, prodrug esters and pharmaceutically acceptable salts thereof include hydrocortisone and compounds which are derived from hydrocortisone, such as 21-acetoxypregnenolone, alclomerasone, algestone, amcinonide, beclomethasone, betamethasone, betamethasone valerate, budesonide, chloroprednisone, clobetasol, clobetasol propionate, clobetasone, clobetasone butyrate, clocortolone, cloprednol, corticosterone, cortisone, cortivazol, deflazacon, desonide, desoximerasone, dexamethasone, diflorasone, diflucortolone, difluprednate, enoxolone, fluazacort, flucloronide, flumethasone, flumethasone pivalate, flucinolone acetonide, flu
  • the present invention is directed to the use of an IL-IRa fusion proteins in combination (pretreatment, post-treatment, or concurrent treatment) with any of one or more slow-acting antirheumatic drugs (SAARDs) or disease modifying antirheumatic drags (DMARDS), prodrug esters, or pharmaceutically acceptable salts thereof for the treatment of the diseases and disorders recited herein, including acute and chronic inflammation such as rheumatic diseases, graft versus host disease and multiple sclerosis.
  • SAARDs slow-acting antirheumatic drugs
  • DARDS disease modifying antirheumatic drags
  • prodrug esters or pharmaceutically acceptable salts thereof for the treatment of the diseases and disorders recited herein, including acute and chronic inflammation such as rheumatic diseases, graft versus host disease and multiple sclerosis.
  • SAARDs or DMARDS, prodrug esters and pharmaceutically acceptable salts thereof comprise: allocupreide sodium, auranof ⁇ n, aurothioglucose, aurothioglycanide, azathioprine, brequinar sodium, bucillamine, calcium 3-aurothio-2-propanol-l-sulfonate, chlorambucil, chloroquine, clobuzarit, cuproxoline, cyclo-phosphamide, cyclosporin, dapsone, 15- deoxyspergualin, diacerein, glucosamine, gold salts (e.g., cycloquine gold salt, gold sodium thiomalate, gold sodium thiosulfate), hydroxychloroquine, hydroxychloroquine sulfate, hydroxyurea, kebuzone, levamisole, lobenzarit, melittin, 6-mercaptopurine, methotrexate, mi
  • the present invention is directed to the use of an IL-IRa fusion proteins in combination (pretreatment, post-treatment, or concurrent treatment) with any of one or more COX2 inhibitors, prodrug esters or pharmaceutically acceptable salts thereof for the treatment of the diseases and disorders recited herein, including acute and chronic inflammation.
  • COX2 inhibitors, prodrug esters or pharmaceutically acceptable salts thereof include, for example, celecoxib.
  • Structurally related COX2 inhibitors having similar analgesic and anti-inflammatory properties are also intended to be encompassed by this group.
  • Examples of COX-2 selective inhibitors include but not limited to etoricoxib, valdecoxib, celecoxib, licofelone, lumiracoxib, rofecoxib, and the like.
  • the present invention is directed to the use of an IL-IRa fusion proteins in combination (pretreatment, post-treatment, or concurrent treatment) with any of one or more antimicrobials, prodrug esters or pharmaceutically acceptable salts thereof for the treatment of the diseases and disorders recited herein, including acute and chronic inflammation.
  • Antimicrobials include, for example, the broad classes of penicillins, cephalosporins and other beta-lactams, aminoglycosides, azoles, quinolones, macrolides, rifamycins, tetracyclines, sulfonamides, lincosamides and polymyxins.
  • the penicillins include, but are not limited to penicillin G, penicillin V, methicillin, nafcillin, oxacillin, cloxacillin, dicloxacillin, floxacillin, ampicillin, ampicillin/sulbactam, amoxicillin, amoxicillin/clavulanate, hetacillin, cyclacillin, bacampicillin, carbenicillin, carbenicillin indanyl, ticarcillin, ticarcillin/clavulanate, azlocillin, meziocillin, peperacillin, and mecillinam.
  • cephalosporins and other beta- lactams include, but are not limited to cephalothin, cephapirin, cephalexin, cephradine, cefazolin, cefadroxil, cefaclor, cefamandole, cefotetan, cefoxitin, ceruroxime, cefonicid, ceforadine, cefixime, cefotaxime, moxalactam, ceftizoxime, cetriaxone, cephoperazone, ceftazidime, imipenem and aztreonam.
  • the aminoglycosides include, but are not limited to streptomycin, gentamicin, tobramycin, amikacin, netilmicin, kanamycin and neomycin.
  • the azoles include, but are not limited to fluconazole.
  • the quinolones include, but are not limited to nalidixic acid, norfloxacin, enoxacin, ciprofloxacin, ofloxacin, sparfloxacin and temafloxacin.
