US20190367600A1 - Prophylactic and/or therapeutic agent of infectious diseases or inflammatory diseases - Google Patents

Prophylactic and/or therapeutic agent of infectious diseases or inflammatory diseases Download PDF

Info

Publication number
US20190367600A1
US20190367600A1 US16/481,241 US201816481241A US2019367600A1 US 20190367600 A1 US20190367600 A1 US 20190367600A1 US 201816481241 A US201816481241 A US 201816481241A US 2019367600 A1 US2019367600 A1 US 2019367600A1
Authority
US
United States
Prior art keywords
antibody
apoa2
amino acid
antibodies
present
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Abandoned
Application number
US16/481,241
Other languages
English (en)
Inventor
Kazuo Suzuki
Yosuke Kameoka
Yoshio Yamakawa
Fukuko Kishi
Osamu Suzuki
Minako Koura
Junichiro Matsuda
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
A-Clip Institute Co Ltd
National Institutes of Biomedical Innovation Health and Nutrition
Original Assignee
A-Clip Institute Co Ltd
National Institutes of Biomedical Innovation Health and Nutrition
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by A-Clip Institute Co Ltd, National Institutes of Biomedical Innovation Health and Nutrition filed Critical A-Clip Institute Co Ltd
Assigned to A-CLIP INSTITUTE, CO., LTD., NATIONAL INSTITUTES OF BIOMEDICAL INNOVATION, HEALTH AND NUTRITION reassignment A-CLIP INSTITUTE, CO., LTD. ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: SUZUKI, KAZUO, YAMAKAWA, YOSHIO, MATSUDA, JUNICHIRO, KAMEOKA, Yosuke, KISHI, Fukuko, KOURA, Minako, SUZUKI, OSAMU
Publication of US20190367600A1 publication Critical patent/US20190367600A1/en
Abandoned legal-status Critical Current

Links

Images

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • AHUMAN NECESSITIES
    • A01AGRICULTURE; FORESTRY; ANIMAL HUSBANDRY; HUNTING; TRAPPING; FISHING
    • A01KANIMAL HUSBANDRY; AVICULTURE; APICULTURE; PISCICULTURE; FISHING; REARING OR BREEDING ANIMALS, NOT OTHERWISE PROVIDED FOR; NEW BREEDS OF ANIMALS
    • A01K67/00Rearing or breeding animals, not otherwise provided for; New or modified breeds of animals
    • A01K67/027New or modified breeds of vertebrates
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K49/00Preparations for testing in vivo
    • A61K49/0004Screening or testing of compounds for diagnosis of disorders, assessment of conditions, e.g. renal clearance, gastric emptying, testing for diabetes, allergy, rheuma, pancreas functions
    • A61K49/0008Screening agents using (non-human) animal models or transgenic animal models or chimeric hosts, e.g. Alzheimer disease animal model, transgenic model for heart failure
    • 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
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/02Immunomodulators
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/5005Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving human or animal cells
    • G01N33/5008Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving human or animal cells for testing or evaluating the effect of chemical or biological compounds, e.g. drugs, cosmetics
    • G01N33/5082Supracellular entities, e.g. tissue, organisms
    • G01N33/5088Supracellular entities, e.g. tissue, organisms of vertebrates
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/92Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving lipids, e.g. cholesterol, lipoproteins, or their receptors
    • AHUMAN NECESSITIES
    • A01AGRICULTURE; FORESTRY; ANIMAL HUSBANDRY; HUNTING; TRAPPING; FISHING
    • A01KANIMAL HUSBANDRY; AVICULTURE; APICULTURE; PISCICULTURE; FISHING; REARING OR BREEDING ANIMALS, NOT OTHERWISE PROVIDED FOR; NEW BREEDS OF ANIMALS
    • A01K2267/00Animals characterised by purpose
    • A01K2267/03Animal model, e.g. for test or diseases
    • A01K2267/0337Animal models for infectious diseases
    • AHUMAN NECESSITIES
    • A01AGRICULTURE; FORESTRY; ANIMAL HUSBANDRY; HUNTING; TRAPPING; FISHING
    • A01KANIMAL HUSBANDRY; AVICULTURE; APICULTURE; PISCICULTURE; FISHING; REARING OR BREEDING ANIMALS, NOT OTHERWISE PROVIDED FOR; NEW BREEDS OF ANIMALS
    • A01K2267/00Animals characterised by purpose
    • A01K2267/03Animal model, e.g. for test or diseases
    • A01K2267/035Animal model for multifactorial diseases
    • A01K2267/0368Animal model for inflammation
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/505Medicinal preparations containing antigens or antibodies comprising antibodies
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/60Immunoglobulins specific features characterized by non-natural combinations of immunoglobulin fragments
    • C07K2317/62Immunoglobulins specific features characterized by non-natural combinations of immunoglobulin fragments comprising only variable region components
    • C07K2317/622Single chain antibody (scFv)
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/70Immunoglobulins specific features characterized by effect upon binding to a cell or to an antigen
