WO2014126582A1 - IL-1β INHIBITOR COMPOSITION AND USE THEREOF - Google Patents

IL-1β INHIBITOR COMPOSITION AND USE THEREOF Download PDF

Info

Publication number
WO2014126582A1
WO2014126582A1 PCT/US2013/026349 US2013026349W WO2014126582A1 WO 2014126582 A1 WO2014126582 A1 WO 2014126582A1 US 2013026349 W US2013026349 W US 2013026349W WO 2014126582 A1 WO2014126582 A1 WO 2014126582A1
Authority
WO
WIPO (PCT)
Prior art keywords
acid sequence
human
disease
polypeptide
amino acid
Prior art date
Application number
PCT/US2013/026349
Other languages
French (fr)
Inventor
Yan Lavrovsky
Ting Xu
Alexey REPIK
Tao Xu
Vasily IGNATIEV
Mikhail Samsonov
Original Assignee
R-PHARM, CJSC (Closed Joint Stock Company)
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Family has litigation
First worldwide family litigation filed litigation Critical https://patents.darts-ip.com/?family=51354448&utm_source=google_patent&utm_medium=platform_link&utm_campaign=public_patent_search&patent=WO2014126582(A1) "Global patent litigation dataset” by Darts-ip is licensed under a Creative Commons Attribution 4.0 International License.
Priority to PT138752118T priority Critical patent/PT2956471T/en
Priority to DK13875211.8T priority patent/DK2956471T3/en
Priority to EP21208571.6A priority patent/EP4050019A1/en
Priority to BR112015019729-9A priority patent/BR112015019729B1/en
Priority to PL13875211.8T priority patent/PL2956471T3/en
Priority to FIEP13875211.8T priority patent/FI2956471T3/en
Priority to SI201332081T priority patent/SI2956471T1/en
Priority to PCT/US2013/026349 priority patent/WO2014126582A1/en
Priority to LTEPPCT/US2013/026349T priority patent/LT2956471T/en
Priority to KR1020157025393A priority patent/KR101989551B1/en
Priority to RS20240597A priority patent/RS65559B1/en
Priority to EP13875211.8A priority patent/EP2956471B1/en
Priority to JP2015557984A priority patent/JP6225197B2/en
Priority to CN201380075648.9A priority patent/CN105431448B/en
Priority to AU2013378122A priority patent/AU2013378122B2/en
Priority to NZ71090013A priority patent/NZ710900A/en
Priority to MX2015010438A priority patent/MX2015010438A/en
Priority to EA201500842A priority patent/EA033269B1/en
Priority to SG11201506389YA priority patent/SG11201506389YA/en
Application filed by R-PHARM, CJSC (Closed Joint Stock Company) filed Critical R-PHARM, CJSC (Closed Joint Stock Company)
Publication of WO2014126582A1 publication Critical patent/WO2014126582A1/en
Priority to IL240553A priority patent/IL240553B/en
Priority to PH12015501796A priority patent/PH12015501796A1/en
Priority to HK16107308.4A priority patent/HK1219281A1/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/705Receptors; Cell surface antigens; Cell surface determinants
    • C07K14/715Receptors; Cell surface antigens; Cell surface determinants for cytokines; for lymphokines; for interferons
    • C07K14/7155Receptors; Cell surface antigens; Cell surface determinants for cytokines; for lymphokines; for interferons for interleukins [IL]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/16Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • A61K38/17Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • A61K38/177Receptors; Cell surface antigens; Cell surface determinants
    • A61K38/1793Receptors; Cell surface antigens; Cell surface determinants for cytokines; for lymphokines; for interferons
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/50Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates
    • A61K47/51Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent
    • A61K47/68Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an antibody, an immunoglobulin or a fragment thereof, e.g. an Fc-fragment
    • A61K47/6801Drug-antibody or immunoglobulin conjugates defined by the pharmacologically or therapeutically active agent
    • A61K47/6803Drugs conjugated to an antibody or immunoglobulin, e.g. cisplatin-antibody conjugates
    • A61K47/6811Drugs conjugated to an antibody or immunoglobulin, e.g. cisplatin-antibody conjugates the drug being a protein or peptide, e.g. transferrin or bleomycin
    • 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
    • A61P17/00Drugs for dermatological 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
    • A61P27/00Drugs for disorders of the senses
    • A61P27/02Ophthalmic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P27/00Drugs for disorders of the senses
    • A61P27/02Ophthalmic agents
    • A61P27/04Artificial tears; Irrigation solutions
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P27/00Drugs for disorders of the senses
    • A61P27/02Ophthalmic agents
    • A61P27/14Decongestants or antiallergics
    • 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
    • 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
    • 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
    • A61P9/06Antiarrhythmics
    • 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
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2319/00Fusion polypeptide
    • C07K2319/30Non-immunoglobulin-derived peptide or protein having an immunoglobulin constant or Fc region, or a fragment thereof, attached thereto
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2319/00Fusion polypeptide
    • C07K2319/32Fusion polypeptide fusions with soluble part of a cell surface receptor, "decoy receptors"