  • the macrolides include, but are not limited to erythomycin, spiramycin and azithromycin.
  • the rifamycins include, but are not limited to rifampin.
  • the tetracyclines include, but are not limited to spicycline, chlortetracycline, clomocycline, demeclocycline, deoxycycline, guamecycline, lymecycline, meclocycline, methacycline, minocycline, oxytetracycline, penimepicycline, pipacycline, rolitetracycline, sancycline, senociclin and tetracycline.
  • the sulfonamides include, but are not limited to sulfanilamide, sulfamethoxazole, sulfacetamide, sulfadiazine, sulf ⁇ soxazole and co-trimoxazole (trimethoprim/sulfamethoxazole).
  • the lincosamides include, but are not limited to clindamycin and lincomycin.
  • the polymyxins (polypeptides) include, but are not limited to polymyxin B and colistin.
  • Example 1 Format, production and purification of trimeric IL-IRa
  • IL-IRa can be produced as recombinant protein in E. coli. (Steinkasserer et al 1992. FEBS 310:63-65). The protein is very stable and refolds efficiently. Isoforms of IL-IRa with additional amino acids in the N-terminal have been also described (Haskill et al 1991, PNAS 88:3681-3685; Muzio et al 1995, JEM 182, 623-628)). These molecules bind IL-IR as well as the mature secreted form indicating that it is possible to fuse extra peptide to the N-terminal of the antagonist without compromising the binding to the receptor.
  • IL-IRa Crystal structure analysis of IL-IRa interaction with IL-IR also supports that N-terminal alterations do not affect interactions with ILlR (Schreuder et al 1997, Nature 386: 190-194).
  • IL-IRa was cloned from a human cDNA library derived from bone marrow and/or human placenta.
  • Trimeric IL-IRa was designed as a C-terminal fusion to the Trip-trimerization unit. Eight different fusion proteins were designed, four with full length trimerization units (Trip) and four with a nine amino acid truncation of the trimerization unit (IlOTrip). IL- Ira was than fused with either trimerization unit using four different C-terminal fusions. C- terminal variations termed Trip V, Trip T, Trip Q and Trip K allow for unique presentation of the CTLD domains on the trimerization domain. The Trip K variant is the longest construct and contains the longest and most flexible linker between the CTLD and the trimerization domain.
  • Trip V, Trip T, Trip Q represent fusions of the CTLD molecule directly onto the trimerization module without any structural flexibility but are turning the CTLD molecule l/3 r going from Trip V to Trip T and from Trip T to Trip Q. This is due to the fact that each of these amino acids is in an ⁇ -helical turn and 3.2 aa are needed for a full turn
  • NMPDEGVMVTKFYFQEDE (SEQ ID NO: 49);
  • the sample was then sonicated for 2-5 min with mixing in between.
  • Detergent buffer 0.2 M NaCl, 1 w/v % Deoxycholate, Na salt, 1 w/v % Nonidet P40, 20 mM Tris-HCl, pH 7.5, 2 mM EDTA
  • the inclusion bodies were recovered by centrifugation for 25 min at 8.000 rpm, 4°C.
  • the supernatant was stored at 4 0 C and the pellet resuspended in 100 mL TRITON ® X-IOO buffer (0,5 w/v % TRITON ® X-100, 1 mM EDTA , pH 8) per 50 g original cell pellet.
  • Inclusion bodies were recovered by centrifugation for 25 min at 8.000 rpm, 4°C and the supernatant was stored at 4°C.
  • the TRITON ® X-100 buffer wash is repeated once more and the inclusion bodies were recovered by centrifugation for 5 min at 12.000 rpm, 4°C.
  • the inclusion bodies were re-suspended in 30 mL denaturing buffer/gram original cell paste (6 M urea, 10 mM EDTA, 20 mM Tris/HCl and 20 mM ⁇ -Mercaptoethanol, pH 8.0) at 28 0 C for 2 h.
  • the suspension was centrifuged at 7500 g for 15 min to remove insoluble material.
  • the cleaved protein was purified using SP-Sepharose FF (GE Healthcare) cation exchange step.
  • a ⁇ 50 mL SP-Sepharose FF was packed and equilibrated in buffer A (I x PBS, 1 mM EDTA pH 5,5) until stabile basis line was obtained.
  • the cleavage reaction was diluted 1:3 with buffer A and loaded on the column followed by a wash in buffer A until stabile basis line was monitored.
  • a gradient from 10 CV buffer A to 10 CV Buffer B (1 x PBS 5 1 mM EDTA + 0,5 M NaCl pH 5,5) was setup and fractions collected in 5 mL. Protein containing fractions were analysed on SDS-PAGE before pooling the protein product.
  • the supernatant from the above inclusion body preparation were used to purify the protein.