    • C07K2317/76Antagonist effect on antigen, e.g. neutralization or inhibition of binding
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N2333/00Assays involving biological materials from specific organisms or of a specific nature
    • G01N2333/435Assays involving biological materials from specific organisms or of a specific nature from animals; from humans
    • G01N2333/775Apolipopeptides
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N2500/00Screening for compounds of potential therapeutic value
    • YGENERAL TAGGING OF NEW TECHNOLOGICAL DEVELOPMENTS; GENERAL TAGGING OF CROSS-SECTIONAL TECHNOLOGIES SPANNING OVER SEVERAL SECTIONS OF THE IPC; TECHNICAL SUBJECTS COVERED BY FORMER USPC CROSS-REFERENCE ART COLLECTIONS [XRACs] AND DIGESTS
    • Y02TECHNOLOGIES OR APPLICATIONS FOR MITIGATION OR ADAPTATION AGAINST CLIMATE CHANGE
    • Y02ATECHNOLOGIES FOR ADAPTATION TO CLIMATE CHANGE
    • Y02A50/00TECHNOLOGIES FOR ADAPTATION TO CLIMATE CHANGE in human health protection, e.g. against extreme weather
    • Y02A50/30Against vector-borne diseases, e.g. mosquito-borne, fly-borne, tick-borne or waterborne diseases whose impact is exacerbated by climate change

Definitions

  • This invention is about agents for prophylaxis and/or therapy for infectious diseases or inflammatory diseases.
  • Antibody drugs (Infliximab, Actemura, etc.) have been used for rheumatoid arthritis and related diseases which is an inflammatory disease.
  • steroids and antibody drugs has been studied as there is no standard therapeutic agents for an intractable vasculitis which is a similar inflammatory disease.
  • immunoglobulin (ig) drugs have been used in intractable vasculitis, eosinophilia granulomatosis with polyangiitis: EGPA Churg-Strauss syndrome.
  • the immunoglobulin is a generic term of a protein structurally and/or functionally related to antibodies. In other words, an immunoglobulin, which binds to its specific antigens, is defined as antibody corresponding to specific antigens.
  • Infliximab which has been used as an antibody medicine for a specific antigen as a drug for (Remicade®) is an antibody to antigen an inflammatory cytokine TNF-alpha. It has been developed for an antibody drug to suppress over-produced TNF-alpha in Crohn's disease. Infliximab is an antibody drug that also destroys cells producing TNF alpha (non-patent document-1).
  • Rizximab (Rizane) has been developed for lymphoma treatment as a monoclonal antibody drug to CD20 in overseas, and also has begun to be used for intractable vasculitis (see non-patent document 2).
  • These antibody drug in which the antigens are identified are not specific to the intractable vasculitis, and it is considered that the antigen is neutralized by direct reactions with the antibodies.
  • Antibody medicines currently used are always associated with risks because these antibody drugs were not developed against an antigen specific for an intractable vasculitis.
  • the diversity of immunoglobulins consists of 10 8 clones.
  • Their molecular structure comprises light chains (L chains) and heavy chains (H chains), in which two kinds of polypeptides are linked by disulfide bonds.
  • the heavy chain comprises a constant region (C region) and a variable region (V region) composed of a VH region.
  • the light chain includes a constant region consisting of a CL region, and a variable region consisting of a VL region is connected.
  • Various antibodies to diverse antigens are made in vivo due to diversity of the amino acid sequence of the variable region.
  • the first hypothesis is that many kinds of antibodies against unknown antigens exhibit a pharmacological effect.
  • the second hypothesis is also that antibodies against an auto-antibody myeloperoxidase (MPO) against to MPO (anti-MPO antibody) are effective (non-patent literature 3).
  • MPO myeloperoxidase
  • anti-MPO antibody antibodies against an auto-antibody myeloperoxidase against to MPO
  • a variety of anti-MPO antibodies against a wide variety of epitopes of MPO exhibit pharmacological effects.
  • an antibody against anti-moesin antibody which is a self-antibody that is involved in the severity of intractable vasculitis antibody is effective (patent reference-1 and non-patent references 4-5).
  • the immunoglobulin medicine is a mixture of a lot of varieties of immunoglobulins, that is, polyclonal antibodies contribute to a therapeutic effect.
  • Another hypothesis is that a variety of antibodies against a specific antigens exhibit efficacy.
  • the antibody drug can be produced by recombinant DNA technology using the recombinant ScFv as a model.
  • the chimeric antibody and humanized antibody have already been practically used as antibody drugs (patent references 6-7).
  • immunoglobulin protein fused with chaperon is produced as a technique of production of soluble normal proteins by expression of immunoglobulin genes in host cells. All of these are a single molecule immunoglobulin, that is, a monoclonal immunoglobulin or its fragment.