Definitions

  • the invention relates to the field of biological pharmaceuticals as well as their use in conditions associated with inflammatory disorders (e.g rheumatoid arthritis, Crohn's desease, etc.), diabetes, cardiovascular disease and gout. More specifically, the invention relates to a
  • heterodimeric IL-lRl/IL-lRAcP -derived composition that is capable of inhibiting IL- ⁇ ⁇ cytokine.
  • the interleukin-1 (IL-1) family of cytokines comprises 11 proteins (IL-1F1 to IL-1F11) encoded by 11 distinct genes in humans and mice.
  • IL-l-type cytokines are major mediators of innate immune reactions, and blockade of the founding members IL-1 or IL-1 ⁇ by the interleukin- 1 receptor antagonist (IL-1RA) has demonstrated a central role of IL-1 in a number of human autoinfiammatory diseases.
  • IL-1 or IL- ⁇ rapidly increase messenger R A expression of hundreds of genes in multiple different cell types.
  • IL-1 and IL- ⁇ are restricted at three major levels: (i) synthesis and release, (ii) membrane receptors, and (iii) intracellular signal transduction.
  • This pathway summarizes extracellular and intracellular signaling of IL-1 or IL- ⁇ , including positive- and negative-feedback mechanisms that amplify or terminate the IL-1 response.
  • combinatorial phosphorylation and ubiquitination events results in activation of nuclear factor kappa-B signaling and the JNK and p38 mitogen-activated protein kinase pathways, which, cooperatively, induce the expression of canonical IL-1 target genes (such as IL-6, IL-8, MCP-1, COX-2, IB, IL-1, IL- ⁇ , MKP-1) by transcriptional and posttranscriptional mechanisms.
  • canonical IL-1 target genes such as IL-6, IL-8, MCP-1, COX-2, IB, IL-1, IL- ⁇ , MKP-1
  • IL-1 interleukin-1
  • IL-18 and IL-33 cytokines
  • TLRs Toll-like-receptors
  • cytotoxic stresses see Weber A, et al, Sci Signal, 2010 Jan 19;3(105), the entire teachings of which are incorporated by reference herein).
  • IL-1 and IL- ⁇ independently bind the type I IL-1 receptor (IL-1R1), which is ubiquitously expressed.
  • a third specific ligand, the IL-1 receptor antagonist (IL-IRA) binds the IL-1RI with similar specificity and affinity but does not activate the receptor and trigger downstream signaling.
  • the IL-1 receptor accessory protein (IL-lRAcP) serves as a co-receptor that is required for signal transduction of IL-1/IL-1RI complexes, and this co-receptor is also necessary for activation of IL- 1R1 by other IL-1 family members, in particular IL-18 and IL-33.
  • the type II IL-1 receptor (IL- 1R2) binds IL-1 and IL- ⁇ but lacks a signaling-competent cytosolic part and thus serves as a decoy receptor.
  • the IL-IRA, the plasma membrane-anchored IL-1R2, and the naturally occurring "shed" domains of each of the extracellular IL-1 receptor chains (termed sIL-lRI, sIL-lRII, and sIL-lRAcP, where "s” stands for soluble) provide inducible negative regulators of IL-1 signaling in the extracellular space whose abundance, which is regulated by a combination of increased transcription and controlled release, can limit or terminate IL-1 effects.
  • the initial step in IL-1 signal transduction is a ligand-induced conformational change in the first extracellular domain of the IL-1RI that facilitates recruitment of IL-lRacP.
  • TIR Toll- and IL-lR-like domains
  • the trimeric complex rapidly assembles two intracellular signaling proteins, myeloid differentiation primary response gene 88 (MYD88) and interleukin-1 receptor-activated protein kinase (IRAK) 4. Mice lacking MYD88 or IRAK4 show severe defects in IL-1 signaling.
  • humans with mutations in the IRAK4 gene have defects in IL-1RI and Toll-like receptor (TLR) signaling.
  • IL-1, IL-1RI, IL-RAcP Toll-like receptor
  • MYD88, and IRAK4 form a stable IL-l-induced first signaling module. This is paralled by the (auto)phosphorylation of IRAK4, which subsequently phosphorylates IRAKI and IRAK2, and then this is followed by the recruitment and oligomerization of tumor necrosis factor-associated factor (TRAF) 6.
  • IRAKI and 2 function as both adaptors and protein kinases to transmit downstream signals. Complexes of IRAKI, IRAK2, and TRAF6 dissociate from the initial receptor complex, and cells lacking these proteins have impaired activation of the transcription factors nuclear factor kappa-B (NF-kappa-B) and activator protein 1 (AP-1).
  • IL-1 has been linked to the pathology of diabetes, cardiovascular disease, gout and certain types of arthritis (e.g. rheumatoid arthritis (RA)).
  • RA rheumatoid arthritis
  • Rilonacept is an IL-1 antagonist which includes an IL-1 -specific fusion protein which comprises an IL-1 binding portion of the extracellular domain of human ILl-RAcP, an IL-1 binding portion of the extracellular domain of human IL-1RI, and a multimerizing component.
  • This IL-l-specific fusion protein is described in U.S. Pat. No. 6,472,179, U.S. patent publication No. 2003/0143697, published 31 Jul. 2003, U.S. Pat. No. 7,361,350, and U.S. patent publication No. 2005/0197293, published 8 Sep. 2005 (all of which are incorporated by reference herein in their entirety).
  • Rilonacept under the trade name ARCALYST was approved by U.S.
  • FDA Food and Drug Administration
  • Cryopyrin- Associated Periodic Syndromes including Familial Cold Auto-inflammatory Syndrome (FCAS) and Muckle- Wells Syndrome (MWS) in adults and children 12 and older.
  • FCAS Familial Cold Auto-inflammatory Syndrome
  • MFS Muckle- Wells Syndrome
  • the present invention provides for a heterodimeric protein composition capable of binding human IL- ⁇ (GenBank: AAH08678.1).
  • the protein composition comprises a first polypeptide which includes a first amino acid sequence which contains amino acids 18 through 333 of human IL1-R1 (GenBank: AAM88423.1), and a second amino acid sequence which contains a first mutant of a Fc portion of human immunoglobulin gamma- 1 Fc (GenBank: J00228.1).
  • the protein composition also comprises a second polypeptide which includes another first amino acid sequence containing amino acids 21 through 358 of human ILl-RAcP (GenBank: BAA25421.1), and another second amino acid sequence which contains a second mutant of the Fc portion of human immunoglobulin gamma- 1 Fc.
  • the first and second mutants are selected as to favor heterodimeric assembly between the first and second mutants over any homodimeric assembly.
  • the protein composition may be capable of exhibiting human IL- ⁇ binding activity in an ELISA assay with an EC50 of about 50 ng/ml.
  • the first polypeptide of the protein composition may contain amino acid sequence of SEQ ID NO. 1, while the second polypeptide may contain amino acid sequence of SEQ ID NO. 2.
  • the present invention provides for a therapeutic composition.
  • the therapeutic composition comprises a heterodimeric protein composition capable of binding human IL- ⁇ .
  • the protein composition comprises a first polypeptide which includes a first amino acid sequence which contains amino acids 18 through 333 of human IL1-R1, and a second amino acid sequence which contains a first mutant of the Fc portion of human immunoglobulin gamma- 1 Fc.
  • the protein composition also comprises a second polypeptide which includes another first amino acid sequence containing amino acids 21 through 358 of human ILl-RAcP, and another second amino acid sequence which contains a second mutant of the Fc portion of human immunoglobulin gamma- 1 Fc.
  • the first and second mutants are selected as to favor heterodimeric assembly between the first and second mutants over any homodimeric assembly.
  • the therapeutic composition may exhibit a half-life of the heterodimeric protein composition in systemic circulation in mice after a subcutaneous administration at a dose of 5 mg/kg of at least about 88 hours, as assayed by human Fc ELISA.
  • the therapeutic composition may comprise a heterodimeric protein comprised of a first polypeptide containing amino acid sequence of SEQ ID NO. 1 and a second polypeptide containing amino acid sequence of SEQ ID NO. 2.
  • the present invention provides for an isolated nucleic acid encoding a polypeptide comprising amino acid sequence of SEQ ID NO. 3.
  • the codon usage of the nucleic acid may be optimized for high expression of the polypeptide in a mammalian cell.
  • the nucleic acid may contain the sequence of SEQ ID NO. 5.
  • the nucleic acid may comprise an expression vector
  • the present invention provides for an isolated nucleic acid encoding a polypeptide comprising amino acid sequence of SEQ ID NO. 4.
  • the codon usage of the nucleic acid may be optimized for high expression of the polypeptide in a mammalian cell.
  • the nucleic acid may contain the sequence of SEQ ID NO. 6.
  • the nucleic acid may comprise an expression vector.
  • the present invention provides for an isolated nucleic acid of SEQ ID NO. 7.
  • the present invention provides for a heterologous expression system.
  • the expression system harbors an expression vector comprising a nucleic acid sequence encoding a first polypeptide containing amino acid sequence of SEQ ID NO. 3 and another nucleic acid sequence encoding a second polypeptide containing amino acid sequence of SEQ ID NO. 4.
  • the expression vector of the expression system may be harbored in a mammalian cell.
  • the mammalian cell may be a CHO cell.
  • the expression system may be capable of expressing a heterodimeric protein comprising a first polypeptide containing amino acid sequence of SEQ ID NO.
  • the present invention provides for use of a substance for manufacture of a medicament for the treatment or prevention of a disease associated with modulation of activity of human IL- ⁇ ⁇ .
  • the substance comprises a heterodimeric protein comprised of a first polypeptide containing amino acid sequence of SEQ ID NO. 1 and a second polypeptide containing amino acid sequence of SEQ ID NO. 2.
  • the disease associated with modulation of activity of human IL- ⁇ may be an arthritis, a gout, a rheumatoid arthritis, a Cryopyrin- Associated Periodic Syndromes (CAPS), a scleroderma, a diabetes, a atherosclerosis, a dry eye disease, an ocular allergy, or an uveitis.
  • the present invention provides for a method of treating or preventing a disease or condition associated with modulation of activity of human IL- 1 ⁇ .
  • the method comprising administering to a patient in need for treating or preventing a disease associated with modulation of activity of human IL- ⁇ a therapeutically effective amount of a pharmaceutical composition.
  • the pharmaceutical composition comprising a heterodimeric protein comprised of a first polypeptide containing amino acid sequence of SEQ ID NO. 1 and a second polypeptide containing amino acid sequence of SEQ ID NO. 2.
  • the disease associated with modulation of activity of human IL- ⁇ may be an arthritis, a gout, a rheumatoid arthritis, a Cryopyrin- Associated Periodic Syndromes (CAPS), a scleroderma, a diabetes, a atherosclerosis, a dry eye disease, an ocular allergy, or an uveitis.
  • the present invention provides for a heterodimeric protein composition capable of binding human IL- ⁇ .
  • the protein composition is capable of inhibiting human IL-6 production in human lung fibroblasts in response to treating the fibroblasts with human IL- ⁇ ⁇ .
  • the inhibiting is characterized by an IC50 value of about 2.3 pM.
  • Figure 1 illustratively shows a heterodimeric protein assembly of the present teachings comprising an extracellular portion of IL1-R1 fused with an IgG-Fc domain (Fc-II) via a flexible linker and an extracellular portion of ILl-RAcP fused with another IgG-Fc domain (Fc-V) via another flexible linker;
  • Fc-II IgG-Fc domain
  • Fc-V IgG-Fc domain
  • Figure 2 schematically shows the map of PKN012 plasmid and annotated sequence used in the cloning of the polypeptides of the present invention
  • Figure 3 shows representative transfection growth curve obtained in the process of generating stable cell lines for expressing the polypeptides of the present invention
  • Figure 4 shown a size-exclusion HPLC analytical chromatogram of the sample containing heterodimers comprising polypeptide of SEQ ID NO. 1 and polypeptide of SEQ ID NO. 2 after the anion exchange chromatography purification step;
  • Figure 5 shown a SDS-PAGE analysis of the sample containing heterodimers comprising polypeptide of SEQ ID NO. 1 and polypeptide of SEQ ID NO. 2 after the anion exchange chromatography purification step;
  • Figure 6 shows a typical binding curve of a purified sample containing heterodimers comprising polypeptide of SEQ ID NO. 1 and polypeptide of SEQ ID NO. 2 in an ELISA assay using commercially available human IL- 1 ⁇ ;
  • Figure 7 shows the calibration curve obtained in the IL-1 antibody mediated inhibition of IL- 6 production assay experiment
  • Figure 8 shows the results of IL-i -mediated IL-6 production and IL-6 recovery from the culture medium where MRC5 cells were incubated for 24 hours with IL- ⁇ at a final concentration of 50 pg/ml (IL-1 alone) or left untreated (Cells Ctrl) while culture medium not exposed to the cells was spiked with IL-6 at a final concentration of 20 pg/ml to estimate IL-6 recovery;
  • Figure 9 shows the results of the measurements of IL-i -mediated IL-6 production and IL-6 recovery from the culture medium where MRC5 cells were incubated for 24 hours with IL- ⁇ at a final concentration of 50 pg/ml (IL-1 alone) or left untreated (Cells Ctrl). Culture medium not exposed to the cells was spiked with IL-6 at a final concentration of 20 pg/ml to estimate IL-6 recovery;
  • Figure 10 shows the calibration curve obtained in the inhibition of IL-6 production assay conducted with ILlR-FcV-RAcP-FcII heterodimer of the present teachings
  • Figure 11 shows the results of inhibition measurements of IL-i -mediated IL-6 production with ILlR-FcV-RAcP-FcII heterodimer and IL-6 recovery from the culture medium where MRC5 cells were incubated for 24 hours with IL- ⁇ at a final concentration of 50 pg/ml (IL-1 alone) or left untreated (Cells Ctrl) while culture medium not exposed to the cells was spiked with IL-6 at a final concentration of 30 pg/ml to estimate IL-6 recovery (RPH-10 + 30 pg/ml IL6); to ensure consistency between two cell culture plates used for this experiment, the effects of IL-1 on IL-6 production from both plates were compared (IL-1 alone and IL1 alone Plate 2, for plates 1 and 2 respectively); the effect of the highest concentration (20 ⁇ g/ml) of ILlR-FcV-RAcP-FcII heterodimer on IL-6 production was also tested (RPH-10 alone); ILlR-Fc
  • Figure 12 shows the results of the titration curve measurements for LIR-FcV-RAcP-FcII heterodimer experiment where MRC5 cells were incubated with IL- 1 ⁇ or IL- 1 ⁇ pre -mixed with various concentrations of LIR-FcV-RAcP-FcII heterodimer; IL-6 production was measured by Quantakine ELISA and the data was analyzed using 4-parameter fit algorithm; coefficient C at the resulting equation is numerically equal to an IC50 value of 0.3 ng/ml or about 2.4 pM.
  • the teachings disclosed herein are based, in part, upon engineering of a heterodimeric protein assembly that is capable of binding to human IL- ⁇ and attenuating its function.
  • the heterodimeric protein assembly of the present teachings comprises an extracellular portions of IL1-R1 (GenBank: AAM88423.1) and of IL-lRAcP (GenBank: BAA25421.1), or functional fragments thereof. Each, the IL1-R1 portion and the IL-lRAcP portion, is fused to a distinct mutant of Fc portion of the human Ig Gamma- 1 (GenBank: J00228.1).
  • the two distinct Fc mutants in the heterodimeric protein assembly are engineered as to favor the heteromeric dimer formation between the two Fc mutants over any homomeric assembly.
  • a DNA expression vector has been constructed for overproducing the heterodimeric protein assembly in a heterologous protein expression system, and mammalian cells have been prepared stably expressing the heterodimeric protein assembly to a high expression level.
  • a protein purification procedure has been devised allowing obtaining a physiologically relevant substantially pure preparation of the heterodimeric protein assembly of the present teachings.
  • purified protein molecule demonstrates a high degree of specific activity in an in vitro Enzyme -Linked Immunosorbent Assay (ELISA) using human IL- 1 ⁇ (GenBank: AAH08678.1).
  • ELISA Enzyme -Linked Immunosorbent Assay
  • human IL- 1 ⁇ GenBank: AAH08678.1.
  • the protein molecule exhibits an acceptable pharmacokinetics profile upon subcutaneous animal administration, while not resulting in any body weight loss or adverse clinical events.
  • the methods of the invention may include steps of comparing sequences to each other, including wild-type sequence to one or more mutants (sequence variants).
  • Such comparisons typically comprise alignments of polymer sequences, e.g., using sequence alignment programs and/or algorithms that are well known in the art (for example, BLAST, FASTA and MEGALIGN, to name a few).
  • sequence alignment programs and/or algorithms that are well known in the art (for example, BLAST, FASTA and MEGALIGN, to name a few).
  • sequence alignment programs and/or algorithms that are well known in the art (for example, BLAST, FASTA and MEGALIGN, to name a few).
  • the methods of the invention may include statistical calculations, e.g. determination of IC50 or EC50 values, etc..
  • the skilled artisan can readily appreciate that such can be performed using a variety of commercially available software, e.g. PRISM (GraphPad Software Inc, La Jolla, CA, USA) or similar.
  • "Homologous,” in all its grammatical forms and spelling variations, refers to the relationship between two proteins that possess a "common evolutionary origin,” including proteins from superfamilies in the same species of organism, as well as homologous proteins from different species of organism. Such proteins (and their encoding nucleic acids) have sequence homology, as reflected by their sequence similarity, whether in terms of percent identity or by the presence of specific residues or motifs and conserved positions.
  • the term “homologous,” when modified with an adverb such as "highly” may refer to sequence similarity and may or may not relate to a common evolutionary origin.
  • sequence similarity in all its grammatical forms, refers to the degree of identity or correspondence between nucleic acid or amino acid sequences that may or may not share a common evolutionary origin.
  • polypeptides described herein may be comprised of more than one contiguous amino acid chain, thus forming dimers or other oligomeric formations.
  • the polypetides of the present teachings for use in mammals are expressed in mammalian cells that allow for proper post-translational modifications, such as CHO or HEK293 cell lines, although other mammalian expression cell lines are expected to be useful as well. It is therefore anticipated that the polypeptides of the present teachings may be post-translationally modified without substantially effecting its biological function.
  • fusion proteins having at least a biologically active portion of the human IL1-R1 or IL-lRAcP or a functional fragment thereof, and one or more fusion domains.
  • fusion domains include, but are not limited to, polyhistidine, Glu-Glu, glutathione S transferase (GST), thioredoxin, protein A, protein G, an immunoglobulin heavy chain constant region (e.g., an Fc), maltose binding protein (MBP), or human serum albumin.
  • a fusion domain may be selected so as to confer a desired property.
  • the IL1-R1 or IL- IPVACP polypeptide portions may be fused with a domain that stabilizes the IL1-R1 or IL-lRAcP polypeptides in vivo (a "stabilizer” domain), optionally via a suitable peptide linker.
  • a stabilizer means anything that increases the half life of a polypeptide in systemic circulation, regardless of whether this is because of decreased destruction, decreased clearance, or other pharmacokinetic effect. Fusions with the Fc portion of an immunoglobulin are known to confer desirable pharmacokinetic properties on certain proteins. Likewise, fusions to human serum albumin can confer desirable properties.
  • fusion domains that may be selected include multimerizing (e.g., dimerizing, tetramerizing) domains and functional domains that confer an additional biological function, e.g. promoting accumulation at the targeted site of action in vivo.
  • multimerizing e.g., dimerizing, tetramerizing
  • functional domains that confer an additional biological function, e.g. promoting accumulation at the targeted site of action in vivo.
  • the heterodimeric protein assemblies of the present teachings comprise an extracellular portion of ILl-Rl, or a functional fragment thereof, fused with a IgG-Fc domain, and an extracellular portion IL-lRAcP, or a functional fragment thereof, fused with another IgG-Fc domain.
  • the IgG-Fc domain and the another IgG-Fc domain are chosen as to favor a heterodimeric protein assembly over any homodimeric protein assembly.
  • ILl-Rl may be fused with the IgG-Fc domain via a flexible linker, while IL-lRAcP, or a functional fragment thereof, may be fused with the another IgG-Fc domain via the flexible linker of the same amino acid sequence or via another flexible linker.
  • the extracellular portion of ILl-Rl fused with IgG-Fc domain (Fc-II) via a flexible linker may comprise the amino acid sequence of SEQ. ID NO. 1, while IL-lRAcP fused with another IgG-Fc domain (Fc-V) via a flexible linker may comprise the amino acid sequence of SEQ. ID NO. 2.
  • hILl-Rl-hlgGl-Fc polypeptide SEQ ID NO. 1
  • the present teachings provides for a recombinant DNA molecule having an open reading frame coding for a polypeptide comprising the leading 333 amino acids of the human IL1-R1 fused with IgG-Fc domain (Fc-II) via a flexible linker, and for another recombinant DNA molecule having an open reading frame coding for another polypeptide comprising the leading 358 amino acids of the human IL-lRAcP fused with another IgG-Fc domain (Fc-V) via a flexible linker.
  • the polypeptide comprising the leading 333 amino acids of the human IL1-R1 fused with IgG-Fc domain (Fc-II) via a flexible linker comprises the amino acid sequence of SEQ. ID NO. 3.
  • the corresponding to it DNA molecule may comprise the nucleotide sequence of SEQ ID NO. 4.
  • the another polypeptide comprises the leading 358 amino acids of the human IL-lRAcP fused with another IgG-Fc domain (Fc-V) via a flexible linker may comprise the amino acid sequence of SEQ. ID NO. 5.
  • the corresponding to it DNA molecule may comprise the nucleotide sequence of SEQ ID NO. 6.
  • hILl-Rl-hlgGl-Fc polypeptide SEQ ID NO. 3
  • FPPKPKDTLM ISRTPEVTCV WDVSHEDPE VKFNWYVDGV EVHNAKTKPR EEQYNSTYRV 420
  • hILl-Rl-hlgGl-Fc DNA SEQ ID NO. 4
  • GAGCCTGCCA GGATTAAGTG CCCCCTCTTC GAGCACTTTC TGAAGTTCAA CTACAGCACC 180
  • AAAGCCAAAG GGCAGCCCCG AGAACCACAG GTGTACACCC TGCCCCCATG TCGGGATGAG 1500
  • the present invention provides for a recombinant malian expression plasmid for high expression of a polypeptide comprising the leading 333 amino acids of the human ILl-Rl fused with IgG-Fc domain (Fc-II) via a flexible linker, and for another recombinant DNA molecule having an open reading frame coding for another polypeptide comprising the leading 358 amino acids of the human IL-lRAcP fused with another IgG-Fc domain (Fc-V) via a flexible linker.
  • This plasmid comprises two cytomegalovirus (CMV) promoters to drive transcription of the two genes coding for said polypeptide and said another polypeptide, each followed by a transcription termination sequence and a polyadenylation sequence.
  • CMV cytomegalovirus
  • the plasmid also contains an origin of replication and a gene conferring ampicillin resistance, for supporting plasmid propagation and selection in bacteria.
  • the plasmid further contains a gene for Glutamine synthetase, a selectable marker widely used for establishing stable CHOK1 and NSO cell lines.
  • the plasmid of the present invention is illustratively shown in Figure 2.
  • the mammalian expression plasmid of the present teachings comprises the nucleotide sequence of SEQ ID NO. 7.
  • hILl-Rl-hlgGl-Fc-II/ IL-lRAcP- hlgGl-Fc-V expression plasmid (SEQ ID NO. 7)
  • AAAGCAATAG CATCACAAAT TTCACAAATA AAGCATTTTT TTCACTGCAT TCTAGTTGTG 1920
  • GTTCAGACCC ACCCTCCTCA ACGACACAGG CAACTACACC TGCATGCTCA GGAACACCAC 4320 CTACTGCAGC AAGGTGGCCT TCCCTCTCGA GGTGGTCCAG AAGGACAGCT GCTTCAACAG 4380
  • GAGCATCAGC CACAGCAGGA CCGAGGACGA GACCAGGACC CAGATCCTGA GCATCAAGAA 4920
  • GTTTGTGTAT TTTAGATTCC AACCTATGGA ACTGATGAAT GGGAGCAGTG GTGGAATGCC 10020
  • AAATATTCTG TAACCTTTAT AAGTAGGCAT AACAGTTATA ATCATAACAT ACTGTTTTTT 10320
  • GCATACGCGA TATCTGGCGA TAGCGCTTAT ATCGTTTACG GGGGATGGCG ATAGACGACT 10980
  • CTCCCATGCC TCCAGCGACT CATGGTCGCT CGGCAGCTCC TTGCTCCTAA CAGTGGAGGC 12480
  • the present teachings provide for a mammalian expression system for production of a heterodimeric protein assembly comprising a polypeptide comprising amino acid residues 18 through 333 of the human IL1-R1 fused with IgG-Fc domain (Fc-II) via a flexible linker, and another polypeptide comprising amino acid residues 21 through 358 of the human IL- lRAcP fused with another IgG-Fc domain (Fc-V) via a flexible linker.
  • the mammalian expression system of the present invention comprises Chinese hamster ovary cells (CHO-K1) harboring a plasmid comprising nucleotide sequence of SEQ ID NO. 7.
  • the present teachings provide for a method of treatment of a mammal effected by the following disorders associated with IL- ⁇ modulation: arthritis, a gout, a rheumatoid arthritis, a Cryopyrin-Associated Periodic Syndromes (CAPS), a scleroderma, a diabetes, a atherosclerosis, a dry eye disease, an ocular allergy, or an uveitis.
  • disorders associated with IL- ⁇ modulation arthritis, a gout, a rheumatoid arthritis, a Cryopyrin-Associated Periodic Syndromes (CAPS), a scleroderma, a diabetes, a atherosclerosis, a dry eye disease, an ocular allergy, or an uveitis.
  • Example 1 Construction of plasmids for expression of polypeptides of the present invention.
  • Optimized gene sequences comprising sequences encoding residues amino acid residues 1 through 333 of IL-1R1 or residues 1 through 358 of IL-lRAcP were chemically synthesized. The resulting fragments were cloned in-frame into the recipient intermediate vectors comprising sequences coding for Fc-V or Fc-II, via Hindlll and BspEI restriction sites. Obtained positive clones were verified by DNA sequencing. The resulting constructions were termed PKN001 '-IL- lR-FcV and PKNOOl '-RAcP-FcII, respectively.
  • PKN002 The gene encoding IL-lRl-Fc-V was then cloned into a second intermediate vector, termed PKN002, via Hindlll and EcoRI restriction sites. Positive clones were screened by double digestion and the insertion sequence of the correct plasmids was verified by DNA sequencing. The resulting constructions were termed PKN002-IL-lR-FcV.
  • PKN012-ILlR-FcV-RAcP-FcII The final plasmid was termed PKN012-ILlR-FcV-RAcP-FcII.
  • Recombinant plasmid PKN012-ILlR-FcV-RAcP-FcII combines expression cassettes for both RAcP-FcII and IL-lR-FcV.
  • the plasmid can be used for co-expressing RAcP-FcII and IL- lR-FcV proteins in a 1 to 1 ratio under the control of a CMV promoter. The majority of these two fusion proteins would then form ILlR-FcV/RAcP-FcII heterodimers after secretion.
  • the plasmid also expresses Glutamine synthetase (GS) protein via a SV40 promoter, which can be used as a selection marker to generate stable cell lines for ILlR-FcV/RAcP-FcII heterodimer production.
  • GS Glutamine synthetase
  • the plasmid map for PKN012-ILlR-FcV-RAcP-FcII is illustratively shown in Figure 2.
  • Example 2 Generation of stable cell lines for expressing polypeptides of the present invention.
  • a stable clone of CHO-K1 cells co-expressing hILl-Rl-hlgGl-Fc polypeptide of SEQ ID NO. 1 and hIL-lRAcP-hlgGl-Fc polypeptide of SEQ ID NO. 2 has been generated through standard cell biology protocols.
  • the expression plasmid PKN012-ILlR-FcV-RAcP-FcII described in Example 1 was used for generating stable cell lines for high expression of said polypeptides. Expression levels of said polypeptides in a plurality of clones were about or over 100 mg/L in a 7-day batch culture.
  • One clonal cell line showed expression levels of about or over 300 mg/L in shake flask batch culture.
  • CHO-K1 Chinese hamster ovary cells
  • the cells were adapted in house into a CD CHO media. Media and reagent were obtained from commercial source. Upon full adaptation, the cells were grown to high density for a few passages. The resulting cells were subcloned. One of the resulting clones with a doubling time under 20 hrs and good morphology was selected as parental cell line.
  • CHO-K1 cells at passage 4 were cultured in CD-CHO chemically defined media (Invitrogen) containing 6 mM L-Glutamine. The cells were maintained by 1 :3 splits after reaching a cell density of 4xl0 6 cells/ml. Cells were span down by centrifugation and resuspended into 1 ml of CD-CHO chemically defined media (Invitrogen).
  • the chosen highest production cell line was selected and thawed into CD-CHO. Growth curves on the cell line were assessed, and samples were collected daily for cell count, cell viability and ILlR-FcV-RAcP-FcII productivity. Based on these studies, it was determined that selected highest productivity cell line was expressing ILlR-FcV-RAcP-FcII heterodimer in CD-CHO media in amounts necessary to support commercial production. The yield of the heterodimer after purification was at least about 300 mg/L (without any production optimization).
  • Example 3 Purification of polypeptides of the present invention.
  • hILl-Rl-hlgGl-Fc polypeptide of SEQ ID NO. 1 and hIL-lRAcP-hlgGl-Fc polypeptide of SEQ ID NO. 2 were co-expressed in CHO-K1 essentially as described in foregoing Example 2.
  • Cells were harvested and lysed utilizing well established protocols. After cell lysate clarification, the supernatant at protein concentration of about 0.4 mg/ml, containing expressed hlLl-Rl-hlgGl- Fc/ hIL-lRAcP-hlgGl-Fc polypeptides, was applied to a Protein A affinity column.
  • the affinity purification step was carried out according to the procedure outlined in Table 2.
  • the Protein A eluate containing hILl-Rl-hlgGl-Fc/ hIL-lRAcP-hlgGl-Fc at pH of about 3.5 - 3.7 is incubated for 45-60 minutes to inactivate potentially existing in the contaminating materials.
  • the pH adjusted Protein A column eluate was further purified by anion-exchange chromatography (AIEX) utilizing Q Sepharose resin.
  • AIEX anion-exchange chromatography
  • the AIEX step is operated according to a step-elution procedure outlined in Table 3. Pooled elution peak fractions are concentrated by microfiltration to a concentration of about 20 mg/ml, followed by addition of a 20% Sucrose stock solution to a final Sucrose concentration of about 1% (w/v) and freezing at -80°C.
  • hILl-Rl-hlgGl-Fc/ hIL-lRAcP-hlgGl-Fc heterodimer has an apparent molecular weight of about 180 kDa, consisting of two di-sulfide linked monomers, each of an apparent molecular weight of about 90 kDa.
  • the SEC-HPLC operational procedure is outlined in Table 4. Loading sample is diluted with mobile phase to reach protein concentration of about 5 mg/ml.
  • Example 4 Characterization of inhibition of IL-i -induced IL-6 production with IL1R- FcV-RAcP-FcII heterodimer in human cells.
  • the present Example demonstrates potent functional (inhibitory) properties of ILlR-FcV- RAcP-FcII heterodimer in targeting human IL- ⁇ .
  • As a functional comparator previously characterized mouse monoclonal antibodies against IL- ⁇ were used.
  • Human lung fibroblasts MRC5 produce IL-6 in response to treatment with IL- ⁇ ⁇ .
  • Quantification of IL- ⁇ -induced IL-6 production in MRC5 cells was used as the assay functional output.
  • Inhibition of IL-6 production by ILlR-FcV-RAcP-FcII heterodimer and by control anti IL- ⁇ antibody provides a quantitative measure of inhibitory properties of the ILlR-FcV-RAcP-FcII heterodimer preparation.
  • Polypeptides of ILlR-FcV-RAcP-FcII heterodimer (SEQ ID NO. l and SEQ ID NO. 2) were co-expressed and purified essentially as described in the forgoing examples.
  • Cells MRC5 cells, Human Lung Fibroblasts, ATCC Cat # CCL-171, Lot # 59474707.
  • Medium DMEM, Dulbecco's Modification of Eagle's Medium, high glucose (4.5 g/L), Mediatech, Cat # 10-017-CM, Lot # 10017204, supplemented with L-glutamine and lx penn/strep and 10% Fetal Bovine Serum, Omega Scientific, Cat # FB-05, Lot # 105104
  • Reagents IL- ⁇ , Human recombinant, E.
  • the assay employs the quantitative sandwich enzyme immunoassay technique.
  • a microplate is pre-coated with a monoclonal antibody specific for IL-6.
  • MRC5 cells Passage 5 were plated into a 48-well plate (Costar, Cat # 3548) at 2 x 10 3 cells per well, 0.4 ml per well, and incubated for 24 hours;
  • IL- ⁇ was used at a final concentration of 50 pg/ml throughout the experiment;
  • Anti-human IL- ⁇ antibodies were used at final concentrations of 0.192, 0.96, 4.8, 24, 120 and 600 ng/ml;
  • Table 5 Summary of IL-6 production values.
  • control anti-IL- ⁇ antibody is a very potent inhibitor of IL- ⁇ signaling pathway with an IC50 of about 2.1 ng/ml or about 14 pM; (2) plating cell density of 2 x 10 3 cells per well 24 hours prior to the assay setup is adequate; (3) recovery of IL-6 spiked into the culture medium at a final concentration of 20 pg/ml is about 114%.
  • MRC5 cells Passage 6 were plated into 48-well plate (Costar, Cat # 3548) at 2 x 103 cells per well, 0.4 ml per well and incubated for 24 hours;
  • IL- ⁇ was used at a final concentration of 50 pg/ml throughout the experiment;
  • ILlR-FcV-RAcP-FcII heterodimer was used at final concentrations of 0.0536, 0.134, 0.335, 0.839, 2.097, 5.24,13.1, 32.8, 81.9, 204.8, 512, 1280, 3200 and 20000 ng/ml;
  • the cells were incubated in the presence of the test substances for 24 hours and supernatants were collected, centrifuged at 300 x g for 10 min and used for IL-6 ELISA assay.
  • Example 5 Pharmacokinetics (PK) of ILlR-FcV-RAcP-FcII heterodimer after subcutaneous administration in mice.
  • Polypeptides of ILlR-FcV-RAcP-FcII heterodimer were co-expressed and purified essentially as described in the forgoing examples.
  • the polypeptides were formulated in the following buffer: 1% w/v Sucrose, lOOmM Sodium Chloride, 20 mM L-Arginine Hydrochloride, 25 mM Sodium Bicarbonate, pH 6.3.
  • the dosing stock concentration used was 0.5 mg/mL of the polypeptide.
  • mice Fourteen female Balb/c nu/nu mice were randomized based on body weight into seven groups of two animals on Day 0 of the study. A single treatment of the polypeptides (5 mg/kg) was administered subcutaneously (dorsal) on Day 0 to all groups except mice in Group 1 , which were bled via cardiac puncture for plasma preparation on Day 0 of the study. Blood samples were collected from mice via the orbital sinus in the remaining groups at various times throughout the study for preparation of plasma.
  • Body weights were recorded for all animals on the treatment day (Day 0) and then three times per week, including the termination day of each group.
  • mice were culled at specific time points for plasma preparation. Body weight changes were not measured in groups culled for sample collection at 0 hours and within 36 hours of dose administration. All other mice gained body weight and no adverse clinical signs were reported during the study period.
  • Table 7 Mean Human-Fc Protein Concentration ⁇ SEM ⁇ g/mL) at each Time Post- Administration
  • the Human-Fc Protein Concentration was determined by Prism Software based on the mean absorbance of the triplicate samples
  • Example 6 Evaluation of the toxicity of ILlR-FcV-RAcP-FcII heterodimer following 28- days repeated dosing in C57BL6 mice. Following is a repeat dose murine toxicity evaluation study results summary. Treatment of
  • C57BL6 mice with polypeptides of TLlR-FcV-RAcP-FcII heterodimer (SEQ ID NO.l and SEQ ID NO. 2) test article by twice weekly subcutaneous injection at 10, 30 and 100 mg/kg dose levels was well tolerated in the study. There were no unscheduled deaths prior to completion of the study. Test article treatment was not associated with morbidity or clinical signs of toxicity. There was no consistent gender or treatment related effect on body weight. There was a finding in increased food consumption which was considered to be a possible effect of treatment but this was not considered to be adverse.