  • the soluble Trimeric ILl-ra in the supernatant was purified on Ni- NTA Superflow (Qiagen) column equilibrated in Buffer A (2OmM TrisHCL, 5OmM NaCl pH 8.0.
  • Buffer A 2OmM TrisHCL, 5OmM NaCl pH 8.0.
  • a pool was made of the washes from the inclusion body purification and it was centrifuged at 10000 rpm for lOmin before CaCl 2 was added to 5 mM and Tris-HCl to 20 mM and the pH adjusted to 6.0 with HCl/NaOH. The pool was loaded on the column and washed in buffer A until stabile basis line.
  • Example 2 Trimeric IL-IRa compounds ability to inhibit IL-I induction of IL-8 in U937 cells
  • GG-TripV-IL-lra trip V-ILlRa
  • GG-TripK-IL-lra trip K-ILlRa
  • GG-TripT- IL-lra trip T-ILlRa
  • CII-H6-GrB-GG-TripT-IL-lra trip Q-ILlRa
  • the compounds are essentially equally effective in blocking the response and they appear all to be as effective as KINERET ® (when compared on w/w). Due to buffer effects in the assay, at the highest protein concentration used (100 ⁇ g/mL) IL-8 production increases instead of further decreasing. Based on several in vitro efficacy assays as well as Biacore assays, it was determined that TripT ILlRa was the best compound based on blocking and binding efficacy as well as production yields.
  • Example 3 Pegylated trimeric IL-IRa compounds
  • KINERET ® Since the in vivo half life is a crucial parameter in the efficacy of KINERET (KINERET ® has only a half life in humans of 4-6 hours and has therefore, to be applied once daily) the ability to pegylate the TripT ILlRa by N-terminal pegylation was tested.
  • the trimeric ILl-Ra is pegylated at the N terminus.
  • Tr ⁇ neric ILl-Ra antagonist proteins after the final step of the purification procedure described above were used as starting point for pegylations. The proteins were buffer changed into PBS buffer pH 6.0 for the pegylation reaction.
  • the protein concentration in the reaction was between 0.5 and 3.5 mg/mL and a 5- 10 molar excess of mPeg5K- Aldehyde or mPeg20K- Aldehyde (Nektar) supplemented with 20 mM cyanoborohydride (NaCNBH3) was used.
  • the reaction was carried out at 2O 0 C for 16 hours.
  • Source 15S column GE Healtcare
  • antagonistic activity of the pegylated version was reduced compared to the unpegylated protein.
  • the pegylated protein still has good ILl blocking efficacy.
  • Example 4 Pharmacokinetic analysis of trimeric ILlRa proteins in male Lewis rats after i.v. injection
  • ILlRa polypeptides Three of the trimeric ILlRa polypeptides described in the previous examples were chosen for pharmacokinetic analysis. The differences in the constructs were in the N- terminus of the trimerization domain: full length (FL), first nine amino acids truncated (110) and the first 16 amino acids truncated (V 17).
  • the 110 construct represents a naturally occurring deletion variant of the trimerization domain and lacks the O-glycosylation site at Thr 4.
  • the Vl 7 derivative represents a deletion of the first exon encoding the trimerization domain and lacks a characterized heparin binding site. This site is also partially removed in the 110 construct. In vitro efficacy of the IL-IRa molecules was verified in a U937 cell assay as shown in Figure 6.
  • test compound was dissolved in vehicle (4.4 mM NaCitrate, pH 6.5, 93.8 mM NaCl, 0.33 mM EDTA, 0.7 g TWEEN ® -80) and administered through the tail vein (vena sacralis media) or the hind paw vein (vena saphena).
  • vehicle 4.4 mM NaCitrate, pH 6.5, 93.8 mM NaCl, 0.33 mM EDTA, 0.7 g TWEEN ® -80
  • Plasma samples were then collected from four animals per time-point at baseline (zero hours) and 0.5, 1, 2, 4, 8, 12, 24, 48, 72 h post dosing. Blood samples of approximately 100 ⁇ l were collected from the tip of the tails in MicrotainersTM. Plasma was collected and transferred into polypropylene tubes. Plasma samples were then stored at ⁇ -70°C until measurements were performed. Animals were then sacrificed by CO 2 inhalation and the carcasses were discarded without pathological examination. The IL-IRa compound levels and KINERET ® levels in plasma were then determined by ELISA.
  • the average body weight of each rat was 250 grams. Assuming that the rat average blood volume was 16.5 mL a theoretical maximum initial concentration of the compounds of 1,500,000 ng/mL was calculated after i.v. injection. These concentrations are shown in Figure 7. This starting level was used as starting value for the analysis. No observations of side effects or changes in animal well being were observed.