  • Non-patent document 1 Gilberto Poggioli, Silvio Laureti, Massimo Campieri, Filippo Pierangeli, Paolo Gionchetti, Federica Ugolini, Lorenzo Gentilini, Piero Bazzi, Fernando Rizzello, and Maurizio Coscia. Infliximab in the treatment of Crohn's disease. TherClinRiskManag. 2007 June; 3(2): 301-308.
  • Non-patent document 2 Watts R A, Scott D G. Vasculitis and inflammatory arthritis. Best Pract Res Clin Rheumatol. 2016 October; 30(5):916-931.
  • Non-patent document 3 Goeken J A. Antineutrophil cytoplasmic antibody-A useful serological marker for vasculitis. J Clin Immunol 1991; 11: 161-174
  • Non-patent document 4 Suzuki K, Nagao T, Itabashi M, Hamano Y, Sugamata R, Yamazaki Y, Yumura W, Tsukita S, Wang P C, Nakayama T, Suzuki K. A novel autoantibody against moesin in the serum of patients with MPO-ANCA-associated vasculitis. Nephrol Dial Transplant. 2014 June; 29(6):1168-77.
  • Non-patent document 5 Ellen F. Carney.
  • VASCULITIS Potential role of an antimoesin autoantibody in AAV Nature Reviews Nephrology 2014; 10:3.
  • Non-patent document 6 Ito-Ihara T, Ono T, Nogaki F, Suyama K, Tanaka M, Yonemoto S, Fukatsu A, Kita T, Suzuki K, E. Muso. Clinical efficacy of intravenous immunoglobulin for patients with MPO-ANCA-associated rapidly progressive glomerulonephritis. Nephron Clin Pract. 2005; 102: c35-c42.
  • Non-patent document 7 Unizony S, Villarreal M, Miloslaysky E M, Lu N, Merkel P A, Spiera R, Seo P, Langford C A, Hoffman G S, Kallenberg C M, St Clair E W, Ikle D, Tchao N K, Ding L, Brunetta P, Choi H K, Monach P A, Fervenza F, Stone J H, Specks U; RAVE-ITN Research Group. Clinical outcomes of treatment of anti-neutrophil cytoplasmic antibody (ANCA)-associated vasculitis based on ANCA type. Ann Rheum Dis. 2015 Nov. 30. pii: annrheumdis-2015-208073. doi: 10.1136/annrheumdis-2015-208073. [Epub ahead of print]
  • ANCA anti-neutrophil cytoplasmic antibody
  • Non-patent document 8 de Joode A A, Roozendaal C, van der Leij M J, Bacheer L B, Sanders J S, Stegeman C A. Performance of two strategies for urgent ANCA and anti-GBM analysis in vasculitis. Eur J Intern Med. 2014 February; 25(2):182-6. doi: 10.1016/j.ejim.2013.11.011. Epub 2013 Dec. 19.
  • Non-patent document 9 Kallenberg C G, Stegeman C A, Heeringa P. Autoantibodies vex the vasculature. Nat. Med. 2008 October; 14(10):1018-9.
  • Non-patent document 10 Kain R, Exner M, Brandes R, Ziebermayr R, Cunningham D, Alderson C A, Davidovits A, Raab I, Jahn R, Ashour O, Spitzauer S, Sunder-Plassmann G, Fukuda M, Klemm P, Rees A J, Kerjaschki D. Molecular mimicry in pauci-immune focal necrotizing glomerulonephritis. Nat. Med. 2008 October; 14(10):1088-96. Epub 2008 Oct. 5.
  • Non-patent document 11 Tomizawa K, Nagao T, Kusunoki R, Saiga K, Oshima M, Kobayashi K, Nakayama T, Tanokura M, Suzuki K. Reduction of MPO-ANCA epitopes in SCG/Kj mice by 15-Deoxyspergualin treatment restricted by IgG2b associated with crescentic glomerulonephritis. Rheumatology (Oxford). 2010; 49:1245-56.
  • the present invention provides a therapeutic method specialized for inflammatory diseases such as intractable vasculitis and infectious diseases according to determination of target molecules of these diseases which is different therapeutic methods from the prior to the method with unknown target molecules.
  • the present inventors have intensively studied to solve the above-described problems. As a results of overcoming much difficulties in the process, the present inventors determined that the target antigen of the clone (“VasSF”), which shows various therapeutic effects of a human single-chain variable fragment (hScFv) on a mouse model of intractable vasculitis (patent document 5) as in Sequence No. 31 comprising an amino acid sequence represented, was apolipoprotein a2 (APOA 2). As these results, the present inventors the present invention has been completed as resent invention has been completed. As a result, the present inventors have found that use of an antibody that specifically recognizes APOA2 makes it possible to prevent and/or treat inflammatory diseases such as vasculitis and infectious diseases, and completed the present invention.