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Engineering & Computer Science (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • General Health & Medical Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Organic Chemistry (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Animal Behavior & Ethology (AREA)
  • Public Health (AREA)
  • Veterinary Medicine (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • General Chemical & Material Sciences (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Immunology (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Ophthalmology & Optometry (AREA)
  • Rheumatology (AREA)
  • Diabetes (AREA)
  • Zoology (AREA)
  • Gastroenterology & Hepatology (AREA)
  • Molecular Biology (AREA)
  • Epidemiology (AREA)
  • Cell Biology (AREA)
  • Cardiology (AREA)
  • Pain & Pain Management (AREA)
  • Physical Education & Sports Medicine (AREA)
  • Heart & Thoracic Surgery (AREA)
  • Genetics & Genomics (AREA)
  • Biochemistry (AREA)
  • Biophysics (AREA)
  • Toxicology (AREA)
  • Emergency Medicine (AREA)
  • Hematology (AREA)
  • Urology & Nephrology (AREA)
  • Endocrinology (AREA)
  • Obesity (AREA)
  • Orthopedic Medicine & Surgery (AREA)
  • Dermatology (AREA)

Abstract

A therapeutic composition is described that can be used for treating or prevention of diseases association with modulation of activity of human IL-1β. In certain aspects, the disclosed invention is based, on engineering of a heterodimeric protein assembly that is capable of binding to human IL-1β and attenuating its function. The heterodimeric protein assembly comprises an extracellular portions of human IL1-R1 and of human IL-1RAcP, or their functional fragments. Each, the IL1-R1 portion and the IL-1RAcP portion, is fused to a distinct mutant of Fc portion of the human Ig Gamma-1. The two distinct Fc mutants in the heterodimeric protein assembly are engineered as to favor the heteromeric dimer formation between the two Fc mutants over any homomeric assembly. DNA expression vectors and expression systems for overproducing the polypeptides in mammalian cells are also provided for.