  • Example 5 Production of Met-IlO-TripT-ILlra and GG-V17-TripT-ILlra and Rat CIA model
  • Both molecules were produced by BL21 AI bacteria in 10 L fermentor runs using either 2 x TY medium (Met-I10-TripT-IL-lra ) or chemically defined minimal medium (GG- V17-TripT-IL-lra).
  • Cell pellets were obtained by centrifugation at 5887 x g for 20 min, then resuspended in 10 mJVI Na 2 HPO 4 pH 6.
  • Met-IO-TripT-IL-lra the soluble cell fraction containing the protein of interest was obtained by high pressure homogenization (2 x 17.000 psi) followed by 10 min centrifugation at 10.000 x g.
  • the supernatant was diluted with 10 mM Na 2 HPO 4 pH 7.4 and run over a SP-Sepharose FF column (cation exchange, GE Healthcare) followed by Q-Sepharose FF (anion exchange. GE Healthcare) using an AKTA fPLC.
  • proteins were run through a Mustang E filter (Pall) to remove endotoxin, followed by buffer exchange into PBS pH 7.4 and concentration to 50 mg/mL.
  • the GG-Vl 7 -TripT-IL-lRa protein was expressed as a fusion protein comprising an N-terminal booster domain, phage CII protein, followed by a human Granzyme B cleavage site.
  • the GG-V 17 - TripT-IL-lRa was purified from fermentation cell pellets by homogenization in lysis buffer containing lysozyme followed by centrifugation for 25 min at 8000 rpm. The supernatant was then run through a Fractogel ® EMD Chelate (M) column (EMD Chemicals Inc.), and the eluate was buffer exchanged into 20 mM Tris pH 7.5, 150 mM NaCl. The protein fraction was then digested with recombinant human Granzyme B (made in house, ref to patent).
  • Aggregates were ⁇ 0.5% as determined by analytical SEC ( Figure 9) and host cell protein ⁇ 6 ng/mL.
  • Two batches (LM022, LM023) of Met-I10-TripT-IL-lra and two batches (CF019, CF020) of GG-Vl 7-TripT-IL- Ira were tested in above assays.
  • mice with 4-day established type II collagen arthritis were treated subcutaneously (SC), daily (QD) on arthritis days 1-3 with Vehicle (10 mM phosphate buffer pH 7.4), or equimolar amounts of IL-lra administering either monomeric IL-lra (100 mg/kg KINERET ® ), or trimerized ILlra (120 mg/kg Met-IlO-TripT-ILlra, or 120 mg/kg GG-V17- TripT-ILlra).
  • Rats were weighed on days 0-4 of arthritis, and caliper measurements of ankles were taken every day beginning on day 0 of arthritis (study day 9). After final body weight measurement, animals were euthanized, and hind paws were transected at the level of the medial and lateral malleolus and weighed (paired).
  • Met-IlO-TripT-ILlra QD treatment resulted in significantly reduced ankle diameter AUC compared to KINERET ® QD treatment (p ⁇ 0.035 at the end of the study).
  • GG-V 17-TripT-IL Ira QD treatment resulted in significantly reduced ankle diameter AUC compared to KINERET ® QD treatment at the end of the study (pO.OOl).
  • STZ Sigma Aldrich
  • mice were administered once daily for five successive days at 50 mg/kg i.p. to fasted C57BL/6J male mice.
  • the mice gradually developed higher levels of blood glucose from Day 1 to Day 4.
  • the levels rose from 6.9 nmol/L to 13.1 nmol/L during the STZ induction period.
  • the treatment groups were as shown in Table 5.
  • the study period was 28 days and the mice were weighed once weekly during the treatment period. Blood glucose levels were measured every other day during the study period in order to monitor development of diabetes. A droplet of whole blood was collected by tail vein bleeding and placed on an Ascensia ELITE ® blood glucose test strip and analyzed with an Ascensia ELITE ® blood glucose meter (Bayer). The values were recorded, and x-fold increase in any given group compared to levels at treatment initiation was calculated. Clinical symptoms were observed daily or as appropriate in groups where adverse symptoms occurred.
  • any numerical values recited herein include all values from the lower value to the upper value in increments of one unit provided that there is a separation of at least two units between any lower value and any higher value.
  • concentration of a component or value of a process variable such as, for example, size, angle size, pressure, time and the like, is, for example, from 1 to 90, specifically from 20 to 80, more specifically from 30 to 70, it is intended that values such as 15 to 85, 22 to 68, 43 to 51, 30 to 32, etc. are expressly enumerated in this specification.
  • one unit is considered to be 0.0001, 0.001, 0.01 or 0.1 as appropriate.