  • VasSF target antigen of the clone
  • APOA2 apolipoprotein a2
  • a prophylactic and/or therapeutic agent which contains an antibody recognizing apolipoprotein a2 (APOP, 2) as a specific antigen, is provided for infectious diseases and inflammatory.
  • APOP apolipoprotein a2
  • recombinant fragments of immunoglobulins of anti-APOA2 (SEQ ID NO: 4) and polypeptides of immunoglobulins of anti-APOA2 containing the same or substantially the same amino acid sequences as the amino acid sequence represented by the expression (SEQ ID NO: 4) are provided.
  • a vector containing polynucleotide encoding a polypeptide containing the same or substantially identical amino acid sequence as the amino acid sequence represented by the structure containing the polynucleotide and its polynucleotide, and a host cells containing its vector are provided.
  • methods including administration of an effective amount of anti-APOA2 antibody for preventing and/or treating infectious diseases or inflammatory diseases, and methods for screening a prophylactic and/or therapeutic agent for infectious diseases or inflammatory diseases using apolipoprotein A2 (APOA2) are provided.
  • APOA2 apolipoprotein A2
  • induction reagents containing an apolipoprotein A2 (APOA2) for infectious diseases or inflammatory diseases as an effective component procedures for preparation of a model mouse of infectious diseases or inflammatory diseases by administration of apolipoprotein A2 (APOA2) into a non-transgenic animal except humans, and the pathological model animal of the infectious diseases or inflammatory diseases induced with this procedures are provided.
  • APOA2 apolipoprotein A2
  • the target molecule for the effective on treating inflammatory diseases or infectious diseases such as an intractable vasculitis was identified. Based on the evidence, it is possible that a specific treatment method is provided which is different from the previous methods.
  • FIG. 1A Illustration describing a method used for identification of an antigen targeted by a specific antibody.
  • FIG. 1B A photograph showing the results of attempting to identify the target antigen by the ProteoSeekTM method in Embodiment 1.
  • FIG. 1C A photograph showing the results of attempting to identify the target antigen by the HiTrap® using an NHS-activated HP column in Embodiment 1.
  • FIG. 2 A photograph showing the results of attempting to identify the target antigen by the HiTrap® using the tosyl group activation of DYNABEADS® in Embodiment 1.
  • FIG. 3 The left is the same photograph as FIG. 2 , and the right panel shows a table of 11 kinds of candidate molecules hit by the MS ion search.
  • FIG. 4 A microscope photograph showing a result of evaluating a therapeutic effect of anti-APOA 2 polyclonal antibody on the kidney tissues in Embodiment 4.
  • FIG. 5 A graph showing the results of evaluation of the therapeutic effect of anti-APOA 2 polyclonal antibody the crescent formation in kidney glomerular tissues in Embodiment 4.
  • FIG. 6 A microscope photograph showing the results of evaluation of the therapeutic effect of anti-APOA 2 polyclonal antibody on kidney tissues in Embodiment 4.
  • FIG. 7 A graph showing the results of evaluation of the therapeutic effect of anti-APOA 2 polyclonal antibody on spleen weight in Embodiment 4.
  • FIG. 8 A graph showing the results of evaluation of the therapeutic effect of anti-APOA 2 polyclonal antibody on levels of MPO-ANCA and anti-moesin antibodies in sera in Embodiment 4.
  • FIG. 9 A graph showing the results of evaluation of the therapeutic effect of anti-APOA 2 polyclonal antibody on levels of cytokines and chemokines in sera in Embodiment 4.
  • FIG. 10 A graph showing the results of evaluation of the therapeutic effect of anti-APOA 2 polyclonal antibody on levels of white blood cells in the peripheral blood in Embodiment 4.
  • FIG. 11 A microscope photograph showing the results of evaluation of the therapeutic effect of anti-APOA 2 polyclonal antibody on lung tissues in Embodiment 4.
  • FIG. 12 Figure for explaining the plasmid ptac2-URq01_OptE.coli prepared in Embodiment 5.
  • FIG. 13 Figure for explaining a method for incorporating the fragment containing a gene encoding an antibody in the invention a built-in a site where a T7 promoter control region of a pET32 vector in Embodiment 5.
  • FIG. 14 Figure for explanatory diagram for explaining the whole operation in Embodiment 5.
  • FIG. 15 Figure for results in purification of the recombinant protein (VasAP) from the culture of E. coli cells DE32) in Embodiment 5.
  • FIG. 16 Figure for results in evaluation of the therapeutic effect on the rate of crescent formation in kidney glomeruli by treatment with anti-APOA2 monoclonal antibody (VasAP) in Embodiment 6.
  • FIG. 17 Figure for results in evaluation of the therapeutic effect on weight of spleen by treatment with anti-APOA2 monoclonal antibody (VasAP) in Embodiment 6.