Description

IL-ip INHIBITOR COMPOSITION AND USE THEREOF
FIELD OF THE INVENTION
Generally, the invention relates to the field of biological pharmaceuticals as well as their use in conditions associated with inflammatory disorders (e.g rheumatoid arthritis, Crohn's desease, etc.), diabetes, cardiovascular disease and gout. More specifically, the invention relates to a
heterodimeric IL-lRl/IL-lRAcP -derived composition that is capable of inhibiting IL-Ι β cytokine.
BACKGROUND
The interleukin-1 (IL-1) family of cytokines comprises 11 proteins (IL-1F1 to IL-1F11) encoded by 11 distinct genes in humans and mice. IL-l-type cytokines are major mediators of innate immune reactions, and blockade of the founding members IL-1 or IL-1 β by the interleukin- 1 receptor antagonist (IL-1RA) has demonstrated a central role of IL-1 in a number of human autoinfiammatory diseases. IL-1 or IL-Ιβ rapidly increase messenger R A expression of hundreds of genes in multiple different cell types. The potent proinflammatory activities of IL-1 and IL-Ιβ are restricted at three major levels: (i) synthesis and release, (ii) membrane receptors, and (iii) intracellular signal transduction. This pathway summarizes extracellular and intracellular signaling of IL-1 or IL-Ιβ, including positive- and negative-feedback mechanisms that amplify or terminate the IL-1 response. In response to ligand binding of the receptor, a complex sequence of
combinatorial phosphorylation and ubiquitination events results in activation of nuclear factor kappa-B signaling and the JNK and p38 mitogen-activated protein kinase pathways, which, cooperatively, induce the expression of canonical IL-1 target genes (such as IL-6, IL-8, MCP-1, COX-2, IB, IL-1, IL-Ιβ, MKP-1) by transcriptional and posttranscriptional mechanisms. Of note, most intracellular components that participate in the cellular response to IL-1 also mediate responses to other cytokines (IL-18 and IL-33), Toll-like-receptors (TLRs), and many forms of cytotoxic stresses (see Weber A, et al, Sci Signal, 2010 Jan 19;3(105), the entire teachings of which are incorporated by reference herein).
IL-1 and IL-Ιβ independently bind the type I IL-1 receptor (IL-1R1), which is ubiquitously expressed. A third specific ligand, the IL-1 receptor antagonist (IL-IRA), binds the IL-1RI with similar specificity and affinity but does not activate the receptor and trigger downstream signaling. The IL-1 receptor accessory protein (IL-lRAcP) serves as a co-receptor that is required for signal transduction of IL-1/IL-1RI complexes, and this co-receptor is also necessary for activation of IL- 1R1 by other IL-1 family members, in particular IL-18 and IL-33. The type II IL-1 receptor (IL- 1R2) binds IL-1 and IL-Ιβ but lacks a signaling-competent cytosolic part and thus serves as a decoy receptor. The IL-IRA, the plasma membrane-anchored IL-1R2, and the naturally occurring "shed" domains of each of the extracellular IL-1 receptor chains (termed sIL-lRI, sIL-lRII, and sIL-lRAcP, where "s" stands for soluble) provide inducible negative regulators of IL-1 signaling in the extracellular space whose abundance, which is regulated by a combination of increased transcription and controlled release, can limit or terminate IL-1 effects.
The initial step in IL-1 signal transduction is a ligand-induced conformational change in the first extracellular domain of the IL-1RI that facilitates recruitment of IL-lRacP. Through conserved cytosolic regions called Toll- and IL-lR-like (TIR) domains, the trimeric complex rapidly assembles two intracellular signaling proteins, myeloid differentiation primary response gene 88 (MYD88) and interleukin-1 receptor-activated protein kinase (IRAK) 4. Mice lacking MYD88 or IRAK4 show severe defects in IL-1 signaling. Similarly, humans with mutations in the IRAK4 gene have defects in IL-1RI and Toll-like receptor (TLR) signaling. IL-1, IL-1RI, IL-RAcP,
MYD88, and IRAK4 form a stable IL-l-induced first signaling module. This is paralled by the (auto)phosphorylation of IRAK4, which subsequently phosphorylates IRAKI and IRAK2, and then this is followed by the recruitment and oligomerization of tumor necrosis factor-associated factor (TRAF) 6. IRAKI and 2 function as both adaptors and protein kinases to transmit downstream signals. Complexes of IRAKI, IRAK2, and TRAF6 dissociate from the initial receptor complex, and cells lacking these proteins have impaired activation of the transcription factors nuclear factor kappa-B (NF-kappa-B) and activator protein 1 (AP-1).
Overproduction of IL-1 is the cause of many inflammatory disorders. For example, IL-1 has been linked to the pathology of diabetes, cardiovascular disease, gout and certain types of arthritis (e.g. rheumatoid arthritis (RA)).
Rilonacept is an IL-1 antagonist which includes an IL-1 -specific fusion protein which comprises an IL-1 binding portion of the extracellular domain of human ILl-RAcP, an IL-1 binding portion of the extracellular domain of human IL-1RI, and a multimerizing component. This IL-l-specific fusion protein is described in U.S. Pat. No. 6,472,179, U.S. patent publication No. 2003/0143697, published 31 Jul. 2003, U.S. Pat. No. 7,361,350, and U.S. patent publication No. 2005/0197293, published 8 Sep. 2005 (all of which are incorporated by reference herein in their entirety). Rilonacept under the trade name ARCALYST was approved by U.S. Food and Drug Administration (FDA) for the treatment of Cryopyrin- Associated Periodic Syndromes (CAPS), including Familial Cold Auto-inflammatory Syndrome (FCAS) and Muckle- Wells Syndrome (MWS) in adults and children 12 and older. Further clinical trials of rilonacept are currently under way, i.e. for gout.
SUMMARY OF THE INVENTION This summary is provided to introduce a selection of concepts in a simplified form that are further described below in the Detailed Description. This summary is not intended to identify key features or essential features of the claimed subject matter, nor is it intended to be used as an aid in determining the scope of the claimed subject matter.
In certain aspects, the present invention provides for a heterodimeric protein composition capable of binding human IL-Ιβ (GenBank: AAH08678.1). The protein composition comprises a first polypeptide which includes a first amino acid sequence which contains amino acids 18 through 333 of human IL1-R1 (GenBank: AAM88423.1), and a second amino acid sequence which contains a first mutant of a Fc portion of human immunoglobulin gamma- 1 Fc (GenBank: J00228.1). The protein composition also comprises a second polypeptide which includes another first amino acid sequence containing amino acids 21 through 358 of human ILl-RAcP (GenBank: BAA25421.1), and another second amino acid sequence which contains a second mutant of the Fc portion of human immunoglobulin gamma- 1 Fc. In the protein composition, the first and second mutants are selected as to favor heterodimeric assembly between the first and second mutants over any homodimeric assembly. The protein composition may be capable of exhibiting human IL-Ιβ binding activity in an ELISA assay with an EC50 of about 50 ng/ml. The first polypeptide of the protein composition may contain amino acid sequence of SEQ ID NO. 1, while the second polypeptide may contain amino acid sequence of SEQ ID NO. 2.
In certain aspects, the present invention provides for a therapeutic composition. The therapeutic composition comprises a heterodimeric protein composition capable of binding human IL-Ιβ. The protein composition comprises a first polypeptide which includes a first amino acid sequence which contains amino acids 18 through 333 of human IL1-R1, and a second amino acid sequence which contains a first mutant of the Fc portion of human immunoglobulin gamma- 1 Fc. The protein composition also comprises a second polypeptide which includes another first amino acid sequence containing amino acids 21 through 358 of human ILl-RAcP, and another second amino acid sequence which contains a second mutant of the Fc portion of human immunoglobulin gamma- 1 Fc. In the protein composition, the first and second mutants are selected as to favor heterodimeric assembly between the first and second mutants over any homodimeric assembly. The therapeutic composition may exhibit a half-life of the heterodimeric protein composition in systemic circulation in mice after a subcutaneous administration at a dose of 5 mg/kg of at least about 88 hours, as assayed by human Fc ELISA. The therapeutic composition may comprise a heterodimeric protein comprised of a first polypeptide containing amino acid sequence of SEQ ID NO. 1 and a second polypeptide containing amino acid sequence of SEQ ID NO. 2.
In certain aspects, the present invention provides for an isolated nucleic acid encoding a polypeptide comprising amino acid sequence of SEQ ID NO. 3. The codon usage of the nucleic acid may be optimized for high expression of the polypeptide in a mammalian cell. 1. The nucleic acid may contain the sequence of SEQ ID NO. 5. The nucleic acid may comprise an expression vector
In certain aspects, the present invention provides for an isolated nucleic acid encoding a polypeptide comprising amino acid sequence of SEQ ID NO. 4. The codon usage of the nucleic acid may be optimized for high expression of the polypeptide in a mammalian cell. The nucleic acid may contain the sequence of SEQ ID NO. 6. The nucleic acid may comprise an expression vector.
In certain aspects, the present invention provides for an isolated nucleic acid of SEQ ID NO. 7. In certain aspects, the present invention provides for a heterologous expression system. The expression system harbors an expression vector comprising a nucleic acid sequence encoding a first polypeptide containing amino acid sequence of SEQ ID NO. 3 and another nucleic acid sequence encoding a second polypeptide containing amino acid sequence of SEQ ID NO. 4. The expression vector of the expression system may be harbored in a mammalian cell. The mammalian cell may be a CHO cell. The expression system may be capable of expressing a heterodimeric protein comprising a first polypeptide containing amino acid sequence of SEQ ID NO. 1 and a second polypeptide containing amino acid sequence of SEQ ID NO. 2. The level of expression of the heterodimeric protein may be at least 300 mg per liter of cell culture. In certain aspects, the present invention provides for use of a substance for manufacture of a medicament for the treatment or prevention of a disease associated with modulation of activity of human IL-Ι β . The substance comprises a heterodimeric protein comprised of a first polypeptide containing amino acid sequence of SEQ ID NO. 1 and a second polypeptide containing amino acid sequence of SEQ ID NO. 2. The disease associated with modulation of activity of human IL-Ιβ may be an arthritis, a gout, a rheumatoid arthritis, a Cryopyrin- Associated Periodic Syndromes (CAPS), a scleroderma, a diabetes, a atherosclerosis, a dry eye disease, an ocular allergy, or an uveitis.
In certain aspects, the present invention provides for a method of treating or preventing a disease or condition associated with modulation of activity of human IL- 1 β. The method comprising administering to a patient in need for treating or preventing a disease associated with modulation of activity of human IL-Ιβ a therapeutically effective amount of a pharmaceutical composition. The pharmaceutical composition comprising a heterodimeric protein comprised of a first polypeptide containing amino acid sequence of SEQ ID NO. 1 and a second polypeptide containing amino acid sequence of SEQ ID NO. 2. The disease associated with modulation of activity of human IL-Ιβ may be an arthritis, a gout, a rheumatoid arthritis, a Cryopyrin- Associated Periodic Syndromes (CAPS), a scleroderma, a diabetes, a atherosclerosis, a dry eye disease, an ocular allergy, or an uveitis.
In certain aspects the present invention provides for a heterodimeric protein composition capable of binding human IL-Ιβ. The protein composition is capable of inhibiting human IL-6 production in human lung fibroblasts in response to treating the fibroblasts with human IL-Ι β. The inhibiting is characterized by an IC50 value of about 2.3 pM.
BRIEF DESCRIPTION OF THE DRAWINGS
The following drawings and descriptions are provided to aid in the understanding of the invention:
Figure 1 illustratively shows a heterodimeric protein assembly of the present teachings comprising an extracellular portion of IL1-R1 fused with an IgG-Fc domain (Fc-II) via a flexible linker and an extracellular portion of ILl-RAcP fused with another IgG-Fc domain (Fc-V) via another flexible linker;
Figure 2 schematically shows the map of PKN012 plasmid and annotated sequence used in the cloning of the polypeptides of the present invention;
Figure 3 shows representative transfection growth curve obtained in the process of generating stable cell lines for expressing the polypeptides of the present invention;
Figure 4 shown a size-exclusion HPLC analytical chromatogram of the sample containing heterodimers comprising polypeptide of SEQ ID NO. 1 and polypeptide of SEQ ID NO. 2 after the anion exchange chromatography purification step;
Figure 5 shown a SDS-PAGE analysis of the sample containing heterodimers comprising polypeptide of SEQ ID NO. 1 and polypeptide of SEQ ID NO. 2 after the anion exchange chromatography purification step;
Figure 6 shows a typical binding curve of a purified sample containing heterodimers comprising polypeptide of SEQ ID NO. 1 and polypeptide of SEQ ID NO. 2 in an ELISA assay using commercially available human IL- 1 β;
Figure 7 shows the calibration curve obtained in the IL-1 antibody mediated inhibition of IL- 6 production assay experiment;
Figure 8 shows the results of IL-i -mediated IL-6 production and IL-6 recovery from the culture medium where MRC5 cells were incubated for 24 hours with IL-Ιβ at a final concentration of 50 pg/ml (IL-1 alone) or left untreated (Cells Ctrl) while culture medium not exposed to the cells was spiked with IL-6 at a final concentration of 20 pg/ml to estimate IL-6 recovery;
Figure 9 shows the results of the measurements of IL-i -mediated IL-6 production and IL-6 recovery from the culture medium where MRC5 cells were incubated for 24 hours with IL-Ιβ at a final concentration of 50 pg/ml (IL-1 alone) or left untreated (Cells Ctrl). Culture medium not exposed to the cells was spiked with IL-6 at a final concentration of 20 pg/ml to estimate IL-6 recovery;
Figure 10 shows the calibration curve obtained in the inhibition of IL-6 production assay conducted with ILlR-FcV-RAcP-FcII heterodimer of the present teachings;
Figure 11 shows the results of inhibition measurements of IL-i -mediated IL-6 production with ILlR-FcV-RAcP-FcII heterodimer and IL-6 recovery from the culture medium where MRC5 cells were incubated for 24 hours with IL-Ιβ at a final concentration of 50 pg/ml (IL-1 alone) or left untreated (Cells Ctrl) while culture medium not exposed to the cells was spiked with IL-6 at a final concentration of 30 pg/ml to estimate IL-6 recovery (RPH-10 + 30 pg/ml IL6); to ensure consistency between two cell culture plates used for this experiment, the effects of IL-1 on IL-6 production from both plates were compared (IL-1 alone and IL1 alone Plate 2, for plates 1 and 2 respectively); the effect of the highest concentration (20 μg/ml) of ILlR-FcV-RAcP-FcII heterodimer on IL-6 production was also tested (RPH-10 alone); ILlR-FcV-RAcP-FcII heterodimer dilution corresponding to a final concentration of 204.8 ng/ml was also included to demonstrate uniformity of the data between the two plates (RPHIO Plate 2); and
Figure 12 shows the results of the titration curve measurements for LIR-FcV-RAcP-FcII heterodimer experiment where MRC5 cells were incubated with IL- 1 β or IL- 1 β pre -mixed with various concentrations of LIR-FcV-RAcP-FcII heterodimer; IL-6 production was measured by Quantakine ELISA and the data was analyzed using 4-parameter fit algorithm; coefficient C at the resulting equation is numerically equal to an IC50 value of 0.3 ng/ml or about 2.4 pM.
DETAILED DESCRIPTION OF THE INVENTION
The teachings disclosed herein are based, in part, upon engineering of a heterodimeric protein assembly that is capable of binding to human IL-Ιβ and attenuating its function. The heterodimeric protein assembly of the present teachings comprises an extracellular portions of IL1-R1 (GenBank: AAM88423.1) and of IL-lRAcP (GenBank: BAA25421.1), or functional fragments thereof. Each, the IL1-R1 portion and the IL-lRAcP portion, is fused to a distinct mutant of Fc portion of the human Ig Gamma- 1 (GenBank: J00228.1). The two distinct Fc mutants in the heterodimeric protein assembly are engineered as to favor the heteromeric dimer formation between the two Fc mutants over any homomeric assembly. To enable recombinant production of the heterodimeric protein assembly of the present teachings, a DNA expression vector has been constructed for overproducing the heterodimeric protein assembly in a heterologous protein expression system, and mammalian cells have been prepared stably expressing the heterodimeric protein assembly to a high expression level. A protein purification procedure has been devised allowing obtaining a physiologically relevant substantially pure preparation of the heterodimeric protein assembly of the present teachings. Thus purified protein molecule demonstrates a high degree of specific activity in an in vitro Enzyme -Linked Immunosorbent Assay (ELISA) using human IL- 1β (GenBank: AAH08678.1). Unexpectedly, the protein molecule exhibits an acceptable pharmacokinetics profile upon subcutaneous animal administration, while not resulting in any body weight loss or adverse clinical events.
The terms used in this specification generally have their ordinary meanings in the art, within the context of this invention and in the specific context where each term is used. Certain terms are discussed below or elsewhere in the specification, to provide additional guidance to the
practitioner in describing the compositions and methods of the invention and how to make and use them. The scope or meaning of any use of a term will be apparent from the specific context in which the term is used. "About" and "approximately" shall generally mean an acceptable degree of error for the quantity measured given the nature or precision of the measurements. Typically, exemplary degrees of error are within 20 percent (%), preferably within 10%, and more preferably within 5% of a given value or range of values. Alternatively, and particularly in biological systems, the terms "about" and "approximately" may mean values that are within an order of magnitude, preferably within 5- fold and more preferably within 2-fold of a given value.
Numerical quantities given herein are approximate unless stated otherwise, meaning that the term "about" or "approximately" can be inferred when not expressly stated.