Landscapes

  • Health & Medical Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Organic Chemistry (AREA)
  • General Health & Medical Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Engineering & Computer Science (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Animal Behavior & Ethology (AREA)
  • General Chemical & Material Sciences (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Public Health (AREA)
  • Veterinary Medicine (AREA)
  • Biomedical Technology (AREA)
  • Genetics & Genomics (AREA)
  • Physical Education & Sports Medicine (AREA)
  • Neurology (AREA)
  • Rheumatology (AREA)
  • Neurosurgery (AREA)
  • Molecular Biology (AREA)
  • Immunology (AREA)
  • Zoology (AREA)
  • Diabetes (AREA)
  • Biochemistry (AREA)
  • Hematology (AREA)
  • Orthopedic Medicine & Surgery (AREA)
  • Biophysics (AREA)
  • Pulmonology (AREA)
  • Pain & Pain Management (AREA)
  • Toxicology (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Gastroenterology & Hepatology (AREA)
  • Oncology (AREA)
  • Wood Science & Technology (AREA)
  • General Engineering & Computer Science (AREA)
  • Biotechnology (AREA)
  • Urology & Nephrology (AREA)
  • Obesity (AREA)
EP08837224A 2007-10-08 2008-10-08 Trimeres il-1ra Withdrawn EP2195337A1 (de)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US97825407P 2007-10-08 2007-10-08
PCT/US2008/079216 WO2009048961A1 (en) 2007-10-08 2008-10-08 Trimeric il-1ra

Publications (1)

Publication Number Publication Date
EP2195337A1 true EP2195337A1 (de) 2010-06-16

Family

ID=40336426

Family Applications (1)

Application Number Title Priority Date Filing Date
EP08837224A Withdrawn EP2195337A1 (de) 2007-10-08 2008-10-08 Trimeres il-1ra

Country Status (6)

Country Link
EP (1) EP2195337A1 (de)
JP (1) JP2010539995A (de)
CN (1) CN101889022A (de)
AU (1) AU2008310902A1 (de)
CA (1) CA2701705A1 (de)
WO (1) WO2009048961A1 (de)