  • FIG. 18 Figure for results in evaluation of the therapeutic effect on serum levels of MPO-ANCA and anti-moesin antibody by treatment with anti-APOA2 monoclonal antibody (VasAP) in Embodiment 6.
  • FIG. 19 Figure for results in evaluation of the therapeutic effect on serum levels of cytokines and chemokines by treatment with anti-APOA2 monoclonal antibody (VasAP) in Embodiment 6.
  • FIG. 20 Figure for results in evaluation of the therapeutic effect on cell counts in the peripheral blood by treatment with anti-APOA2 monoclonal antibody (VasAP) in Embodiment 6.
  • VasAP anti-APOA2 monoclonal antibody
  • FIG. 21 Graph of results in daily evaluation of the urinary score in wild-type mice after administration of APOA2 protein in Embodiment 7.
  • FIG. 22 Graph of results in the rate of crescent formation in kidney glomeruli in wild-type mice by administration of APOA2 protein in Embodiment 7.
  • FIG. 23 Photograph of microscope in kidney tissues of wild-type mouse by administration of APOA2 protein in Embodiment 7.
  • FIG. 24 Photograph of microscope in spleen and lung tissues of wild-type mouse by administration of APOA2 protein in Embodiment 7.
  • the first embodiment of the present invention is a prophylactic and/or therapeutic agent for infectious diseases or inflammatory diseases relates to apolipoprotein A2 (APOA2) as an active ingredient.
  • APOA2 apolipoprotein A2
  • the present inventors have outlined how the present inventors have completed the present invention, first, the inventors of the present invention (5) showing various therapeutic effects on the intractable vasculitis model mice:
  • hscFv human single-chain variable fragment
  • VasSF human single-chain variable fragment
  • APOA2 apolipoprotein a2
  • the human apolipoprotein A2 (hPOA2) is encoded in 473-bp-long APOA2 gene (RefSeq accession no. NM 001643; CDS sequence corresponds to SEQ ID NO:1) and comprises 100 amino acids (RefSeq accession No. NP 001634, SEQ ID NO: 2)
  • Apolipoprotein a2 used in the present invention; “the protein of the present invention is also referred to as “protein of the present invention”) is a protein having the same sequence of amino acids or substantially the same sequence by the formula 2.
  • the proteins of the present invention include humans and other warm-blooded animals (for example, guinea pigs, rats, mice, chicken, rabbits, pigs, sheep, bovine, monkey, and the like) of cells (e.g., hepatocytes, spleen cells, and nerve cells), glial cells, pancreatic beta cells, bone marrow cells, mesangial cells, Langerhans cells, and epidermal cells, epithelial cells, goblet cells, endothelial cells, smooth muscle cells, and fibroblast cells, muscle cells, adipocytes, immune cells (e.g., macrophages, T cells, and B cells, natural killer cells, mast cells, neutrophils, basophils, eosinophils, monocytes), mega
  • the protein may be a purified protein from above source.
  • the protein may be a protein which is chemically synthesized by a chemical synthesis or a cell-free translation system, or a recombinant protein produced from a transformant in which a nucleic acid having a nucleotide sequence encoding the amino acid sequence is introduced
  • the amino acid sequence substantially identical to the amino acid sequence represented by SEQ ID NO 2 is the amino acid sequence represented by the formula about 50% or more, and preferably about 60% or more, more preferably about 70% or more, even more preferably about 80% or more, and particularly preferably about 90% or more, and most preferably, an amino acid sequence having homology (homology) of about 95% or more in the SEQ ID NO 2.
  • “homology” means and the ratio (%) of the overlapping residues of the same amino acid and similar amino acid in total amino acids which are referred to a mathematical algorithm known in the art when one of the amino acid sequence is aligned with another amino acid sequence (the algorithm may take into account the introduction of a gap into one or both of the sequences for optimal alignment).
  • Similar amino acid refers to amino acids similar in physicochemical properties, for example, an aromatic amino acid (Phe, Trp, Tyr), aliphatic amino acids (Ala, Leu, Ile, Val), polar amino acids (Gln, Asn); basic amino acids (Lys, Arg, His), acidic amino acids (Glu, Asp), an amino acid having a hydroxyl group (Ser, Thr), and an amino acid having a small side chain (Gly, Ala, Ser, Thr, Met).
  • substitution with similar amino acids does not result in changes in the phenotype of the protein (that is, conservative amino acid substitutions).
  • Specific examples of conservative amino acid substitutions are well known in the art and are described in various references (see, for example, Bowie et al. Science, 247: 1306-1310 (1990))
  • the amino acid sequence having substantially the same amino acid sequence as the amino acid sequence represented by SEQ ID NO: (2) is the amino acid sequence with about 50% or more, and preferably about 60% or more, more preferably about 70% or more, even more preferably about 80% or more, and particularly preferably about 90% or more, most preferably at least about 95% identity.
  • the protein used in the present invention is a protein with substantially the same amino acid sequence as represented by SEQ ID NO: (2), and with substantially identical activities as those of a protein containing the amino acid sequence represented by SEQ ID NO: (2).