The methods of the invention may include steps of comparing sequences to each other, including wild-type sequence to one or more mutants (sequence variants). Such comparisons typically comprise alignments of polymer sequences, e.g., using sequence alignment programs and/or algorithms that are well known in the art (for example, BLAST, FASTA and MEGALIGN, to name a few). The skilled artisan can readily appreciate that, in such alignments, where a mutation contains a residue insertion or deletion, the sequence alignment will introduce a "gap" (typically represented by a dash, or "A") in the polymer sequence not containing the inserted or deleted residue.
The methods of the invention may include statistical calculations, e.g. determination of IC50 or EC50 values, etc.. The skilled artisan can readily appreciate that such can be performed using a variety of commercially available software, e.g. PRISM (GraphPad Software Inc, La Jolla, CA, USA) or similar. "Homologous," in all its grammatical forms and spelling variations, refers to the relationship between two proteins that possess a "common evolutionary origin," including proteins from superfamilies in the same species of organism, as well as homologous proteins from different species of organism. Such proteins (and their encoding nucleic acids) have sequence homology, as reflected by their sequence similarity, whether in terms of percent identity or by the presence of specific residues or motifs and conserved positions. However, in common usage and in the instant application, the term "homologous," when modified with an adverb such as "highly," may refer to sequence similarity and may or may not relate to a common evolutionary origin.
The term "sequence similarity," in all its grammatical forms, refers to the degree of identity or correspondence between nucleic acid or amino acid sequences that may or may not share a common evolutionary origin.
The terms "protein" and "polypeptide" are used interchangeably. The polypeptides described herein may be comprised of more than one contiguous amino acid chain, thus forming dimers or other oligomeric formations. In general, the polypetides of the present teachings for use in mammals are expressed in mammalian cells that allow for proper post-translational modifications, such as CHO or HEK293 cell lines, although other mammalian expression cell lines are expected to be useful as well. It is therefore anticipated that the polypeptides of the present teachings may be post-translationally modified without substantially effecting its biological function.
In certain aspects, functional variants of the heterodimeric protein assemblies of the present teachings include fusion proteins having at least a biologically active portion of the human IL1-R1 or IL-lRAcP or a functional fragment thereof, and one or more fusion domains. Well known examples of such fusion domains include, but are not limited to, polyhistidine, Glu-Glu, glutathione S transferase (GST), thioredoxin, protein A, protein G, an immunoglobulin heavy chain constant region (e.g., an Fc), maltose binding protein (MBP), or human serum albumin. A fusion domain may be selected so as to confer a desired property. For example, the IL1-R1 or IL- IPVACP polypeptide portions may be fused with a domain that stabilizes the IL1-R1 or IL-lRAcP polypeptides in vivo (a "stabilizer" domain), optionally via a suitable peptide linker. The term "stabilizing" means anything that increases the half life of a polypeptide in systemic circulation, regardless of whether this is because of decreased destruction, decreased clearance, or other pharmacokinetic effect. Fusions with the Fc portion of an immunoglobulin are known to confer desirable pharmacokinetic properties on certain proteins. Likewise, fusions to human serum albumin can confer desirable properties. Other types of fusion domains that may be selected include multimerizing (e.g., dimerizing, tetramerizing) domains and functional domains that confer an additional biological function, e.g. promoting accumulation at the targeted site of action in vivo.
In certain aspects, the heterodimeric protein assemblies of the present teachings comprise an extracellular portion of ILl-Rl, or a functional fragment thereof, fused with a IgG-Fc domain, and an extracellular portion IL-lRAcP, or a functional fragment thereof, fused with another IgG-Fc domain. The IgG-Fc domain and the another IgG-Fc domain are chosen as to favor a heterodimeric protein assembly over any homodimeric protein assembly. The extracellular portion of ILl-Rl may be fused with the IgG-Fc domain via a flexible linker, while IL-lRAcP, or a functional fragment thereof, may be fused with the another IgG-Fc domain via the flexible linker of the same amino acid sequence or via another flexible linker.
In an example embodiment, illustratively shown in Figure 1, the extracellular portion of ILl-Rl fused with IgG-Fc domain (Fc-II) via a flexible linker may comprise the amino acid sequence of SEQ. ID NO. 1, while IL-lRAcP fused with another IgG-Fc domain (Fc-V) via a flexible linker may comprise the amino acid sequence of SEQ. ID NO. 2. hILl-Rl-hlgGl-Fc polypeptide (SEQ ID NO. 1)
LEADKCKERE EKIILVSSAN EIDVRPCPLN PNEHKGTITW YKDDSKTPVS TEQASRIHQH 60
KEKLWFVPAK VEDSGHYYCV VRNSSYCLRI KISAKFVENE PNLCYNAQAI FKQKLPVAGD 120
GGLVCPYMEF FKNENNELPK LQWYKDCKPL LLDNIHFSGV KDRLIVMNVA EKHRGNYTCH 1 80
ASYTYLGKQY PITRVIEFIT LEENKPTRPV IVSPANETME VDLGSQIQLI CNVTGQLSDI 2 40
AYWKWNGSVI DEDDPVLGED YYSVENPANK RRSTLITVLN ISEIESRFYK HPFTCFAKNT 300
HGIDAAYIQL IYPVTNGSGG GDKTHTCPPC PAPELLGGPS VFLFPPKPKD TLMISRTPEV 3 60 TCWVDVSHE DPEVKFNWYV DGVEVHNAKT KPREEQYNST YRWSVLTVL HQDWLNGKEY 420
KCKVSNKALP APIEKTI SKA KGQPREPQVC TLPPSRDELT KNQVSLSCAV KGFYPSDIAV 4 80
EWESNGQPEN NYKTTPPVLD SDGSFKLVSK LTVDKSRWQQ GNVFSCSVMH EALHNHYTQK 540
SLSLSPGK 548 hIL-lRAcP-hlgGl-Fc polypeptide (SEQ ID NO. 2)
SERCDDWGLD TMRQIQVFED EPARIKCPLF EHFLKFNYST AHSAGLTLIW YWTRQDRDLE 60
EPINFRLPEN RISKEKDVLW FRPTLLNDTG NYTCMLRNTT YCSKVAFPLE VVQKDSCFNS 120 PMKLPVHKLY IEYGIQRITC PNVDGYFPSS VKPTITWYMG CYKIQNFNNV I PEGMNLSFL 1 80
IALISNNGNY TCVVTYPENG RTFHLTRTLT VKVVGSPK A VPPVIHSPND HWYEKEPGE 2 40
ELLI PCTVYF SFLMDSRNEV WWTIDGKKPD DITIDVTINE SISHSRTEDE TRTQILSIKK 300
VTSEDLKRSY VCHARSAKGE VAKAAKVKQK VPAPRYTVGS GGGDKTHTCP PCPAPELLGG 3 60 PSVFLFPPKP KDTLMISRTP EVTCWVDVS HEDPEVKFNW YVDGVEVHNA KTKPREEQYN 420
STYRWSVLT VLHQDWLNGK EYKCKVSNKA LPAPIEKTIS KAKGQPREPQ VYTLPPCRDE 4 80
LTKNQVSLWC LVKGFYPSDI AVEWESNGQP ENNYKTTPPV LDSDGSFFLY SALTVDKSRW 540
QQGNVFSCSV MHEALHNHYT QKSLSLSPGK 570
In certain aspects, the present teachings provides for a recombinant DNA molecule having an open reading frame coding for a polypeptide comprising the leading 333 amino acids of the human IL1-R1 fused with IgG-Fc domain (Fc-II) via a flexible linker, and for another recombinant DNA molecule having an open reading frame coding for another polypeptide comprising the leading 358 amino acids of the human IL-lRAcP fused with another IgG-Fc domain (Fc-V) via a flexible linker.
In an example embodiment, the polypeptide comprising the leading 333 amino acids of the human IL1-R1 fused with IgG-Fc domain (Fc-II) via a flexible linker comprises the amino acid sequence of SEQ. ID NO. 3. The corresponding to it DNA molecule may comprise the nucleotide sequence of SEQ ID NO. 4. The another polypeptide comprises the leading 358 amino acids of the human IL-lRAcP fused with another IgG-Fc domain (Fc-V) via a flexible linker may comprise the amino acid sequence of SEQ. ID NO. 5. The corresponding to it DNA molecule may comprise the nucleotide sequence of SEQ ID NO. 6. hILl-Rl-hlgGl-Fc polypeptide (SEQ ID NO. 3)
MKVLLRLICF IALLI SSLEA DKCKEREEKI ILVSSANEID VRPCPLNPNE HKGTITWYKD 60
DSKTPVSTEQ ASRIHQHKEK LWFVPAKVED SGHYYCVVRN SSYCLRIKIS AKFVENEPNL 120
CYNAQAIFKQ KLPVAGDGGL VCPYMEFFKN ENNELPKLQW YKDCKPLLLD NIHFSGVKDR 1 80
LIVMNVAEKH RGNYTCHASY TYLGKQYPIT RVIEFITLEE NKPTRPVIVS PANETMEVDL 2 40 GSQIQLICNV TGQLSDIAYW KWNGSVIDED DPVLGEDYYS VENPANKRRS TLITVLNISE 300
IESRFYKHPF TCFAKNTHGI DAAYIQLIYP VTNGSGGGDK THTCPPCPAP ELLGGPSVFL 3 60
FPPKPKDTLM ISRTPEVTCV WDVSHEDPE VKFNWYVDGV EVHNAKTKPR EEQYNSTYRV 420
VSVLTVLHQD WLNGKEYKCK VSNKALPAPI EKTISKAKGQ PREPQVCTLP PSRDELTKNQ 4 80 VSLSCAVKGF YPSDIAVEWE SNGQPENNYK TTPPVLDSDG SFKLVSKLTV DKSRWQQGNV 540
FSCSVMHEAL HNHYTQKSLS LSPGK 565
hILl-Rl-hlgGl-Fc DNA (SEQ ID NO. 4)
ATGAAGGTCC TGCTCAGGCT GATCTGCTTC ATTGCCCTGC TCATCAGCAG CCTGGAAGCC 60 GACAAGTGCA AGGAGAGGGA GGAGAAGATC ATCCTCGTCA GCTCCGCCAA CGAGATTGAT 120 GTCAGGCCCT GCCCCCTCAA CCCCAATGAG CACAAGGGCA CAATCACCTG GTACAAGGAC 180 GACAGCAAGA CCCCTGTCTC CACCGAGCAG GCCAGCAGAA TCCACCAGCA CAAAGAGAAG 240 CTGTGGTTCG TGCCTGCCAA GGTGGAAGAC AGCGGCCACT ACTACTGTGT GGTGAGGAAC 300 AGCTCCTACT GCCTCAGGAT CAAGATCTCC GCCAAGTTCG TGGAGAACGA GCCCAACCTC 360 TGTTACAACG CTCAGGCTAT TTTCAAGCAA AAGCTCCCCG TGGCTGGAGA CGGAGGCCTG 420 GTCTGTCCCT ACATGGAGTT CTTCAAGAAT GAGAATAATG AGCTCCCCAA GCTCCAGTGG 480 TACAAGGACT GTAAGCCTCT GCTCCTGGAC AACATCCACT TCTCCGGCGT GAAGGACAGA 540 CTGATCGTCA TGAACGTGGC CGAGAAGCAC AGGGGAAACT ACACCTGTCA CGCCTCCTAC 600 ACCTACCTCG GCAAGCAATA TCCCATCACC AGGGTCATCG AGTTCATCAC CCTCGAAGAG 660 AACAAGCCCA CAAGGCCTGT CATCGTCAGC CCCGCCAATG AAACCATGGA GGTGGACCTC 720 GGCAGCCAGA TCCAGCTGAT CTGCAACGTG ACAGGCCAGC TCAGCGACAT TGCCTACTGG 780 AAGTGGAACG GCTCCGTGAT CGACGAAGAT GATCCCGTGC TGGGCGAGGA CTACTATAGC 840 GTGGAGAACC CCGCCAACAA AAGAAGGAGC ACCCTGATCA CCGTGCTGAA CATCAGCGAG 900 ATCGAGTCCA GATTCTATAA GCATCCTTTC ACCTGCTTTG CCAAGAACAC CCACGGCATC 960 GACGCCGCTT ACATCCAGCT GATCTATCCC GTGACCAACG GATCCGGTGG AGGTGACAAA 1020 ACTCACACAT GCCCACCGTG CCCAGCTCCG GAACTCCTGG GCGGACCGTC AGTCTTCCTC 1080 TTCCCCCCAA AACCCAAGGA CACCCTCATG ATCTCCCGGA CCCCTGAGGT CACATGCGTG 1140 GTGGTGGACG TGAGCCACGA AGACCCTGAG GTCAAGTTCA ACTGGTACGT GGACGGCGTG 1200 GAGGTGCATA ATGCCAAGAC AAAGCCGCGG GAGGAGCAGT ACAACAGCAC GTACCGTGTG 1260 GTCAGCGTCC TCACCGTCCT GCACCAGGAC TGGCTGAATG GCAAGGAGTA CAAGTGCAAG 1320 GTCTCCAACA AAGCCCTCCC AGCCCCCATC GAGAAAACCA TCTCCAAAGC CAAAGGGCAG 1380 CCCCGAGAAC CACAGGTGTG TACCCTGCCC CCATCCCGGG ATGAGCTGAC CAAGAACCAG 1440 GTCAGCCTGA GTTGCGCGGT CAAAGGCTTC TATCCCAGCG ACATCGCCGT GGAGTGGGAG 1500 AGCAATGGGC AGCCGGAGAA CAACTACAAG ACCACGCCTC CCGTGTTGGA CTCCGACGGC 1560 TCCTTCAAGC TCGTCAGCAA GCTCACCGTG GACAAGAGCA GGTGGCAGCA GGGGAACGTC 1620 TTCTCATGCT CCGTGATGCA TGAGGCTCTG CACAACCACT ACACGCAGAA GAGCCTCTCC 1680 CTGTCTCCGG GTAAA hIL-lRAcP-hlgGl-Fc polypeptide (SEQ ID NO. 5)
MTLLWCWSL YFYGILQSDA SERCDDWGLD TMRQIQVFED EPARIKCPLF EHFLKFNYST 60
AHSAGLTLIW YWTRQDRDLE EPINFRLPEN RISKEKDVLW FRPTLLNDTG NYTCMLRNTT 120
YCSKVAFPLE VVQKDSCFNS PMKLPVHKLY IEYGIQRITC PNVDGYFPSS VKPTITWYMG 180
CYKIQNFNNV I PEGMNLSFL IALISNNGNY TCVVTYPENG RTFHLTRTLT VKVVGSPKNA 240
VPPVIHSPND HWYEKEPGE ELLI PCTVYF SFLMDSRNEV WWTI DGKKPD DITIDVTINE 300
SISHSRTEDE TRTQILSIKK VTSEDLKRSY VCHARSAKGE VAKAAKVKQK VPAPRYTVGS 360
GGGDKTHTCP PCPAPELLGG PSVFLFPPKP KDTLMISRTP EVTCWVDVS HEDPEVKFNW 420
YVDGVEVHNA KTKPREEQYN STYRWSVLT VLHQDWLNGK EYKCKVSNKA LPAPIEKTIS 480
KAKGQPREPQ VYTLPPCRDE LTKNQVSLWC LVKGFYPSDI AVEWESNGQP ENNYKTTPPV 540
LDSDGSFFLY SALTVDKSRW QQGNVFSCSV MHEALHNHYT QKSLSLSPGK 590 hIL-lRAcP-hlgGl-Fc DNA (SEQ ID NO. 6)
ATGACTCTGC TGTGGTGCGT CGTGTCCCTC TACTTCTACG GCATCCTCCA GTCCGACGCC 60
AGCGAGAGGT GCGATGACTG GGGCCTGGAC ACCATGAGGC AGATCCAGGT GTTTGAGGAC 120
GAGCCTGCCA GGATTAAGTG CCCCCTCTTC GAGCACTTTC TGAAGTTCAA CTACAGCACC 180
GCTCACAGCG CTGGCCTGAC ACTGATCTGG TACTGGACAA GGCAGGACAG GGATCTCGAG 240
GAGCCCATCA ACTTCAGGCT GCCCGAAAAC AGAATCAGCA AGGAGAAGGA CGTGCTGTGG 300
TTCAGACCCA CCCTCCTCAA CGACACAGGC AACTACACCT GCATGCTCAG GAACACCACC 360
TACTGCAGCA AGGTGGCCTT CCCTCTCGAG GTGGTCCAGA AGGACAGCTG CTTCAACAGC 420
CCCATGAAGC TGCCCGTCCA TAAACTGTAC ATCGAGTACG GCATCCAGAG GATCACATGC 480
CCCAACGTGG ACGGCTACTT CCCCAGCTCC GTGAAGCCCA CCATCACATG GTACATGGGC 540
TGTTACAAAA TCCAGAACTT TAACAACGTC ATCCCCGAGG GCATGAATCT GTCCTTCCTG 600
ATCGCCCTGA TCAGCAACAA CGGCAATTAC ACCTGCGTCG TGACCTACCC CGAAAACGGC 660
AGGACCTTCC ACCTGACCAG GACCCTGACC GTGAAAGTCG TGGGAAGCCC CAAGAATGCC 720
GTGCCCCCCG TGATCCATTC CCCCAACGAC CACGTGGTGT ACGAGAAGGA GCCTGGAGAG 780
GAGCTGCTGA TCCCCTGCAC AGTGTACTTC TCCTTCCTGA TGGACTCCAG GAATGAAGTG 840
TGGTGGACCA TCGACGGCAA GAAGCCTGAC GACATCACCA TCGATGTGAC CATCAACGAG 900
AGCATCAGCC ACAGCAGGAC CGAGGACGAG ACCAGGACCC AGATCCTGAG CATCAAGAAA 960
GTCACCAGCG AGGACCTCAA GAGAAGCTAC GTCTGTCACG CCAGAAGCGC CAAAGGCGAG 1020 GTGGCCAAGG CTGCTAAGGT GAAACAGAAA GTGCCCGCTC CTAGGTACAC AGTCGGATCC 1080
GGTGGAGGTG ACAAAACTCA CACATGCCCA CCGTGCCCAG CTCCGGAACT CCTGGGCGGA 1140
CCGTCAGTCT TCCTCTTCCC CCCAAAACCC AAGGACACCC TCATGATCTC CCGGACCCCT 1200
GAGGTCACAT GCGTGGTGGT GGACGTGAGC CACGAAGACC CTGAGGTCAA GTTCAACTGG 1260
TACGTGGACG GCGTGGAGGT GCATAATGCC AAGACAAAGC CGCGGGAGGA GCAGTACAAC 1320
AGCACGTACC GTGTGGTCAG CGTCCTCACC GTCCTGCACC AGGACTGGCT GAATGGCAAG 1380
GAGTACAAGT GCAAGGTCTC CAACAAAGCC CTCCCAGCCC CCATCGAGAA AACCATCTCC 1440
AAAGCCAAAG GGCAGCCCCG AGAACCACAG GTGTACACCC TGCCCCCATG TCGGGATGAG 1500
CTGACCAAGA ACCAGGTCAG CCTGTGGTGC CTGGTCAAAG GCTTCTATCC CAGCGACATC 1560
GCCGTGGAGT GGGAGAGCAA TGGGCAGCCG GAGAACAACT ACAAGACCAC GCCTCCCGTG 1620
TTGGACTCCG ACGGCTCCTT CTTCCTCTAC AGCGCGCTCA CCGTGGACAA GAGCAGGTGG 1680
CAGCAGGGGA ACGTCTTCTC ATGCTCCGTG ATGCATGAGG CTCTGCACAA CCACTACACG 1740
CAGAAGAGCC TCTCCCTGTC TCCGGGTAAA 1770 In certain aspects, the present invention provides for a recombinant mamalian expression plasmid for high expression of a polypeptide comprising the leading 333 amino acids of the human ILl-Rl fused with IgG-Fc domain (Fc-II) via a flexible linker, and for another recombinant DNA molecule having an open reading frame coding for another polypeptide comprising the leading 358 amino acids of the human IL-lRAcP fused with another IgG-Fc domain (Fc-V) via a flexible linker. This plasmid comprises two cytomegalovirus (CMV) promoters to drive transcription of the two genes coding for said polypeptide and said another polypeptide, each followed by a transcription termination sequence and a polyadenylation sequence. The plasmid also contains an origin of replication and a gene conferring ampicillin resistance, for supporting plasmid propagation and selection in bacteria. The plasmid further contains a gene for Glutamine synthetase, a selectable marker widely used for establishing stable CHOK1 and NSO cell lines. The plasmid of the present invention is illustratively shown in Figure 2.
In an example embodiment, the mammalian expression plasmid of the present teachings comprises the nucleotide sequence of SEQ ID NO. 7. hILl-Rl-hlgGl-Fc-II/ IL-lRAcP- hlgGl-Fc-V expression plasmid (SEQ ID NO. 7)
AGCTTGCCAC CATGAAGGTC CTGCTCAGGC TGATCTGCTT CATTGCCCTG CTCATCAGCA 60 GCCTGGAAGC CGACAAGTGC AAGGAGAGGG AGGAGAAGAT CATCCTCGTC AGCTCCGCCA 120 ACGAGATTGA TGTCAGGCCC TGCCCCCTCA ACCCCAATGA GCACAAGGGC ACAATCACCT 180
GGTACAAGGA CGACAGCAAG ACCCCTGTCT CCACCGAGCA GGCCAGCAGA ATCCACCAGC 240
ACAAAGAGAA GCTGTGGTTC GTGCCTGCCA AGGTGGAAGA CAGCGGCCAC TACTACTGTG 300
TGGTGAGGAA CAGCTCCTAC TGCCTCAGGA TCAAGATCTC CGCCAAGTTC GTGGAGAACG 360
AGCCCAACCT CTGTTACAAC GCTCAGGCTA TTTTCAAGCA AAAGCTCCCC GTGGCTGGAG 420
ACGGAGGCCT GGTCTGTCCC TACATGGAGT TCTTCAAGAA TGAGAATAAT GAGCTCCCCA 480
AGCTCCAGTG GTACAAGGAC TGTAAGCCTC TGCTCCTGGA CAACATCCAC TTCTCCGGCG 540
TGAAGGACAG ACTGATCGTC ATGAACGTGG CCGAGAAGCA CAGGGGAAAC TACACCTGTC 600
ACGCCTCCTA CACCTACCTC GGCAAGCAAT ATCCCATCAC CAGGGTCATC GAGTTCATCA 660
CCCTCGAAGA GAACAAGCCC ACAAGGCCTG TCATCGTCAG CCCCGCCAAT GAAACCATGG 720
AGGTGGACCT CGGCAGCCAG ATCCAGCTGA TCTGCAACGT GACAGGCCAG CTCAGCGACA 780
TTGCCTACTG GAAGTGGAAC GGCTCCGTGA TCGACGAAGA TGATCCCGTG CTGGGCGAGG 840
ACTACTATAG CGTGGAGAAC CCCGCCAACA AAAGAAGGAG CACCCTGATC ACCGTGCTGA 900
ACATCAGCGA GATCGAGTCC AGATTCTATA AGCATCCTTT CACCTGCTTT GCCAAGAACA 960
CCCACGGCAT CGACGCCGCT TACATCCAGC TGATCTATCC CGTGACCAAC GGATCCGGTG 1020
GAGGTGACAA AACTCACACA TGCCCACCGT GCCCAGCTCC GGAACTCCTG GGCGGACCGT 1080
CAGTCTTCCT CTTCCCCCCA AAACCCAAGG ACACCCTCAT GATCTCCCGG ACCCCTGAGG 1140
TCACATGCGT GGTGGTGGAC GTGAGCCACG AAGACCCTGA GGTCAAGTTC AACTGGTACG 1200
TGGACGGCGT GGAGGTGCAT AATGCCAAGA CAAAGCCGCG GGAGGAGCAG TACAACAGCA 1260
CGTACCGTGT GGTCAGCGTC CTCACCGTCC TGCACCAGGA CTGGCTGAAT GGCAAGGAGT 1320
ACAAGTGCAA GGTCTCCAAC AAAGCCCTCC CAGCCCCCAT CGAGAAAACC ATCTCCAAAG 1380
CCAAAGGGCA GCCCCGAGAA CCACAGGTGT GTACCCTGCC CCCATCCCGG GATGAGCTGA 1440
CCAAGAACCA GGTCAGCCTG AGTTGCGCGG TCAAAGGCTT CTATCCCAGC GACATCGCCG 1500
TGGAGTGGGA GAGCAATGGG CAGCCGGAGA ACAACTACAA GACCACGCCT CCCGTGTTGG 1560
ACTCCGACGG CTCCTTCAAG CTCGTCAGCA AGCTCACCGT GGACAAGAGC AGGTGGCAGC 1620
AGGGGAACGT CTTCTCATGC TCCGTGATGC ATGAGGCTCT GCACAACCAC TACACGCAGA 1680
AGAGCCTCTC CCTGTCTCCG GGTAAATAAT AGAATTCATT GATCATAATC AGCCATACCA 1740
CATTTGTAGA GGTTTTACTT GCTTTAAAAA ACCTCCCACA CCTCCCCCTG AACCTGAAAC 1800
ATAAAATGAA TGCAATTGTT GTTGTTAACT TGTTTATTGC AGCTTATAAT GGTTACAAAT 1860
AAAGCAATAG CATCACAAAT TTCACAAATA AAGCATTTTT TTCACTGCAT TCTAGTTGTG 1920
GTTTGTCCAA ACTCATCAAT GTATCTTATC ATGTCTGGCG GCCGCCGATA TTTGAAAATA 1980
TGGCATATTG AAAATGTCGC CGATGTGAGT TTCTGTGTAA CTGATATCGC CATTTTTCCA 2040
AAAGTGATTT TTGGGCATAC GCGATATCTG GCGATAGCGC TTATATCGTT TACGGGGGAT 2100
GGCGATAGAC GACTTTGGTG ACTTGGGCGA TTCTGTGTGT CGCAAATATC GCAGTTTCGA 2160
TATAGGTGAC AGACGATATG AGGCTATATC GCCGATAGAG GCGACATCAA GCTGGCACAT 2220 GGCCAATGCA TATCGATCTA TACATTGAAT CAATATTGGC CATTAGCCAT ATTATTCATT 2280
GGTTATATAG CATAAATCAA TATTGGCTAT TGGCCATTGC ATACGTTGTA TCCATATCAT 2340
AATATGTACA TTTATATTGG CTCATGTCCA ACATTACCGC CATGTTGACA TTGATTATTG 2400
ACTAGTTATT AATAGTAATC AATTACGGGG TCATTAGTTC ATAGCCCATA TATGGAGTTC 2460
CGCGTTACAT AACTTACGGT AAATGGCCCG CCTGGCTGAC CGCCCAACGA CCCCCGCCCA 2520
TTGACGTCAA TAATGACGTA TGTTCCCATA GTAACGCCAA TAGGGACTTT CCATTGACGT 2580
CAATGGGTGG AGTATTTACG GTAAACTGCC CACTTGGCAG TACATCAAGT GTATCATATG 2640
CCAAGTACGC CCCCTATTGA CGTCAATGAC GGTAAATGGC CCGCCTGGCA TTATGCCCAG 2700
TACATGACCT TATGGGACTT TCCTACTTGG CAGTACATCT ACGTATTAGT CATCGCTATT 2760
ACCATGGTGA TGCGGTTTTG GCAGTACATC AATGGGCGTG GATAGCGGTT TGACTCACGG 2820
GGATTTCCAA GTCTCCACCC CATTGACGTC AATGGGAGTT TGTTTTGGCA CCAAAATCAA 2880
CGGGACTTTC CAAAATGTCG TAACAACTCC GCCCCATTGA CGCAAATGGG CGGTAGGCGT 2940
GTACGGTGGG AGGTCTATAT AAGCAGAGCT CGTTTAGTGA ACCGTCAGAT CGCCTGGAGA 3000
CGCCATCCAC GCTGTTTTGA CCTCCATAGA AGACACCGGG ACCGATCCAG CCTCCGCGGC 3060
CGGGAACGGT GCATTGGAAC GCGGATTCCC CGTGCCAAGA GTGACGTAAG TACCGCCTAT 3120
AGAGTCTATA GGCCCACCCC CTTGGCTTCT TATGCATGCT ATACTGTTTT TGGCTTGGGG 3180
TCTATACACC CCCGCTTCCT CATGTTATAG GTGATGGTAT AGCTTAGCCT ATAGGTGTGG 3240
GTTATTGACC ATTATTGACC ACTCCCCTAT TGGTGACGAT ACTTTCCATT ACTAATCCAT 3300
AACATGGCTC TTTGCCACAA CTCTCTTTAT TGGCTATATG CCAATACACT GTCCTTCAGA 3360
GACTGACACG GACTCTGTAT TTTTACAGGA TGGGGTCTCA TTTATTATTT ACAAATTCAC 3420
ATATACAACA CCACCGTCCC CAGTGCCCGC AGTTTTTATT AAACATAACG TGGGATCTCC 3480
ACGCGAATCT CGGGTACGTG TTCCGGACAT GGGCTCTTCT CCGGTAGCGG CGGAGCTTCT 3540
ACATCCGAGC CCTGCTCCCA TGCCTCCAGC GACTCATGGT CGCTCGGCAG CTCCTTGCTC 3600
CTAACAGTGG AGGCCAGACT TAGGCACAGC ACGATGCCCA CCACCACCAG TGTGCCGCAC 3660
AAGGCCGTGG CGGTAGGGTA TGTGTCTGAA AATGAGCTCG GGGAGCGGGC TTGCACCGCT 3720
GACGCATTTG GAAGACTTAA GGCAGCGGCA GAAGAAGATG CAGGCAGCTG AGTTGTTGTG 3780
TTCTGATAAG AGTCAGAGGT AACTCCCGTT GCGGTGCTGT TAACGGTGGA GGGCAGTGTA 3840
GTCTGAGCAG TACTCGTTGC TGCCGCGCGC GCCACCAGAC ATAATAGCTG ACAGACTAAC 3900
AGACTGTTCC TTTCCATGGG TCTTTTCTGC AGTCACCGTC CTTGACACGA AGCTTGCCAC 3960
CATGACTCTG CTGTGGTGCG TCGTGTCCCT CTACTTCTAC GGCATCCTCC AGTCCGACGC 4020