Families Citing this family (14)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20090143276A1 (en) * 2007-10-08 2009-06-04 Anaphore, Inc. Trimeric IL-1Ra
CN102317314A (zh) * 2008-10-10 2012-01-11 阿纳福公司 结合trail-r1和trail-r2的多肽
CN101690801B (zh) * 2009-10-26 2012-08-01 上海交通大学 白细胞介素-1受体拮抗剂的用途及其药物组合物
MX360097B (es) 2010-07-29 2018-10-22 Eleven Biotherapeutics Inc Agonistas y antagonistas del receptor tipo i de il-1 quimérico.
EP2611419A2 (de) * 2010-08-30 2013-07-10 F.Hoffmann-La Roche Ag Verfahren zur herstellung eines tetranectin-apolipoprotein-a1-partikels, dieses partikel selbst und seine verwendung
KR20130117760A (ko) * 2010-08-30 2013-10-28 에프. 호프만-라 로슈 아게 테트라넥틴-아포지단백질 a-i 입자의 제조 방법, 이에 의해 제조된 지질 입자 및 이의 용도
US9359405B2 (en) * 2011-03-14 2016-06-07 Phlogo Aps Antagonists of the interleukin-1 receptor
CA2838070A1 (en) * 2011-08-25 2013-02-28 F. Hoffmann-La Roche Ag Shortened tetranectin-apolipoprotein a-i fusion protein, a lipid particle containing it, and uses thereof
CN112142855A (zh) 2012-05-18 2020-12-29 爱德迪安(北京)生物技术有限公司 用于糖尿病治疗的蛋白、蛋白缀合物及其应用
WO2014160371A1 (en) 2013-03-13 2014-10-02 Eleven Biotherapeutics, Inc. Chimeric cytokine formulations for ocular delivery
CN105367663A (zh) * 2014-08-31 2016-03-02 复旦大学 一种长效白细胞介素-1受体拮抗剂重组融合蛋白及其制备方法和用途
CN104587457B (zh) * 2015-01-13 2017-03-22 广东海大畜牧兽医研究院有限公司 一种利用难溶或不溶蛋白、多肽抗原制备纳米微粒疫苗的方法
AU2020449791A1 (en) * 2020-05-25 2023-02-02 Beijing Vdjbio Co., Ltd. An interleukin-1 receptor antagonist and a fusion protein containing the same
CN114457086B (zh) * 2022-03-02 2022-11-15 上海勉亦生物科技有限公司 白介素1受体拮抗蛋白的表达盒以及基于aav的基因递送系统

Family Cites Families (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN1268641C (zh) * 2000-11-10 2006-08-09 普罗蒂奥制药公司 载脂蛋白类似物
CN1665931A (zh) * 2002-05-31 2005-09-07 西根控股有限公司 从膜转运序列衍生的自身聚集或自身团聚嵌合蛋白
CN101899106A (zh) * 2002-10-29 2010-12-01 阿纳福公司 三聚细胞因子的三聚结合蛋白
EP1720899A2 (de) * 2004-02-23 2006-11-15 Borean Pharma A/S Multimerisierte hiv-fusionsinhibitoren
KR20070094909A (ko) * 2004-12-02 2007-09-27 도만티스 리미티드 혈청 알부민 및 glp-1 또는 pyy를 표적으로 삼는이중특이성 도메인을 갖는 항체

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
See references of WO2009048961A1 *

Also Published As

Publication number Publication date
JP2010539995A (ja) 2010-12-24
CN101889022A (zh) 2010-11-17
CA2701705A1 (en) 2009-04-16
WO2009048961A1 (en) 2009-04-16
AU2008310902A1 (en) 2009-04-16

Similar Documents

Publication Publication Date Title
WO2009048961A1 (en) Trimeric il-1ra
US20100028995A1 (en) Tetranectin Trimerizing Polypeptides
EP1615952B1 (de) Behandlungsmethoden von entzündungskrankheiten mit spezifisch an menschliches angiopoietin-2 bindenden wirkstoffen
US20090143276A1 (en) Trimeric IL-1Ra
AU2009244878B2 (en) IL-17RA-IL-17RB antagonists and uses thereof
JP6938565B2 (ja) Uti融合タンパク質
WO2013016220A1 (en) Il-17 receptor a is required for il-17c biology
AU2008242665B2 (en) Identification and method for using the Pre-Ligand Assembly Domain of the IL-17 receptor
KR20020073139A (ko) 인터루킨-1 수용체 길항물질 - 유사 분자 및 이의 용도
AU2013205228A1 (en) Identification and method for using the Pre-Ligand assembly domain of the IL-17 receptor

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: 20100331

AK Designated contracting states

Kind code of ref document: A1

Designated state(s): AT BE BG CH CY CZ DE DK EE ES FI FR GB GR HR HU IE IS IT LI LT LU LV MC MT NL NO PL PT RO SE SI SK TR

AX Request for extension of the european patent

Extension state: AL BA MK RS

DAX Request for extension of the european patent (deleted)
REG Reference to a national code

Ref country code: HK

Ref legal event code: DE

Ref document number: 1145088

Country of ref document: HK

17Q First examination report despatched

Effective date: 20110415

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: 20110826

REG Reference to a national code

Ref country code: HK

Ref legal event code: WD

Ref document number: 1145088

Country of ref document: HK