  • the substantially identical activities include, for example, ligand binding activity and signal transduction.
  • substantially identical means qualitatively (e.g., physiologically or pharmacologically) identical.
  • the protein of the present invention has the identical activity.
  • the degree of the activity e.g., about 0.01 to about 100 times, preferably from about 0.1 to about 10 times, more preferably from 0.5 to 2 times
  • a quantitative element such as a molecular weight of the protein may be different.
  • Measurement of activities can be carried out in accordance with known methods.
  • the activities can be measured by a method used in the screening method, and the like, for compounds or their salts, which inhibit the activity of a protein used in the present invention, which will be described later.
  • the proteins used in the present invention include proteins with, for example, (i) one or more amino acid residues (for example, from one to 50, and preferably from one to 30, more preferably from one to 10, and even more preferably from one to five, four, three, or two amino acid residues) are deleted from the amino acid sequence represented by SEQ ID NO: 2, (ii) one or more amino acid residues (for example, from one to 50, and preferably from one to 30, more preferably from one to 10, and even more preferably from one to five, four, three, or two amino acid residues) are added to the amino acid sequence represented by SEQ ID NO: 2, (iii) one or more amino acid residues (for example, from one to 50, and preferably from one to 30, more preferably from one to 10, and even more preferably from one to five, four, three, or two amino acid residues) are inserted into the amino acid sequence represented by SEQ ID NO: 2, (iv) one or more amino acid residues (for example, from one to 50, and preferably from one to 30, more
  • proteins used in the present invention include, for example, human apolipoprotein A2 (RefSeq Accession No. NP 001634) containing the amino acid sequence shown in SEQ ID NO: (2), and it's homologs in another mammals.
  • Proteins used in the present invention including a protein containing an amino acid sequence represented by SEQ ID NO: 2 may have a carboxyl group (—COOH), a carboxylate (—COO ⁇ ), an amide (—CONH 2 ) or an ester (—COOR) at the C-terminal.
  • proteins used in the present invention contain proteins with an amino group of the amino acid residue at N-terminal (e.g., methionine residue)) of which are protected by protecting groups (for example, formyl group, C 1-6 acyl groups such as C 1-6 alkanoyl such as acetyl group), proteins with glutamine residues produced by in vivo cleavage and pyroglutamated, proteins of which substituent groups (e.g., —OH, —SH, amino groups, imidazole groups, indole groups, and guanidine groups) in intramolecular side chains of amino acid resides are protected by appropriate protecting groups (for example, formyl group, C 1-6 acyl groups such as C 1-6 alkanoyl such as acetyl group), complex proteins such as so-called glycoproteins with glycosylation.
  • protecting groups for example, formyl group, C 1-6 acyl groups such as C 1-6 alkanoyl such as acetyl group
  • substituent groups
  • the partial peptides of the protein used in the present invention may be any peptides as long as they have partial amino acid sequence of the protein used in the present invention described above and also have substantially identical activity as that of the protein.
  • substantially identical activity is defined the same as that described above.
  • the “substantially identical activity” can be measured in the same manner for the protein in the present invention as described above.
  • peptides containing at least 20-amino-acid-long or longer, preferably 50- or longer, more preferably 70- or longer, more preferably 100- or longer, and most preferably 150- or longer of the constituent amino acid sequence of the protein used in the present invention can be used.
  • the partial peptides used in the present invention can be used even though one amino acid or more (preferably one to 20, more preferably one to 10, more preferably several (one to five, four, three or two) are deleted, one amino acid or more (preferably one to 20, more preferably one to 10, more preferably several (one to five, four, three or two) are added, one amino acid or more (preferably one to 20, more preferably one to 10, more preferably several (one to five, four, three or two) are inserted, or one amino acid or more (preferably one to 20, more preferably one to 10, more preferably several (one to five, four, three or two) are substituted to different amino acids in their amino acid sequences.
  • the partial peptides used in the present invention can also be used as antigens for antibody preparation.
  • Proteins or its partial peptides used in the present invention may be in free or salt forms (hereinafter, the same unless otherwise specified).
  • Such salts include physiologically acceptable acids (e.g., inorganic acids, and organic acids) and salts with bases (e.g., alkali metal salts).
  • physiologically acceptable acid addition salts are preferred.
  • salts include inorganic acids (e.g., hydrochloric acid, phosphoric acid, hydrobromic acid, and sulfuric acid) and organic acids (e.g., acetic acid, formic acid, propionic acid, fumaric acid, maleic acid, succinic acid, tartaric acid, citric acid, malic acid, oxalic acid, benzoic acid, methanesulfonic acid, and benzenesulfonic acid).