CAGCGAGAGG TGCGATGACT GGGGCCTGGA CACCATGAGG CAGATCCAGG TGTTTGAGGA 4080
CGAGCCTGCC AGGATTAAGT GCCCCCTCTT CGAGCACTTT CTGAAGTTCA ACTACAGCAC 4140
CGCTCACAGC GCTGGCCTGA CACTGATCTG GTACTGGACA AGGCAGGACA GGGATCTCGA 4200
GGAGCCCATC AACTTCAGGC TGCCCGAAAA CAGAATCAGC AAGGAGAAGG ACGTGCTGTG 4260
GTTCAGACCC ACCCTCCTCA ACGACACAGG CAACTACACC TGCATGCTCA GGAACACCAC 4320 CTACTGCAGC AAGGTGGCCT TCCCTCTCGA GGTGGTCCAG AAGGACAGCT GCTTCAACAG 4380
CCCCATGAAG CTGCCCGTCC ATAAACTGTA CATCGAGTAC GGCATCCAGA GGATCACATG 4440
CCCCAACGTG GACGGCTACT TCCCCAGCTC CGTGAAGCCC ACCATCACAT GGTACATGGG 4500
CTGTTACAAA ATCCAGAACT TTAACAACGT CATCCCCGAG GGCATGAATC TGTCCTTCCT 4560
GATCGCCCTG ATCAGCAACA ACGGCAATTA CACCTGCGTC GTGACCTACC CCGAAAACGG 4620
CAGGACCTTC CACCTGACCA GGACCCTGAC CGTGAAAGTC GTGGGAAGCC CCAAGAATGC 4680
CGTGCCCCCC GTGATCCATT CCCCCAACGA CCACGTGGTG TACGAGAAGG AGCCTGGAGA 4740
GGAGCTGCTG ATCCCCTGCA CAGTGTACTT CTCCTTCCTG ATGGACTCCA GGAATGAAGT 4800
GTGGTGGACC ATCGACGGCA AGAAGCCTGA CGACATCACC ATCGATGTGA CCATCAACGA 4860
GAGCATCAGC CACAGCAGGA CCGAGGACGA GACCAGGACC CAGATCCTGA GCATCAAGAA 4920
AGTCACCAGC GAGGACCTCA AGAGAAGCTA CGTCTGTCAC GCCAGAAGCG CCAAAGGCGA 4980
GGTGGCCAAG GCTGCTAAGG TGAAACAGAA AGTGCCCGCT CCTAGGTACA CAGTCGGATC 5040
CGGTGGAGGT GACAAAACTC ACACATGCCC ACCGTGCCCA GCTCCGGAAC TCCTGGGCGG 5100
ACCGTCAGTC TTCCTCTTCC CCCCAAAACC CAAGGACACC CTCATGATCT CCCGGACCCC 5160
TGAGGTCACA TGCGTGGTGG TGGACGTGAG CCACGAAGAC CCTGAGGTCA AGTTCAACTG 5220
GTACGTGGAC GGCGTGGAGG TGCATAATGC CAAGACAAAG CCGCGGGAGG AGCAGTACAA 5280
CAGCACGTAC CGTGTGGTCA GCGTCCTCAC CGTCCTGCAC CAGGACTGGC TGAATGGCAA 5340
GGAGTACAAG TGCAAGGTCT CCAACAAAGC CCTCCCAGCC CCCATCGAGA AAACCATCTC 5400
CAAAGCCAAA GGGCAGCCCC GAGAACCACA GGTGTACACC CTGCCCCCAT GTCGGGATGA 5460
GCTGACCAAG AACCAGGTCA GCCTGTGGTG CCTGGTCAAA GGCTTCTATC CCAGCGACAT 5520
CGCCGTGGAG TGGGAGAGCA ATGGGCAGCC GGAGAACAAC TACAAGACCA CGCCTCCCGT 5580
GTTGGACTCC GACGGCTCCT TCTTCCTCTA CAGCGCGCTC ACCGTGGACA AGAGCAGGTG 5640
GCAGCAGGGG AACGTCTTCT CATGCTCCGT GATGCATGAG GCTCTGCACA ACCACTACAC 5700
GCAGAAGAGC CTCTCCCTGT CTCCGGGTAA ATAATAGAAT TCATTGATCA TAATCAGCCA 5760
TACCACATTT GTAGAGGTTT TACTTGCTTT AAAAAACCTC CCACACCTCC CCCTGAACCT 5820
GAAACATAAA ATGAATGCAA TTGTTGTTGT TAACTTGTTT ATTGCAGCTT ATAATGGTTA 5880
CAAATAAAGC AATAGCATCA CAAATTTCAC AAATAAAGCA TTTTTTTCAC TGCATTCTAG 5940
TTGTGGTTTG TCCAAACTCA TCAATGTATC TTATCATGTC TGGATCCTCT ACGCCGGACG 6000
CATCGTGGCC GGCATCACCG GCGCCACAGG TGCGGTTGCT GGCGCCTATA TCGCCGACAT 6060
CACCGATGGG GAAGATCGGG CTCGCCACTT CGGGCTCATG AGCGCTTGTT TCGGCGTGGG 6120
TATGGTGGCA GGCCCCGTGG CCGGGGGACT GTTGGGCGCC ATCTCCTTGC ATGCACCATT 6180
CCTTGCGGCG GCGGTGCTCA ACGGCCTCAA CCTACTACTG GGCTGCTTCC TAATGCAGGA 6240
GTCGCATAAG GGAGAGCGTC GACCTCGGGC CGCGTTGCTG GCGTTTTTCC ATAGGCTCCG 6300
CCCCCCTGAC GAGCATCACA AAAATCGACG CTCAAGTCAG AGGTGGCGAA ACCCGACAGG 6360
ACTATAAAGA TACCAGGCGT TTCCCCCTGG AAGCTCCCTC GTGCGCTCTC CTGTTCCGAC 6420 CCTGCCGCTT ACCGGATACC TGTCCGCCTT TCTCCCTTCG GGAAGCGTGG CGCTTTCTCA 6480
TAGCTCACGC TGTAGGTATC TCAGTTCGGT GTAGGTCGTT CGCTCCAAGC TGGGCTGTGT 6540
GCACGAACCC CCCGTTCAGC CCGACCGCTG CGCCTTATCC GGTAACTATC GTCTTGAGTC 6600
CAACCCGGTA AGACACGACT TATCGCCACT GGCAGCAGCC ACTGGTAACA GGATTAGCAG 6660
AGCGAGGTAT GTAGGCGGTG CTACAGAGTT CTTGAAGTGG TGGCCTAACT ACGGCTACAC 6720
TAGAAGAACA GTATTTGGTA TCTGCGCTCT GCTGAAGCCA GTTACCTTCG GAAAAAGAGT 6780
TGGTAGCTCT TGATCCGGCA AACAAACCAC CGCTGGTAGC GGTGGTTTTT TTGTTTGCAA 6840
GCAGCAGATT ACGCGCAGAA AAAAAGGATC TCAAGAAGAT CCTTTGATCT TTTCTACGGG 6900
GTCTGACGCT CAGTGGAACG AAAACTCACG TTAAGGGATT TTGGTCATGA GATTATCAAA 6960
AAGGATCTTC ACCTAGATCC TTTTAAATTA AAAATGAAGT TTTAAATCAA TCTAAAGTAT 7020
ATATGAGTAA ACTTGGTCTG ACAGTTACCA ATGCTTAATC AGTGAGGCAC CTATCTCAGC 7080
GATCTGTCTA TTTCGTTCAT CCATAGTTGC CTGACTCCCC GTCGTGTAGA TAACTACGAT 7140
ACGGGAGGGC TTACCATCTG GCCCCAGTGC TGCAATGATA CCGCGAGACC CACGCTCACC 7200
GGCTCCAGAT TTATCAGCAA TAAACCAGCC AGCCGGAAGG GCCGAGCGCA GAAGTGGTCC 7260
TGCAACTTTA TCCGCCTCCA TCCAGTCTAT TAATTGTTGC CGGGAAGCTA GAGTAAGTAG 7320
TTCGCCAGTT AATAGTTTGC GCAACGTTGT TGCCATTGCT ACAGGCATCG TGGTGTCACG 7380
CTCGTCGTTT GGTATGGCTT CATTCAGCTC CGGTTCCCAA CGATCAAGGC GAGTTACATG 7440
ATCCCCCATG TTGTGCAAAA AAGCGGTTAG CTCCTTCGGT CCTCCGATCG TTGTCAGAAG 7500
TAAGTTGGCC GCAGTGTTAT CACTCATGGT TATGGCAGCA CTGCATAATT CTCTTACTGT 7560
CATGCCATCC GTAAGATGCT TTTCTGTGAC TGGTGAGTAC TCAACCAAGT CATTCTGAGA 7620
ATAGTGTATG CGGCGACCGA GTTGCTCTTG CCCGGCGTCA ATACGGGATA ATACCGCGCC 7680
ACATAGCAGA ACTTTAAAAG TGCTCATCAT TGGAAAACGT TCTTCGGGGC GAAAACTCTC 7740
AAGGATCTTA CCGCTGTTGA GATCCAGTTC GATGTAACCC ACTCGTGCAC CCAACTGATC 7800
TTCAGCATCT TTTACTTTCA CCAGCGTTTC TGGGTGAGCA AAAACAGGAA GGCAAAATGC 7860
CGCAAAAAAG GGAATAAGGG CGACACGGAA ATGTTGAATA CTCATACTCT TCCTTTTTCA 7920
ATATTATTGA AGCATTTATC AGGGTTATTG TCTCATGAGC GGATACATAT TTGAATGTAT 7980
TTAGAAAAAT AAACAAATAG GGGTTCCGCG CACATTTCCC CGAAAAGTGC CACCTGACGT 8040
CTAAGAAACC ATTATTATCA TGACATTAAC CTATAAAAAT AGGCGTATCA CGAGGCCCTG 8100
ATGGCTCTTT GCGGCACCCA TCGTTCGTAA TGTTCCGTGG CACCGAGGAC AACCCTCAAG 8160
AGAAAATGTA ATCACACTGG CTCACCTTCG GGTGGGCCTT TCTGCGTTTA TAAGGAGACA 8220
CTTTATGTTT AAGAAGGTTG GTAAATTCCT TGCGGCTTTG GCAGCCAAGC TAGATCCGGC 8280
TGTGGAATGT GTGTCAGTTA GGGTGTGGAA AGTCCCCAGG CTCCCCAGCA GGCAGAAGTA 8340
TGCAAAGCAT GCATCTCAAT TAGTCAGCAA CCAGGTGTGG AAAGTCCCCA GGCTCCCCAG 8400
CAGGCAGAAG TATGCAAAGC ATGCATCTCA ATTAGTCAGC AACCATAGTC CCGCCCCTAA 8460
CTCCGCCCAT CCCGCCCCTA ACTCCGCCCA GTTCCGCCCA TTCTCCGCCC CATGGCTGAC 8520 ΤΑΑΤΤΤΤΤΤΤ TATTTATGCA GAGGCCGAGG CCGCCTCGGC CTCTGAGCTA TTCCAGAAGT 8580
AGTGAGGAGG CTTTTTTGGA GGCCTAGGCT TTTGCAAAAA GCTAGCTTGG GGCCACCGCT 8640
CAGAGCACCT TCCACCATGG CCACCTCAGC AAGTTCCCAC TTGAACAAAA ACATCAAGCA 8700
AATGTACTTG TGCCTGCCCC AGGGTGAGAA AGTCCAAGCC ATGTATATCT GGGTTGATGG 8760
TACTGGAGAA GGACTGCGCT GCAAAACCCG CACCCTGGAC TGTGAGCCCA AGTGTGTAGA 8820
AGAGTTACCT GAGTGGAATT TTGATGGCTC TAGTACCTTT CAGTCTGAGG GCTCCAACAG 8880
TGACATGTAT CTCAGCCCTG TTGCCATGTT TCGGGACCCC TTCCGCAGAG ATCCCAACAA 8940
GCTGGTGTTC TGTGAAGTTT TCAAGTACAA CCGGAAGCCT GCAGAGACCA ATTTAAGGCA 9000
CTCGTGTAAA CGGATAATGG ACATGGTGAG CAACCAGCAC CCCTGGTTTG GAATGGAACA 9060
GGAGTATACT CTGATGGGAA CAGATGGGCA CCCTTTTGGT TGGCCTTCCA ATGGCTTTCC 9120
TGGGCCCCAA GGTCCGTATT ACTGTGGTGT GGGCGCAGAC AAAGCCTATG GCAGGGATAT 9180
CGTGGAGGCT CACTACCGCG CCTGCTTGTA TGCTGGGGTC AAGATTACAG GAACAAATGC 9240
TGAGGTCATG CCTGCCCAGT GGGAACTCCA AATAGGACCC TGTGAAGGAA TCCGCATGGG 9300
AGATCATCTC TGGGTGGCCC GTTTCATCTT GCATCGAGTA TGTGAAGACT TTGGGGTAAT 9360
AGCAACCTTT GACCCCAAGC CCATTCCTGG GAACTGGAAT GGTGCAGGCT GCCATACCAA 9420
CTTTAGCACC AAGGCCATGC GGGAGGAGAA TGGTCTGAAG CACATCGAGG AGGCCATCGA 9480
GAAACTAAGC AAGCGGCACC GGTACCACAT TCGAGCCTAC GATCCCAAGG GGGGCCTGGA 9540
CAATGCCCGT GGTCTGACTG GGTTCCACGA AACGTCCAAC ATCAACGACT TTTCTGCTGG 9600
TGTCGCCAAT CGCAGTGCCA GCATCCGCAT TCCCCGGACT GTCGGCCAGG AGAAGAAAGG 9660
TTACTTTGAA GACCGCGGCC CCTCTGCCAA TTGTGACCCC TTTGCAGTGA CAGAAGCCAT 9720
CGTCCGCACA TGCCTTCTCA ATGAGACTGG CGACGAGCCC TTCCAATACA AAAACTAATT 9780
AGACTTTGAG TGATCTTGAG CCTTTCCTAG TTCATCCCAC CCCGCCCCAG AGAGATCTTT 9840
GTGAAGGAAC CTTACTTCTG TGGTGTGACA TAATTGGACA AACTACCTAC AGAGATTTAA 9900
AGCTCTAAGG TAAATATAAA ATTTTTAAGT GTATAATGTG TTAAACTACT GATTCTAATT 9960
GTTTGTGTAT TTTAGATTCC AACCTATGGA ACTGATGAAT GGGAGCAGTG GTGGAATGCC 10020
TTTAATGAGG AAAACCTGTT TTGCTCAGAA GAAATGCCAT CTAGTGATGA TGAGGCTACT 10080
GCTGACTCTC AACATTCTAC TCCTCCAAAA AAGAAGAGAA AGGTAGAAGA CCCCAAGGAC 10140
TTTCCTTCAG AATTGCTAAG TTTTTTGAGT CATGCTGTGT TTAGTAATAG AACTCTTGCT 10200
TGCTTTGCTA TTTACACCAC AAAGGAAAAA GCTGCACTGC TATACAAGAA AATTATGGAA 10260
AAATATTCTG TAACCTTTAT AAGTAGGCAT AACAGTTATA ATCATAACAT ACTGTTTTTT 10320
CTTACTCCAC ACAGGCATAG AGTGTCTGCT ATTAATAACT ATGCTCAAAA ATTGTGTACC 10380
TTTAGCTTTT TAATTTGTAA AGGGGTTAAT AAGGAATATT TGATGTATAG TGCCTTGACT 10440
AG AG AT CAT A ATCAGCCATA CCACATTTGT AGAGGTTTTA CTTGCTTTAA AAAACCTCCC 10500
ACACCTCCCC CTGAACCTGA AACATAAAAT GAATGCAATT GTTGTTGTTA ACTTGTTTAT 10560
TGCAGCTTAT AATGGTTACA AATAAAGCAA TAGCATCACA AATTTCACAA ATAAAGCATT 10620 TTTTTCACTG CATTCTAGTT GTGGTTTGTC CAAACTCATC AATGTATCTT ATCATGTCTG 10680
GATCTAGCTT CGTGTCAAGG ACGGTGACTG CAGTGAATAA TAAAATGTGT GTTTGTCCGA 10740
AATACGCGTT TTGAGATTTC TGTCGCCGAC TAAATTCATG TCGCGCGATA GTGGTGTTTA 10800
TCGCCGATAG AGATGGCGAT ATTGGAAAAA TCGATATTTG AAAATATGGC ATATTGAAAA 10860
TGTCGCCGAT GTGAGTTTCT GTGTAACTGA TATCGCCATT TTTCCAAAAG TGATTTTTGG 10920
GCATACGCGA TATCTGGCGA TAGCGCTTAT ATCGTTTACG GGGGATGGCG ATAGACGACT 10980
TTGGTGACTT GGGCGATTCT GTGTGTCGCA AATATCGCAG TTTCGATATA GGTGACAGAC 11040
GATATGAGGC TATATCGCCG ATAGAGGCGA CATCAAGCTG GCACATGGCC AATGCATATC 11100
GATCTATACA TTGAATCAAT ATTGGCCATT AGCCATATTA TTCATTGGTT ATATAGCATA 11160
AATCAATATT GGCTATTGGC CATTGCATAC GTTGTATCCA TATCATAATA TGTACATTTA 11220
TATTGGCTCA TGTCCAACAT TACCGCCATG TTGACATTGA TTATTGACTA GTTATTAATA 11280
GTAATCAATT ACGGGGTCAT TAGTTCATAG CCCATATATG GAGTTCCGCG TTACATAACT 11340
TACGGTAAAT GGCCCGCCTG GCTGACCGCC CAACGACCCC CGCCCATTGA CGTCAATAAT 11400
GACGTATGTT CCCATAGTAA CGCCAATAGG GACTTTCCAT TGACGTCAAT GGGTGGAGTA 11460
TTTACGGTAA ACTGCCCACT TGGCAGTACA TCAAGTGTAT CATATGCCAA GTACGCCCCC 11520
TATTGACGTC AATGACGGTA AATGGCCCGC CTGGCATTAT GCCCAGTACA TGACCTTATG 11580
GGACTTTCCT ACTTGGCAGT ACATCTACGT ATTAGTCATC GCTATTACCA TGGTGATGCG 11640
GTTTTGGCAG TACATCAATG GGCGTGGATA GCGGTTTGAC TCACGGGGAT TTCCAAGTCT 11700
CCACCCCATT GACGTCAATG GGAGTTTGTT TTGGCACCAA AATCAACGGG ACTTTCCAAA 11760
ATGTCGTAAC AACTCCGCCC CATTGACGCA AATGGGCGGT AGGCGTGTAC GGTGGGAGGT 11820
CTATATAAGC AGAGCTCGTT TAGTGAACCG TCAGATCGCC TGGAGACGCC ATCCACGCTG 11880
TTTTGACCTC CATAGAAGAC ACCGGGACCG ATCCAGCCTC CGCGGCCGGG AACGGTGCAT 11940
TGGAACGCGG ATTCCCCGTG CCAAGAGTGA CGTAAGTACC GCCTATAGAG TCTATAGGCC 12000
CACCCCCTTG GCTTCTTATG CATGCTATAC TGTTTTTGGC TTGGGGTCTA TACACCCCCG 12060
CTTCCTCATG TTATAGGTGA TGGTATAGCT TAGCCTATAG GTGTGGGTTA TTGACCATTA 12120
TTGACCACTC CCCTATTGGT GACGATACTT TCCATTACTA ATCCATAACA TGGCTCTTTG 12180
CCACAACTCT CTTTATTGGC TATATGCCAA TACACTGTCC TTCAGAGACT GACACGGACT 12240
CTGTATTTTT ACAGGATGGG GTCTCATTTA TTATTTACAA ATTCACATAT ACAACACCAC 12300
CGTCCCCAGT GCCCGCAGTT TTTATTAAAC ATAACGTGGG ATCTCCACGC GAATCTCGGG 12360
TACGTGTTCC GGACATGGGC TCTTCTCCGG TAGCGGCGGA GCTTCTACAT CCGAGCCCTG 12420
CTCCCATGCC TCCAGCGACT CATGGTCGCT CGGCAGCTCC TTGCTCCTAA CAGTGGAGGC 12480
CAGACTTAGG CACAGCACGA TGCCCACCAC CACCAGTGTG CCGCACAAGG CCGTGGCGGT 12540
AGGGTATGTG TCTGAAAATG AGCTCGGGGA GCGGGCTTGC ACCGCTGACG CATTTGGAAG 12600
ACTTAAGGCA GCGGCAGAAG AAGATGCAGG CAGCTGAGTT GTTGTGTTCT GATAAGAGTC 12660
AGAGGTAACT CCCGTTGCGG TGCTGTTAAC GGTGGAGGGC AGTGTAGTCT GAGCAGTACT 12720 CGTTGCTGCC GCGCGCGCCA CCAGACATAA TAGCTGACAG ACTAACAGAC TGTTCCTTTC 12780 CATGGGTCTT TTCTGCAGTC ACCGTCCTTG ACACGA 12816
In certain aspects, the present teachings provide for a mammalian expression system for production of a heterodimeric protein assembly comprising a polypeptide comprising amino acid residues 18 through 333 of the human IL1-R1 fused with IgG-Fc domain (Fc-II) via a flexible linker, and another polypeptide comprising amino acid residues 21 through 358 of the human IL- lRAcP fused with another IgG-Fc domain (Fc-V) via a flexible linker.
In an example embodiment, the mammalian expression system of the present invention comprises Chinese hamster ovary cells (CHO-K1) harboring a plasmid comprising nucleotide sequence of SEQ ID NO. 7.
In certain aspects, the present teachings provide for a method of treatment of a mammal effected by the following disorders associated with IL-Ιβ modulation: arthritis, a gout, a rheumatoid arthritis, a Cryopyrin-Associated Periodic Syndromes (CAPS), a scleroderma, a diabetes, a atherosclerosis, a dry eye disease, an ocular allergy, or an uveitis.
EXAMPLES
The following Examples illustrate the forgoing aspects and other aspects of the present teachings. These non-limiting Examples are put forth so as to provide those of ordinary skill in the art with illustrative embodiments as to how the compounds, compositions, articles, devices, and/or methods claimed herein are made and evaluated. The Examples are intended to be purely exemplary of the inventions disclosed herein and are not intended to limit the scope of what the inventors regard as their invention. Efforts have been made to ensure accuracy with respect to numbers (e.g., amounts, temperature, etc.) but some errors and deviations should be accounted for.
Example 1: Construction of plasmids for expression of polypeptides of the present invention.
Optimized gene sequences comprising sequences encoding residues amino acid residues 1 through 333 of IL-1R1 or residues 1 through 358 of IL-lRAcP were chemically synthesized. The resulting fragments were cloned in-frame into the recipient intermediate vectors comprising sequences coding for Fc-V or Fc-II, via Hindlll and BspEI restriction sites. Obtained positive clones were verified by DNA sequencing. The resulting constructions were termed PKN001 '-IL- lR-FcV and PKNOOl '-RAcP-FcII, respectively.
The gene encoding IL-lRl-Fc-V was then cloned into a second intermediate vector, termed PKN002, via Hindlll and EcoRI restriction sites. Positive clones were screened by double digestion and the insertion sequence of the correct plasmids was verified by DNA sequencing. The resulting constructions were termed PKN002-IL-lR-FcV.
Finally, the expression cassette for RAcP-FcII from PKN001 '-RAcP-FcII was integrated into PKN002-IL-lR-FcV plasmid via Notl and Sail restriction sites and yielded a new
recombinant construction that contained expression elements for both RAcP-FcII and IL-lR-FcV. The resulting clones were screened by Notl and Sail double digestion followed by DNA gel electrophoresis, the correct clones were exhibiting one band migrating approximately as an 8kb DNA fragment and another band migrating approximately as a 4kb DNA fragment. The final plasmid was termed PKN012-ILlR-FcV-RAcP-FcII.
Recombinant plasmid PKN012-ILlR-FcV-RAcP-FcII combines expression cassettes for both RAcP-FcII and IL-lR-FcV. The plasmid can be used for co-expressing RAcP-FcII and IL- lR-FcV proteins in a 1 to 1 ratio under the control of a CMV promoter. The majority of these two fusion proteins would then form ILlR-FcV/RAcP-FcII heterodimers after secretion. The plasmid also expresses Glutamine synthetase (GS) protein via a SV40 promoter, which can be used as a selection marker to generate stable cell lines for ILlR-FcV/RAcP-FcII heterodimer production. The plasmid map for PKN012-ILlR-FcV-RAcP-FcII is illustratively shown in Figure 2.
Example 2: Generation of stable cell lines for expressing polypeptides of the present invention.
A stable clone of CHO-K1 cells co-expressing hILl-Rl-hlgGl-Fc polypeptide of SEQ ID NO. 1 and hIL-lRAcP-hlgGl-Fc polypeptide of SEQ ID NO. 2 has been generated through standard cell biology protocols. The expression plasmid PKN012-ILlR-FcV-RAcP-FcII described in Example 1 was used for generating stable cell lines for high expression of said polypeptides. Expression levels of said polypeptides in a plurality of clones were about or over 100 mg/L in a 7-day batch culture. One clonal cell line showed expression levels of about or over 300 mg/L in shake flask batch culture.
Materials
Chinese hamster ovary cells (CHO-K1) were obtained as frozen stocks from ATCC (CCL-
61™). The cells were adapted in house into a CD CHO media. Media and reagent were obtained from commercial source. Upon full adaptation, the cells were grown to high density for a few passages. The resulting cells were subcloned. One of the resulting clones with a doubling time under 20 hrs and good morphology was selected as parental cell line.
Methods
CHO-K1 cells at passage 4 were cultured in CD-CHO chemically defined media (Invitrogen) containing 6 mM L-Glutamine. The cells were maintained by 1 :3 splits after reaching a cell density of 4xl06 cells/ml. Cells were span down by centrifugation and resuspended into 1 ml of CD-CHO chemically defined media (Invitrogen).
The following electroporation protocol was utilized:
40 ug of plasmid DNA in 100 ul sterilized TE buffer was used for an electroporation; on the day of trans fection the viability of cells was at least 95%;
cells were centrifuged at 800rpm for 5 min; the super was removed and the cells were resuspended in 10 ml CD CHO media and centrifuged again;
- the super was removed and the cells were resuspended in a small vol. of CD CHO media to 1.43xl07 cells/ml;
- 0.7 ml cells (107 cells) were added to the DNA and mixed gently by pipetting, avoiding generating bubbles;
- cells were immediately electroporated by delivering a single pulse of 300 volts, 900 uF to each cuvette;
50 ml CD CHO (without L-glutamine) was immediately added to the elecroporated cells and mixed gently;
- the cell suspension was distributed into ten 96 well plates at 50 ul/well; the following day, 150 ul of CD CHO containing 33.3 uM MSX was added to each well. A number of transfections were carried out in CHO-K1 cells in the process of generation of potential ILlR-FcV-RAcP-FcII expression cell line; a representative transfection growth curve is shown in Figure 3 and the data is shown in Table 1. Protein expression levels were analyzed by SDS-PAGE. Cell lines with high levels of protein overexpression exhibit a strong band with an apparent molecular weight of about 180 kDa. Based on the preliminary analysis four clones were selected for further analysis, where expression levels were further assessed by SDS-PAGE and ELISA. Well expressing clones were inoculated into 125 ml shake flasks, the cells were expanded and frozen.
The chosen highest production cell line was selected and thawed into CD-CHO. Growth curves on the cell line were assessed, and samples were collected daily for cell count, cell viability and ILlR-FcV-RAcP-FcII productivity. Based on these studies, it was determined that selected highest productivity cell line was expressing ILlR-FcV-RAcP-FcII heterodimer in CD-CHO media in amounts necessary to support commercial production. The yield of the heterodimer after purification was at least about 300 mg/L (without any production optimization).
Table 1: A Representative Clonal Cell Line Growth curve
Culture period Cell density
Feeding (DAY) (106 cells/ml)
0 0.5
1 1.09
2 1.93
3 2.97 +10% feed B
5 4.13
6 4.55 +10% feed B
7 4.6
8 5.46 +10% feed B
9 4.86
10 2.4
11 1 Example 3: Purification of polypeptides of the present invention. hILl-Rl-hlgGl-Fc polypeptide of SEQ ID NO. 1 and hIL-lRAcP-hlgGl-Fc polypeptide of SEQ ID NO. 2 were co-expressed in CHO-K1 essentially as described in foregoing Example 2. Cells were harvested and lysed utilizing well established protocols. After cell lysate clarification, the supernatant at protein concentration of about 0.4 mg/ml, containing expressed hlLl-Rl-hlgGl- Fc/ hIL-lRAcP-hlgGl-Fc polypeptides, was applied to a Protein A affinity column. The affinity purification step was carried out according to the procedure outlined in Table 2. The Protein A eluate containing hILl-Rl-hlgGl-Fc/ hIL-lRAcP-hlgGl-Fc at pH of about 3.5 - 3.7 is incubated for 45-60 minutes to inactivate potentially existing in the contaminating materials.
After material incubation for about 45 minutes at pH 3.6 at room temperature its pH is adjusted to about 7.9-8.1 with 2 M Tris-HCl pH 9.5 (~ 3.5% v/v of the diluted Pro A eluate). The low pH treated Protein A eluate is pH-adjusted to pH of about 8.0 using 1 M Tris-HCl, pH 9.0. The conductivity is adjusted to about 5.5 mS/cm with deionized water (dH20) if needed.
In order to reduce the contents of DNA, HCP, endotoxin and potential viral contaminants, the pH adjusted Protein A column eluate was further purified by anion-exchange chromatography (AIEX) utilizing Q Sepharose resin. The AIEX step is operated according to a step-elution procedure outlined in Table 3. Pooled elution peak fractions are concentrated by microfiltration to a concentration of about 20 mg/ml, followed by addition of a 20% Sucrose stock solution to a final Sucrose concentration of about 1% (w/v) and freezing at -80°C.
A sample of thus purified protein was analyzed by size-exclusion HPLC (SEC-HPLC) and SDS-PAGE (reducing and non-reducing). The results of the analysis are presented in Figure 4 and Figure 5, respectively. In Figure 5, lanes 1, 3 show hILl-Rl-hlgGl-Fc/ hIL-lRAcP-hlgGl-Fc SDS-PAGE under non-reducing conditions, while lanes 4, 6 - under reducing conditions. Lanes 2, 5 show molecular weight markers. hILl-Rl-hlgGl-Fc/ hIL-lRAcP-hlgGl-Fc heterodimer has an apparent molecular weight of about 180 kDa, consisting of two di-sulfide linked monomers, each of an apparent molecular weight of about 90 kDa.
The SEC-HPLC operational procedure is outlined in Table 4. Loading sample is diluted with mobile phase to reach protein concentration of about 5 mg/ml. The biologically relevant form of hILl-Rl-hlgGl-Fc/ hIL-lRAcP-hlgGl-Fc heterodimer is represented by the major peak with retention time RT=14.979 min.
Table 2: The operational procedure of Protein A Affinity Chromatography
Step Buffer Vol Flow
CV cm/h
Rinse Before-use dH20 3 150
Equilibration lO mM NaPh, pH 6.0 5 150
Sample Load Cell harvest - 150
Wash 1 lO mM NaPh, pH 6.0 3 150
25 mM NaPh, 0.5 M NaCl, 5%
Wash 2 5 150
Isopropanol pH 7.0
Re-Equilibration lO mM NaPh, pH 6.0 3 150
Elution 20 mM Na-Citrate, pH 3.4 4 150
Re-Equilibration lO mM PB, pH 6.0 3 150
0.1 M NaOH (contact time 15 min),
CIP 3 100 reversed flow, CIP every 5 cycles
Rinse with NaCl lM NaCl 3 150
Rinse After-use dH20 3 150
Storage 20% (v/v) Ethanol, 20 mM NaPh, pH 7.0 3 150
Table 3: The operational procedure of AIEX
Step Buffer Vol Flow
CV cm/h
Rinse Before-use dH20 3 150
Recharge 10 mM Tris-HCl, 1 M NaCl, pH 8 3 150
Equilibration 10 mM Tris-HCl, 50 mM NaCl, pH 8 3 150
Sample Load Prepared Q Load - 150
Wash 10 mM Tris-HCl, 50 mM NaCl, pH 8 3 150
Elution 10 mM Tris-HCl, 0.35 M NaCl, pH 8.0 3 150
CIP 1 M NaOH (contact time 1 hr), 3 40 reversed-flow
Regeneration 10 mM Tris-HCl, 1 M NaCl, pH 8.0 3 150
Rinse After-use dH20 3 150
Storage 20% (v/v) Ethanol 3 150 Table 4: The Operational Procedure of SEC-HPLC
Mobile Phase: 20 mM phosphate, 300 mM NaCl, pH 7.4
Flow Rate: 0.5 mL/min
Column: G2000 SWxl, 7.8mmx300mm, TOSOH Bioscience
Guard column: TSK Guard SWxl,6.0mmx40mm, TOSOH Bioscience
Column Temperature: 25 °C
Sampler temperature: Rome temperature
Injection Volume: 10 μΐ
Detector Wavelength: 280 nm
Run Time: 30 min
The activity of thus purified sample, which was expressed and purified essentially as described in this example, was tested in a standard ELISA assay using commercially available human IL-Ι β (PrimGene, Shanghai, China; Cat#: 101-OlB). A typical binding curve obtained in the assay is shown in Figure 6. Based on the curve analysis, the calculated EC50 value is about 50 ng/ml.
Example 4: Characterization of inhibition of IL-i -induced IL-6 production with IL1R- FcV-RAcP-FcII heterodimer in human cells.
The present Example demonstrates potent functional (inhibitory) properties of ILlR-FcV- RAcP-FcII heterodimer in targeting human IL-Ιβ. As a functional comparator, previously characterized mouse monoclonal antibodies against IL-Ιβ were used. Human lung fibroblasts MRC5 produce IL-6 in response to treatment with IL-Ι β. Quantification of IL-^-induced IL-6 production in MRC5 cells was used as the assay functional output. Inhibition of IL-6 production by ILlR-FcV-RAcP-FcII heterodimer and by control anti IL-Ιβ antibody provides a quantitative measure of inhibitory properties of the ILlR-FcV-RAcP-FcII heterodimer preparation. Polypeptides of ILlR-FcV-RAcP-FcII heterodimer (SEQ ID NO. l and SEQ ID NO. 2) were co-expressed and purified essentially as described in the forgoing examples. The protein concentration of substantially pure ILlR-FcV-RAcP-FcII heterodimer in the sample used was 0.9 mg/ml in 100 mM NaCl, 25 mM NaH2P04/Na2HP04, 25 mM Arginine hydrochloride, 1% Sucrose; pH=6.3.
The following materials were used in the assay:
Cells: MRC5 cells, Human Lung Fibroblasts, ATCC Cat # CCL-171, Lot # 59474707. Medium: DMEM, Dulbecco's Modification of Eagle's Medium, high glucose (4.5 g/L), Mediatech, Cat # 10-017-CM, Lot # 10017204, supplemented with L-glutamine and lx penn/strep and 10% Fetal Bovine Serum, Omega Scientific, Cat # FB-05, Lot # 105104 Reagents: IL-Ιβ, Human recombinant, E. coli-derived, Alal l7-Ser269, Accession # NP 000567, R&D systems, Cat # 201-LB, Lot # ADM1411062; Monoclonal antibodies against human IL-Ιβ, clone #8516, R&D systems, Cat # MAB201, Lot # AWE1011081; Human IL-6 Quantakine Immunoassay, R&D systems, Cat # D6050, Lot # 300070.
The following procedures were utilized in the assay:
Cell Maintenance:
1. Propagate MRC5 cells in the DMEM containing 10% FBS on T75 flasks according to the manufacturer's recommendations. Record the passage number;
2. Trypsinize cells, re-suspend in DMEM containing 10%> FBS;
3. Test cell viability using Guava ViaCount Reagent. Standard viability should be >85%>, if less, do not use this batch of cells;
4. Prepare dilutions of the cells for plating at desired density of 2 x 103 cells/well per well of 48-well plate or 5 x 103 per ml;
5. Allow cells to attach for 16-24 hours at 37°C, 5%> C02, then remove medium;
6. Replace with fresh DMEM medium supplemented with 10% FBS, 0.4 ml/well for 48-well plate, incubate 5-10 min and replace the medium again with the same volume; 7. Add to the wells IL- 1 β, Test Substances pre-mixed with IL- 1 β and appropriate controls;
8. Incubate the cells after treatment for 24 hours then harvest the supernatants;
9. Centrifuge the supernatants at 300 x g for 10 min, collect cleared supernatants and use them for ELISA either directly or with 1/3 dilution if appropriate;
ELISA Protocol:
Principle of the assay: the assay employs the quantitative sandwich enzyme immunoassay technique. A microplate is pre-coated with a monoclonal antibody specific for IL-6.
Standards and samples are pipetted into the wells of the microplate and any IL-6 present binds to the immobilized antibodies. After washing away any unbound substances, an enzyme-linked polyclonal antibody specific for IL-6 is added to the wells. Following a wash to remove any unbound antibody-enzyme reagent, a substrate solution is added to the wells and color develops in proportion to the amount of bound IL-6. The color
development is stopped and the color intensity is measured.
Assay procedure:
1. Prepare all reagents and working standards as described in manufacturer's manual (http://www.rndsystems.com product_results.aspx?k=D6050)
2. Remove excess microplate strips from the plate frame, return them to the foil pouch containing the desiccant pack, and reseal.
3. Add 100 of Assay Diluent RD 1 W to each well.
4. Add 100 of Standard, sample, or control per well. Cover with the adhesive strip provided. Incubate for 2 hours at room temperature. A plate layout is provided to record standards and samples assayed.
5. Aspirate each well and wash, repeating the process three times for a total of four washes. Wash by filling each well with Wash Buffer (400 μί) using a squirt bottle, manifold dispenser, or autowasher. Complete removal of liquid at each step is essential to good performance. After the last wash, remove any remaining Wash Buffer by aspirating or decanting. Invert the plate and blot it against clean paper towels.
6. Add 200 μΐ, of IL-6 Conjugate to each well. Cover with a new adhesive strip.
Incubate for 2 hours at room temperature. 7. Repeat the aspiration/wash as in step 5.
8. Add 200 of Substrate Solution to each well. Incubate for 20 minutes at room temperature. Protect from light.
9. Add 50 μΐ, of Stop Solution to each well. The color in the wells should change from blue to yellow. If the color in the wells is green or the color change does not appear uniform, gently tap the plate to ensure thorough mixing.
10. Determine the optical density of each well within 30 minutes, using a microplate reader set to 450 nm. If wavelength correction is available, set to 540 nm or 570 nm. If wavelength correction is not available, subtract readings at 540 nm or 570 nm from the readings at 450 nm. This subtraction will correct for optical imperfections in the plate. Readings made directly at 450 nm without correction may be higher and less accurate.
Following are the experimental data obtained:
Inhibition ofIL-6 production by control anti-IL-1 β antibodies:
1. MRC5 cells (Passage 5) were plated into a 48-well plate (Costar, Cat # 3548) at 2 x 103 cells per well, 0.4 ml per well, and incubated for 24 hours;
2. On the day of the assay, medium from the wells was aspirated and replaced with 0.4 ml of DMEM supplemented with 10% FBS, incubated 10-15 min and replaced with the same amount of the same fresh medium again;
3. IL-Ιβ was used at a final concentration of 50 pg/ml throughout the experiment;
4. Anti-human IL-Ιβ antibodies were used at final concentrations of 0.192, 0.96, 4.8, 24, 120 and 600 ng/ml;
5. Both IL-Ιβ and anti-human IL-Ιβ antibodies were prepared as lOx concentrated solutions;
6. Prior to adding the test substances to the cells, 55 μΐ of lOx IL-Ιβ was mixed with 55 μΐ of lOx corresponding anti-human IL-Ιβ antibody solution (or appropriate control) and incubated at room temperature for 30 min;
7. After the incubation, 100 μΐ of each mixture was added to the wells containing 0.4 ml of growth medium; 8. The cells were incubated in the presence of the test substances for 24 hours and supernatants were collected, centrifuged at 300 x g for 10 min and used for IL-6 ELISA assay;
9. To test for recovery of IL-6 in the presence of anti-IL-Ιβ antibody, the latter was used at a final concentration of 600 ng/ml and were spiked with IL-6 standard to a final concentration of 20 pg/ml.
The calibration curve obtained in the IL-1 antibody mediated inhibition of IL-6 production assay is shown in Figure 7. The results of the measurements of IL-i -mediated IL-6 production and of IL-6 recovery from the culture medium in the experiment are shown in Figure 8 and
Figure 9. The obtained numerical values for IL-6 production along with their standard deviations are presented in Table 5.
Table 5: Summary of IL-6 production values.
Figure imgf000032_0001
The foregoing experiment revealed that: (1) the control anti-IL-Ιβ antibody is a very potent inhibitor of IL-Ιβ signaling pathway with an IC50 of about 2.1 ng/ml or about 14 pM; (2) plating cell density of 2 x 103 cells per well 24 hours prior to the assay setup is adequate; (3) recovery of IL-6 spiked into the culture medium at a final concentration of 20 pg/ml is about 114%. Inhibition of IL-6 production by ILlR-FcV-RAcP-FcII heterodimer preparation:
1. MRC5 cells (Passage 6) were plated into 48-well plate (Costar, Cat # 3548) at 2 x 103 cells per well, 0.4 ml per well and incubated for 24 hours;
2. Plating for this experiment was carried out in duplicates. Each plating duplicate was measured in duplicates on ELISA plate resulting in 4 experimental points per treatment
3. On the day of the assay, medium from the wells was aspirated and replaced with 0.4 ml of DMEM supplemented with 10% FBS, incubated 10-15 min and replaced with the same amount of the same fresh medium again;
4. IL-Ιβ was used at a final concentration of 50 pg/ml throughout the experiment;
5. ILlR-FcV-RAcP-FcII heterodimer was used at final concentrations of 0.0536, 0.134, 0.335, 0.839, 2.097, 5.24,13.1, 32.8, 81.9, 204.8, 512, 1280, 3200 and 20000 ng/ml;
6. Both IL-Ιβ and ILlR-FcV-RAcP-FcII heterodimer was prepared as lOx concentrated solutions;
7. Prior to adding the test substances to the cells, 120 μΐ of lOx IL-i /IL-lF2 was mixed with 120 μΐ of lOx corresponding ILlR-FcV-RAcP-FcII heterodimer solution (or appropriate control) and incubated at room temperature for 30 min;
8. After the incubation, 100 μΐ of each mixture was added to the wells containing 0.4 ml of growth medium;
9. The cells were incubated in the presence of the test substances for 24 hours and supernatants were collected, centrifuged at 300 x g for 10 min and used for IL-6 ELISA assay.
The calibration curve obtained in the ILlR-FcV-RAcP-FcII heterodimer mediated inhibition of IL-6 production assay is shown in Figure 10. The results of the measurements of ILlR-FcV- RAcP-FcII heterodimer -mediated IL-6 production and of IL-6 recovery from the culture medium in the experiment are shown in Figure 11 and Figure 12. The obtained numerical values for IL-6 production along with their standard deviations are presented in Table 6. Table 6: Summary of IL-6 production values.
Figure imgf000034_0001
R - indicates that the value fell outside the assay calibration range Each experimental point was plated in duplicate and IL-6 production was further measured in duplicate resulting in 4 experimental reads per concentration point. The obtained data
demonstrated high reproducibility of the experimental procedure and the assay. The foregoing experiment revealed that the IC50 value for ILlR-FcV-RAcP-FcII heterodimer in this experiment was about 0.3 mg/ml or about 2.4 pM.
Example 5: Pharmacokinetics (PK) of ILlR-FcV-RAcP-FcII heterodimer after subcutaneous administration in mice.
Polypeptides of ILlR-FcV-RAcP-FcII heterodimer (SEQ ID NO. l and SEQ ID NO. 2) were co-expressed and purified essentially as described in the forgoing examples. For administration into animals, the polypeptides were formulated in the following buffer: 1% w/v Sucrose, lOOmM Sodium Chloride, 20 mM L-Arginine Hydrochloride, 25 mM Sodium Bicarbonate, pH 6.3. The dosing stock concentration used was 0.5 mg/mL of the polypeptide.
Fourteen female Balb/c nu/nu mice were randomized based on body weight into seven groups of two animals on Day 0 of the study. A single treatment of the polypeptides (5 mg/kg) was administered subcutaneously (dorsal) on Day 0 to all groups except mice in Group 1 , which were bled via cardiac puncture for plasma preparation on Day 0 of the study. Blood samples were collected from mice via the orbital sinus in the remaining groups at various times throughout the study for preparation of plasma.
Body weights were recorded for all animals on the treatment day (Day 0) and then three times per week, including the termination day of each group.
Groups of mice were culled at specific time points for plasma preparation. Body weight changes were not measured in groups culled for sample collection at 0 hours and within 36 hours of dose administration. All other mice gained body weight and no adverse clinical signs were reported during the study period.
Following the in-life phase of the study, plasma samples were analyzed by ELISA for Hu-Fc proteins. Quantification of Hu-Fc in mouse plasma samples by ELISA was used as a read-out for circulating levels of the polypeptides. The assay was performed on samples from all mice in the study. The polypeptides were detected in the plasma of animals at 1 hour post-administration. One Phase Decay Model equation using Prism 5.0c (GraphPad Software Inc, La Jolla, CA, USA) was then used to determine pharmacokinetics of the polypeptides as detected by Hu-Fc ELISA. Peak circulating level of Hu-Fc (Cmax) was determined to be 1.65 μg/mL, and time to peak circulating levels (Tmax) was 36 hours post treatment. The half-life (Tl/2) was 88.15 hours and the rate constant (K) was 0.0079 hr-1. Hu-Fc levels were negligible in the plasma of the untreated Group 1 animals. The results of the study are summarized in Table 7.
Table 7: Mean Human-Fc Protein Concentration ± SEM ^g/mL) at each Time Post- Administration
Bleeding Schedule Mean Human-Fc
Group Treatment (time post- Protein Concentration SEM
administration) ^g/mL]
1 No treatment 0 hours _*
0.00
2 30 minutes 0.02 0.01
3 1 hour 0.12 0.00
4 2 hours 0.24 0.09
5 4 hours 0.76 0.03
6 8 hours 1.1 1 0.10
polypeptide of SEQ
7 10 hours
IDs NO. 1 and NO. 1.53 0.08
2 2 (5 mg/kg, Once 24 hours" 1.47 0.14
only, s.c.)
3 36 hours" 1.65 0.1 1
4 96 hours* 1.27 0.01
5 7 days" 0.43 0.01
6 14 days" 0.13 0.07
7 21 days" 0.06 0.04 *SEM unable to be calculated as level of Hu-Fc was below detectable limit of ELISA for one of samples in group.
The Human-Fc Protein Concentration was determined by Prism Software based on the mean absorbance of the triplicate samples
Bleed via orbital sinus
Bleed via terminal cardiac puncture
Example 6: Evaluation of the toxicity of ILlR-FcV-RAcP-FcII heterodimer following 28- days repeated dosing in C57BL6 mice. Following is a repeat dose murine toxicity evaluation study results summary. Treatment of
C57BL6 mice with polypeptides of TLlR-FcV-RAcP-FcII heterodimer (SEQ ID NO.l and SEQ ID NO. 2) test article by twice weekly subcutaneous injection at 10, 30 and 100 mg/kg dose levels was well tolerated in the study. There were no unscheduled deaths prior to completion of the study. Test article treatment was not associated with morbidity or clinical signs of toxicity. There was no consistent gender or treatment related effect on body weight. There was a finding in increased food consumption which was considered to be a possible effect of treatment but this was not considered to be adverse.
Although there was evidence of functional changes in parameters related to clinical biochemistry, these were not considered to be of toxicological significance as there was evidence of reversibility in most of these parameters, although not complete in some. In the absence of toxicokinetic evaluation it was not possible to assess the significance of apparent non-reversibility of some of these differences. However, none of the differences was associated with histopathological change or was considered to be adverse. The differences observed were of a type that is typically reversible and it is possible that the reversibility period was no sufficient to demonstrate this.
It was concluded that the No-Observed Adverse Effect Level (NOAEL) in the mouse 28-day ILlR-FcV-RAcP-FcII heterodimer dosing study was therefore considered to be 100 mg/kg.
All publications and patents mentioned herein are hereby incorporated by reference in their entirety as if each individual publication or patent was specifically and individually indicated to be incorporated by reference. While specific embodiments of the subject matter have been discussed, the above specification is illustrative and not restrictive. Many variations will become apparent to those skilled in the art upon review of this specification and the claims below. The full scope of the invention should be determined by reference to the claims, along with their full scope of equivalents, and the specification, along with such variations.