  • inorganic acids e.g., hydrochloric acid, phosphoric acid, hydrobromic acid, and sulfuric acid
  • organic acids e.g., acetic acid, formic acid, propionic acid, fumaric acid, maleic acid, succinic acid, tartaric acid, citric acid, malic acid, oxalic acid, benzoic acid, methanesulfonic acid, and benzenesulfonic acid.
  • proteins used in the present invention can be derived from cells or tissues of humans or other warm-blooded animals mentioned above by purification using known protein purification methods.
  • proteins used in the present invention can be prepared by homogenization of tissues or cells of mammals in the presence of detergents and purification from the crude extract fraction by chromatography, such as reverse phase chromatography, ion exchange chromatography, and affinity chromatography.
  • proteins or their partial peptides used in the present invention can also be produced according to known peptide synthesis methods.
  • Partial peptides of the proteins used in the present invention can be produced by fragmentation of proteins used in the present invention with suitable peptidases.
  • proteins or their partial peptides used in the present invention can be produced by separation and purification of the proteins or their partial peptides from culture products of transformants containing polynucleotides encoding the proteins or their partial peptides
  • Antibodies against proteins or their partial peptides used in the present invention can be either polyclonal or monoclonal antibodies if they specifically recognize the proteins or their partial peptides.
  • the isotypes of the antibodies are not particularly limited, but preferably IgG, IgM or IgA, and particularly preferably IgG.
  • the antibodies of the present invention are not particularly limited if at least they have complementarity determining regions (CDR) for specific recognition and binding to their targets.
  • the antibodies of the present invention may be complete molecules of the antibodies and their fragments such as Fab, Fab′, F(ab′), ScFv, ScFv-Fc, genetically-engineered conjugate molecules such as mini-bodies and dia-bodies, or derivatives modified with molecules having protein stabilizing actions such as polyethylene glycol (PEG).
  • PEG polyethylene glycol
  • the antibodies of the present invention include, for example, polypeptides containing the same or substantially identical amino acid sequence shown in SEQ ID NO 4 (preferably, polypeptides with the same or essentially the same amino acid sequence represented in SEQ ID NO 4).
  • anti-APOA2 recombinant immunoglobulin fragment compounds containing polypeptides containing the same or essentially the same amino acid sequence represented in SEQ ID NO 4 preferably, polypeptides with the same or essentially the same amino acid sequence represented in SEQ ID NO 4
  • active ingredients are provided.
  • These anti-APOA2 recombinant immunoglobulin fragment compounds are preferably used for the prevention and/or treatment of infectious diseases or inflammatory diseases.
  • Antibodies to the proteins or their partial peptides used in the present invention can be produced according to known methods for antibody or anti-serum production.
  • Either proteins or partial peptides in the present invention described above, or (synthetic) peptides containing one, two, or more of the same antigenic determinants thereof may be used as antigens for preparation of antibodies of the present invention.
  • Proteins or their partial peptides of the present invention is produced by, for example, (a) known or similar preparation methods with tissues or cells of warm-blooded animals such as humans, monkeys, rats, mice, and chicken, (b) known chemical preparation methods with peptide synthesizers or similar machines, (c) culture of transformants containing DNA encoding the proteins or their partial peptides in the present invention, or (d) biochemical synthesis using cell-free translation/transcription with DNA encoding the proteins or their partial peptides in the present invention as templates.
  • Antigens of the present invention are administrated to warm-blooded animals at sites capable of antibody production by, e.g., intraperitoneal, intravenous, subcutaneous, or intradermal injections.
  • complete or incomplete Freund's adjuvants may be administered. Administrations are usually performed once every one to six weeks, twice to 10 times in total.
  • the warm-blooded animals used for antibody production include, for example, monkeys, rabbits, dogs, guinea pigs, mice, rats, hamsters, sheep, goats, donkeys, and a chicken.
  • mice and rats are preferably used for monoclonal antibody productions.
  • the artificial immunization for humans is ethically difficult. Therefore, when the antibody of the present invention is to be administered to humans, following methods are preferable for production of human antibodies: (i) immunization of human-antibody-producing animals (for example, a mice) prepared according to a method described later, (ii) preparation of humanized antibodies or complete human antibodies in accordance with a method for production of chimera antibodies described later, or (iii) combinations of in vitro immunizations, virus-based cell immortalizations, human-human (or mouse) hybridoma production techniques, phage display method, etc.
  • human-antibody-producing animals for example, a mice
  • preparation of humanized antibodies or complete human antibodies in accordance with a method for production of chimera antibodies described later or combinations of in vitro immunizations, virus-based cell immortalizations, human-human (or mouse) hybridoma production techniques, phage display method, etc.
  • In vitro immunization methods are also preferably used because antibodies may be produced against antigens which suppress antibody productions in regular in vivo immunization, antibodies can be obtained with only a little amount (ng to ⁇ g order) of antigens, and immunization can be completed in several days.
  • Animal cells used for in vitro immunization methods include lymphocytes, preferably B lymphocytes from peripheral blood, spleens, lymph nodes, etc., of humans and warm-blooded animals described above (preferably mice and rats).