Claims

at is claimed is:
A heterodimeric protein composition capable of binding human IL-Ιβ, said protein composition comprising:
a first polypeptide comprising
a first amino acid sequence comprising amino acids 18 through 333 of human IL 1 -R 1 , and
a second amino acid sequence comprising a first mutant of a Fc portion of human immunoglobulin gamma- 1 Fc;
a second polypeptide comprising
another first amino acid sequence comprising amino acids 21 through 358 of human ILl-RAcP, and
another second amino acid sequence comprising a second mutant of the Fc portion of human immunoglobulin gamma- 1 Fc; and
wherein said first and second mutants are selected as to favor heterodimeric assembly between said first and second mutants over any homodimeric assembly.
The protein composition of claim 1 , wherein said protein composition exhibits human IL- 1β binding activity in an ELISA assay with an EC50 of about 50 ng/ml.
The protein composition of claim 1 , said first polypeptide comprises amino acid sequence of SEQ ID NO. 1 and said second polypeptide comprises amino acid sequence of SEQ ID NO. 2.
A therapeutic composition, the composition comprising a heterodimeric protein
composition capable of binding human IL-Ιβ, said heterodimeric protein composition comprising: a first polypeptide comprising
a first amino acid sequence comprising amino acids 18 through 333 of human IL 1 -R 1 , and a second amino acid sequence comprising a first mutant of a Fc portion of human immunoglobulin gamma- 1 Fc;
a second polypeptide comprising
another first amino acid sequence comprising amino acids 21 through 358 of human ILl-RAcP, and
another second amino acid sequence comprising a second mutant of the Fc portion of human immunoglobulin gamma- 1 Fc; and
wherein said first and second mutants are selected as to favor heterodimeric assembly between said first and second mutants over any homodimeric assembly.
5. The therapeutic composition of claim 4, wherein half-life of said heterodimeric protein composition in systemic circulation in mice after a subcutaneous administration at a dose of 5 mg/kg is at least about 88 hours, as assayed by human Fc ELISA.
6. A therapeutic composition, the composition comprising a heterodimeric protein
composition comprising a first polypeptide comprising amino acid sequence of SEQ ID NO. 1 and a second polypeptide comprising amino acid sequence of SEQ ID NO. 2.
7. An isolated nucleic acid encoding a polypeptide comprising amino acid sequence of SEQ ID NO. 3.
8. An isolated nucleic acid encoding a polypeptide comprising amino acid sequence of SEQ ID NO. 4.
9. The nucleic acid of claims 7 or 8, wherein the codon usage is optimized for high expression of said polypeptide in a mammalian cell.
10. The nucleic acid of claim 7, wherein the nucleic acid sequence comprises the sequence of SEQ ID NO. 5.
11. The nucleic acid of claim 8, wherein the nucleic acid sequence comprises the sequence of SEQ ID NO. 6.
12. The nucleic acid of claims 10 or 11, wherein said nucleic acid comprises an expression vector.
13. An isolated nucleic acid of SEQ ID NO. 7.
14. A heterologous expression system, the expression system harboring an expression vector comprising a nucleic acid sequence encoding a first polypeptide comprising amino acid sequence of SEQ ID NO. 3 and another nucleic acid sequence encoding a second polypeptide comprising amino acid sequence of SEQ ID NO. 4.
15. The expression system of claim 14, wherein said expression vector is harbored in a
mammalian cell.
16. The expression system of claim 15, wherein said mammalian cell is a CHO cell.
17. The expression system of claim 15, wherein said expression system is capable of
expressing a heterodimeric protein comprising a first polypeptide comprising amino acid sequence of SEQ ID NO. 1 and a second polypeptide comprising amino acid sequence of SEQ ID NO. 2, and wherein the level of expression of said heterodimeric protein is at least 300 mg per liter of cell culture.
18. Use of a substance for manufacture of a medicament for the treatment or prevention of a disease associated with modulation of activity of human IL-Ιβ , the substance comprising a heterodimeric protein comprising a first polypeptide comprising amino acid sequence of SEQ ID NO. 1 and a second polypeptide comprising amino acid sequence of SEQ ID NO. 2.
19. The use according to claim 18, wherein said disease is an arthritis.
20. The use according to claim 18, wherein said disease is a gout.
21. The use according to claim 18, wherein said disease is a rheumatoid arthritis.
22. The use according to claim 18, wherein said disease is a Cryopyrin- Associated Periodic Syndromes (CAPS).
23. The use according to claim 18, wherein said disease is a scleroderma.
24. The use according to claim 18, wherein said disease is a diabetes.
25. The use according to claim 18, wherein said disease is atherosclerosis.
26. The use according to claim 18, wherein said disease is a dry eye disease. 27. The use according to claim 18, wherein said disease is an ocular allergy.
28. The use according to claim 18, wherein said disease is an uveitis.
29. A method of treating or preventing a disease or condition associated with modulation of activity of human IL- 1 β, the method comprising administering to a patient in need for treating or preventing a disease associated with modulation of activity of human IL-Ιβ a therapeutically effective amount of a pharmaceutical composition comprising a heterodimeric protein comprising a first polypeptide comprising amino acid sequence of SEQ ID NO. 1 and a second polypeptide comprising amino acid sequence of SEQ ID NO. 2.
30. The method according to claim 29, wherein said disease is an arthritis.
31. The method according to claim 29, wherein said disease is a gout. 32. The method according to claim 29, wherein said disease is a rheumatoid arthritis.
33. The method according to claim 29, wherein said disease is a Cryopyrin- Associated
Periodic Syndromes (CAPS)
The method according claim 29, wherein said disease is a scleroderma.
35. The method according to claim 29, wherein said disease is a diabetes.
36. The method according to claim 29, wherein said disease is atherosclerosis.
37. The method according to claim 29, wherein said disease is a dry eye disease.
38. The method according to claim 29, wherein said disease is an ocular allergy.
39. The method according to claim 29, wherein said disease is an uveitis.
40. A heterodimeric protein composition capable of binding human IL-Ιβ, said protein
composition being capable of inhibiting human IL-6 production in human lung fibroblasts in response to treating the fibroblasts with human IL-Ιβ, said inhibiting having an IC50 value of about 2.3 pM.
PCT/US2013/026349 2013-02-15 2013-02-15 IL-1β INHIBITOR COMPOSITION AND USE THEREOF WO2014126582A1 (en)

Priority Applications (22)

Application Number Priority Date Filing Date Title
RS20240597A RS65559B1 (en) 2013-02-15 2013-02-15 Il-1beta inhibitor composition and use thereof
MX2015010438A MX2015010438A (en) 2013-02-15 2013-02-15 Il-1î² inhibitor composition and use thereof.
EP21208571.6A EP4050019A1 (en) 2013-02-15 2013-02-15 Il-1beta inhibitor composition and use thereof
BR112015019729-9A BR112015019729B1 (en) 2013-02-15 2013-02-15 HETERODIMERIC PROTEIN COMPOSITIONS CAPABLE OF BINDING HUMAN IL-1SS, THERAPEUTIC COMPOSITION COMPRISING THEM, NUCLEIC ACIDS, IN VITRO HETEROLOGY EXPRESSION SYSTEM AND USE OF A SUBSTANCE FOR THE TREATMENT OR PREVENTION OF A DISEASE ASSOCIATED WITH MODULATION OF IL-1SS ACTIVITY 1SS HUMAN
PL13875211.8T PL2956471T3 (en) 2013-02-15 2013-02-15 Il-1beta inhibitor composition and use thereof
FIEP13875211.8T FI2956471T3 (en) 2013-02-15 2013-02-15 Il-1beta inhibitor composition and use thereof
SI201332081T SI2956471T1 (en) 2013-02-15 2013-02-15 Il-1beta inhibitor composition and use thereof
PCT/US2013/026349 WO2014126582A1 (en) 2013-02-15 2013-02-15 IL-1β INHIBITOR COMPOSITION AND USE THEREOF
LTEPPCT/US2013/026349T LT2956471T (en) 2013-02-15 2013-02-15 Il-1beta inhibitor composition and use thereof
KR1020157025393A KR101989551B1 (en) 2013-02-15 2013-02-15 IL-1β INHIBITOR COMPOSITION AND USE THEREOF
EP13875211.8A EP2956471B1 (en) 2013-02-15 2013-02-15 Il-1beta inhibitor composition and use thereof
PT138752118T PT2956471T (en) 2013-02-15 2013-02-15 Il-1beta inhibitor composition and use thereof
AU2013378122A AU2013378122B2 (en) 2013-02-15 2013-02-15 IL-1beta inhibitor composition and use thereof
CN201380075648.9A CN105431448B (en) 2013-02-15 2013-02-15 IL-1 beta inhibitor composition and its application
JP2015557984A JP6225197B2 (en) 2013-02-15 2013-02-15 IL-1β inhibitor composition and use thereof
NZ71090013A NZ710900A (en) 2013-02-15 2013-02-15 Il-1β inhibitor composition and use thereof
DK13875211.8T DK2956471T3 (en) 2013-02-15 2013-02-15 IL-1BETA INHIBITOR COMPOSITION AND USE THEREOF
EA201500842A EA033269B1 (en) 2013-02-15 2013-02-15 INTERLEUKIN-1 BETA (IL-1β) INHIBITOR COMPOSITION AND USE THEREOF
SG11201506389YA SG11201506389YA (en) 2013-02-15 2013-02-15 IL-1β INHIBITOR COMPOSITION AND USE THEREOF
IL240553A IL240553B (en) 2013-02-15 2015-08-12 Il-1ß inhibitor composition and use thereof
PH12015501796A PH12015501796A1 (en) 2013-02-15 2015-08-14 Il-1b inhibitor composition and use thereof
HK16107308.4A HK1219281A1 (en) 2013-02-15 2016-06-23 Il-1beta inhibitor composition and use thereof il-1

Applications Claiming Priority (1)

Application Number Priority Date Filing Date Title
PCT/US2013/026349 WO2014126582A1 (en) 2013-02-15 2013-02-15 IL-1β INHIBITOR COMPOSITION AND USE THEREOF

Publications (1)

Publication Number Publication Date
WO2014126582A1 true WO2014126582A1 (en) 2014-08-21

Family

ID=51354448

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2013/026349 WO2014126582A1 (en) 2013-02-15 2013-02-15 IL-1β INHIBITOR COMPOSITION AND USE THEREOF

Country Status (21)

Country Link
EP (2) EP4050019A1 (en)
JP (1) JP6225197B2 (en)
KR (1) KR101989551B1 (en)
CN (1) CN105431448B (en)
AU (1) AU2013378122B2 (en)
BR (1) BR112015019729B1 (en)
DK (1) DK2956471T3 (en)
EA (1) EA033269B1 (en)
FI (1) FI2956471T3 (en)
HK (1) HK1219281A1 (en)
IL (1) IL240553B (en)
LT (1) LT2956471T (en)
MX (1) MX2015010438A (en)
NZ (1) NZ710900A (en)
PH (1) PH12015501796A1 (en)
PL (1) PL2956471T3 (en)
PT (1) PT2956471T (en)
RS (1) RS65559B1 (en)
SG (1) SG11201506389YA (en)
SI (1) SI2956471T1 (en)
WO (1) WO2014126582A1 (en)

Cited By (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN104628870A (en) * 2015-02-04 2015-05-20 中国药科大学 Human IL12Rbeta1-CHR protein and Fc fusion protein thereof
CN104725514A (en) * 2015-02-06 2015-06-24 中国药科大学 Novel IL23 antagonist
WO2021236091A1 (en) * 2020-05-22 2021-11-25 R-Pharm Overseas, Inc. Il1-r1 derived inhibitor of il-1b and use thereof
WO2022025643A1 (en) 2020-07-29 2022-02-03 (주)메디톡스 Heterodimeric fc fusion protein, and composition, use, and method related to same
EP4050019A1 (en) 2013-02-15 2022-08-31 R-Pharm International, LLC Il-1beta inhibitor composition and use thereof
US11844765B2 (en) 2017-09-26 2023-12-19 Regeneron Pharmaceuticals, Inc. Treatment of inflammatory conditions by delivery of interleukin-1 receptor antagonist fusion protein

Families Citing this family (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2018228442A1 (en) 2017-06-14 2018-12-20 Dingfu Biotarget Co., Ltd. Proteinaceous heterodimer and use thereof
CN116286844A (en) * 2023-02-13 2023-06-23 浙江大学 Homodimer protein combining human IL-1R1 and human IL-1RAcP polypeptide fragments and uses thereof
CN116144668A (en) * 2023-02-13 2023-05-23 杭州吉倍思生物制药有限公司 Human IL-1R1 gene, protein, homodimer protein and application

Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6472179B2 (en) 1998-09-25 2002-10-29 Regeneron Pharmaceuticals, Inc. Receptor based antagonists and methods of making and using
US20030143697A1 (en) 1998-09-25 2003-07-31 Neil Stahl Receptor based antagonists and methods of making and using
US7666622B2 (en) * 2005-10-19 2010-02-23 Regeneron Pharmaceuticals, Inc. Monomeric self-associating fusion polypeptides and therapeutic uses thereof

Family Cites Families (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
DE602006010072D1 (en) * 2005-06-21 2009-12-10 Chen Gang IL-1 BINDING ANTIBODIES AND FRAGMENTS THEREOF
EP2235064B1 (en) * 2008-01-07 2015-11-25 Amgen Inc. Method for making antibody fc-heterodimeric molecules using electrostatic steering effects
BR112013023653A2 (en) 2011-03-17 2016-12-13 Univ Ramot bispecific antibody, antibody preparation method, and pharmaceutical composition
WO2014035361A1 (en) * 2012-08-26 2014-03-06 R-PHARM, CJSC (Closed Joint Stock Company) IL-1β INHIBITOR COMPOSITION AND USE THEREOF
LT2956471T (en) 2013-02-15 2024-06-10 R-Pharm International, Llc Il-1beta inhibitor composition and use thereof

Patent Citations (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6472179B2 (en) 1998-09-25 2002-10-29 Regeneron Pharmaceuticals, Inc. Receptor based antagonists and methods of making and using
US20030143697A1 (en) 1998-09-25 2003-07-31 Neil Stahl Receptor based antagonists and methods of making and using
US20050197293A1 (en) 2002-10-28 2005-09-08 Scott Mellis Use of an IL-1 antagonist for treating arthritis
US7361350B2 (en) 2002-10-28 2008-04-22 Regeneron Pharmaceuticals, Inc. Use of an IL-1 antagonist for treating arthritis
US7666622B2 (en) * 2005-10-19 2010-02-23 Regeneron Pharmaceuticals, Inc. Monomeric self-associating fusion polypeptides and therapeutic uses thereof

Non-Patent Citations (11)

* Cited by examiner, † Cited by third party
Title
BHASKAR V ET AL.: "Monoclonal antibodies targeting IL -1 beta reduce biomarkers of atherosclerosis in vitro and inhibit atherosclerotic plaque formation in Apolipoprotein E-deficient mice.", ATHEROSCLEROSIS, vol. 216, no. 2, 2011, pages 313 - 320, XP028226561 *
DATABASE PUBMED [online] 1 January 2010 (2010-01-01), "ROLE OF IL-1BETA IN TYPE 2 DIABETES.", XP055290630, accession no. NCBI Database accession no. 20588114 *
DATABASE PUBMED [online] 1 January 2011 (2011-01-01), "[CAPS: CRYOPYRIN-ASSOCIATED PERIODIC SYNDROME].", XP055279021, accession no. NCBI Database accession no. 2204124 *
DATABASE PUBMED [online] 28 October 2009 (2009-10-28), "ANALYSIS OF INFLAMMATORY CYTOKINES IN THE TEARS OF DRY EYE PATIENTS", XP055279023, accession no. NCBI Database accession no. 19724208 *
DINARELLO CA ET AL.: "Role of IL -1 beta in type 2 diabetes", CURR OPIN ENDOCRINOL DIABETES OBES, vol. 17, no. 4, 2010, pages 314 - 321, XP009136506 *
ECONOMIDES A.N. ET AL.: "NATURE MEDICINE", vol. 9, 1 January 2003, NATURE PUB. CO, article "Cytokine traps: multi-component, high-affinity blockers of cytokine action", pages: 47 - 52
ECONOMIDES ARIS N. ET AL.: "Cytokine traps: multi-component, high-affinity blockers of cytokine action.", NATURE MEDICINE, vol. 9, no. 1, 2003, pages 47 - 52, XP002256034 *
MASSINGALE ML ET AL.: "Analysis of inflammatory cytokines in the tears of dry e patients.", CORNEA, vol. 28, no. 9, 2009, pages 1023 - 1027 *
SAITO MK .: "CAPS: cryopyrin-associated periodic syndrome.", NIHON RINSHO MENEKI GAKKAI KAISHI, vol. 34, no. 5, 2011, pages 369 - 377, XP055279021 *
See also references of EP2956471A4
WEBER A ET AL., SCI SIGNAL., vol. 3, no. 105, 19 January 2010 (2010-01-19)

Cited By (8)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP4050019A1 (en) 2013-02-15 2022-08-31 R-Pharm International, LLC Il-1beta inhibitor composition and use thereof
CN104628870A (en) * 2015-02-04 2015-05-20 中国药科大学 Human IL12Rbeta1-CHR protein and Fc fusion protein thereof
CN104725514A (en) * 2015-02-06 2015-06-24 中国药科大学 Novel IL23 antagonist
US11844765B2 (en) 2017-09-26 2023-12-19 Regeneron Pharmaceuticals, Inc. Treatment of inflammatory conditions by delivery of interleukin-1 receptor antagonist fusion protein
US11944666B2 (en) 2017-09-26 2024-04-02 Regeneron Pharmaceuticals, Inc. Treatment of inflammatory conditions delivery of interleukin-1 receptor antagonist fusion protein
WO2021236091A1 (en) * 2020-05-22 2021-11-25 R-Pharm Overseas, Inc. Il1-r1 derived inhibitor of il-1b and use thereof
WO2022025643A1 (en) 2020-07-29 2022-02-03 (주)메디톡스 Heterodimeric fc fusion protein, and composition, use, and method related to same
EP4190806A4 (en) * 2020-07-29 2024-08-14 Medytox Inc Heterodimeric fc fusion protein, and composition, use, and method related to same

Also Published As

Publication number Publication date
NZ710900A (en) 2019-09-27
AU2013378122A1 (en) 2015-08-27
CN105431448A (en) 2016-03-23
PL2956471T3 (en) 2024-06-24
SI2956471T1 (en) 2024-06-28
KR20160048028A (en) 2016-05-03
EP2956471A1 (en) 2015-12-23
DK2956471T3 (en) 2024-05-21
JP6225197B2 (en) 2017-11-01
CN105431448B (en) 2019-08-27
BR112015019729A2 (en) 2017-07-18
KR101989551B1 (en) 2019-09-30
IL240553B (en) 2021-02-28
BR112015019729B1 (en) 2023-01-24
PH12015501796B1 (en) 2015-11-09
RS65559B1 (en) 2024-06-28
IL240553A0 (en) 2015-10-29
LT2956471T (en) 2024-06-10
EP2956471A4 (en) 2019-11-13
MX2015010438A (en) 2016-05-05
FI2956471T3 (en) 2024-05-13
EA201500842A1 (en) 2016-02-29
EP2956471B1 (en) 2024-04-10
JP2016513106A (en) 2016-05-12
AU2013378122B2 (en) 2019-05-02
EA033269B1 (en) 2019-09-30
EP4050019A1 (en) 2022-08-31
HK1219281A1 (en) 2017-03-31
SG11201506389YA (en) 2015-09-29
PT2956471T (en) 2024-05-27
PH12015501796A1 (en) 2015-11-09

Similar Documents

Publication Publication Date Title
AU2013378122B2 (en) IL-1beta inhibitor composition and use thereof
CA2502385C (en) Il-1 receptor based antagonists and methods of making and using
KR102587838B1 (en) Hybrid immunoglobulin containing non-peptidyl linkage
KR102424590B1 (en) Fibronectin based scaffold domain proteins that bind to myostatin
KR20180004094A (en) Immunotherapeutics for cancer and autoimmune diseases
KR102666819B1 (en) Thrombin cleavable linker with xten and its uses thereof
KR101805201B1 (en) Bmp-alk3 antagonists and uses for promoting bone growth
KR100511544B1 (en) Recombinant il-5 antagonists useful in treatment of il-5 mediated disorders
KR20160086819A (en) Encapsulated cell therapy cartridge
US6129913A (en) Recombinant IL-5 antagonists useful in treatment of IL-5 mediated disorders
US11155600B2 (en) Human IL1-R1 derived inhibitor of IL-1β
WO1996021000A9 (en) Recombinant il-5 antagonists useful in treatment of il-5 mediated disorders
US20210403534A1 (en) IL1-R1 DERIVED INHIBITOR OF IL-1b AND USE THEREOF
EP0967994A1 (en) Improved method for treatment and diagnosis of il-5 mediated disorders
US10584147B2 (en) Procoagulant fusion compound
EP4153613A1 (en) Il1-r1 derived inhibitor of il-1b and use thereof
CN113637084A (en) Biomacromolecule targeted specific complement inhibitor and preparation method and application thereof

Legal Events

Date Code Title Description
WWE Wipo information: entry into national phase

Ref document number: 201380075648.9

Country of ref document: CN

121 Ep: the epo has been informed by wipo that ep was designated in this application

Ref document number: 13875211

Country of ref document: EP

Kind code of ref document: A1

WWE Wipo information: entry into national phase

Ref document number: 240553

Country of ref document: IL

Ref document number: MX/A/2015/010438

Country of ref document: MX

WWE Wipo information: entry into national phase

Ref document number: P1020/2015

Country of ref document: AE

ENP Entry into the national phase

Ref document number: 2015557984

Country of ref document: JP

Kind code of ref document: A

NENP Non-entry into the national phase

Ref country code: DE

WWE Wipo information: entry into national phase

Ref document number: IDP00201505143

Country of ref document: ID

ENP Entry into the national phase

Ref document number: 2013378122

Country of ref document: AU

Date of ref document: 20130215

Kind code of ref document: A

REG Reference to national code

Ref country code: BR

Ref legal event code: B01A

Ref document number: 112015019729

Country of ref document: BR

REEP Request for entry into the european phase

Ref document number: 2013875211

Country of ref document: EP

WWE Wipo information: entry into national phase

Ref document number: 201500842

Country of ref document: EA

Ref document number: 2013875211

Country of ref document: EP

ENP Entry into the national phase

Ref document number: 20157025393

Country of ref document: KR

Kind code of ref document: A

WWE Wipo information: entry into national phase

Ref document number: 15218431

Country of ref document: CO

ENP Entry into the national phase

Ref document number: 112015019729

Country of ref document: BR

Kind code of ref document: A2

Effective date: 20150817

WWE Wipo information: entry into national phase

Ref document number: P-2024/0597

Country of ref document: RS