  • lymphocytes preferably B lymphocytes from peripheral blood, spleens, lymph nodes, etc.
  • the spleen is excised from animals of 4 to 12 weeks of age and then spleen cells are isolated.
  • the spleen cells are washed in a suitable medium (e.g., Dulbecco's modified eagle's medium (DMEM), RPMI 1640 medium, Ham's F12 medium) and then cultured in a medium with antigens and 5 to 20% of fetal bovine serum (FCS) in a CO 2 incubator for 4 to 10 days.
  • DMEM Dulbecco's modified eagle's medium
  • FCS fetal bovine serum
  • the antigen concentration at 0.05 to 5 ⁇ g is recommended, but not limited.
  • Addition of supernatants from cultures of splenocytes from the same strain of animals (1 to 2 weeks of age is preferable) prepared in accordance with a conventional method to the culture medium is preferable.
  • cytokines such as IL-2, IL-4, IL-5, IL-6, and optionally adjuvant substances (e.g., Muramyl dipeptide) should be added to the medium together with the antigens because it is difficult to obtain supernatants from cultures of human thymus cells.
  • adjuvant substances e.g., Muramyl dipeptide
  • Separation and purification of monoclonal antibodies can be performed by known methods, for example, immunoglobulin separation and purification methods [e.g., salting-out methods, alcohol sedimentation methods, isoelectric point precipitation methods, electrophoretic methods, ion exchanger (e.g., DEAE, and QEAE) adsorption/desorption methods, ultracentrifuge methods, gel filtration methods, or specific purification methods for obtaining antibodies by elution after specific binding of antibodies to active adsorbents such as carriers bound to antigens, protein A or protein G).
  • immunoglobulin separation and purification methods e.g., salting-out methods, alcohol sedimentation methods, isoelectric point precipitation methods, electrophoretic methods, ion exchanger (e.g., DEAE, and QEAE) adsorption/desorption methods, ultracentrifuge methods, gel filtration methods, or specific purification methods for obtaining antibodies by elution after specific binding of antibodies to active adsorbents such as carriers
  • monoclonal antibodies can be produced by collecting the antibodies from the body fluid or culture fluid.
  • the antibodies in the present invention must have a therapeutic activity against infectious diseases and inflammatory diseases when the antibodies are used for prevention and treatment of these diseases, it is necessary to examine the level of the therapeutic activity of the obtained monoclonal antibodies.
  • the therapeutic activity can be measured by comparison of efficacies between disease model animals treated with and without the antibodies.
  • the antibodies of the present invention are used as a medicine to be administered to humans. Therefore, antibodies (preferably monoclonal antibodies) of the present invention have a reduced risk to induce antigenicity when administered to humans (specifically, complete human antibodies, humanized antibodies, mouse-human chimeric antibodies, etc.), and particularly preferably complete human antibodies.
  • the humanized antibodies and the chimeric antibodies can be genetically engineered according to a method described below.
  • the complete human antibodies can be produced from the human-human (or mouse) hybridomas, but the methods with human antibody-producing animals (for example, mice) or phage display methods described later are preferable for stable and inexpensive production of a large amount of antibodies.
  • chimeric antibodies refers to antibodies of which variable regions of H and L chains (VH and VL) are derived from one mammalian species and constant regions (CH and CL) are derived from another mammalian species.
  • Amino acid sequences of variable regions are preferably derived from animal species which can be used for easy production of hybridomas, such as mice.
  • sequences of the constant regions are preferably derived from a mammalian species to be administered.
  • Fab and F(ab′) 2 can be prepared by papain and pepsin digestions, respectively, of obtained chimeric antibodies.
  • the ScFv can be made by combining DNA encoding mouse V H and V L regions with suitable linkers encoding, for example, one to 40 amino acids, preferably three to 30 amino acids, more preferably five to 20 amino acids (e.g., [Ser-(Gly) m ] n or [(Gly) m -ser] n (m is an integer of Zero to 10, n is an integer of one to five) or the like).
  • miniboy monomers can made by combining ScFv with DNA encoding C H3 via suitable likers.
  • ScFv-Fc can be made by combining ScFv with DNA encoding full sequence of C H3 via suitable likers. DNAs encoding such genetically modified (conjugated) antibody molecules can be expressed in E. coli or yeasts when the DNAs are placed under the control of an appropriate promoter and a large amount of antibody molecules can be produced.
  • dimers called Fv are produced by monocistronic gene expression.
  • dimers called dsFv are formed by intermolecular disulfide bonds between both chains.
  • humanized antibodies refer to antibodies in which sequences other than the complementarity determining regions (CDR) in variable regions (i.e., constant regions and framework regions (FR) of variable regions) are derived from humans and only the CDR sequences are derived from other mammalian species.
  • CDR complementarity determining regions
  • FR framework regions
  • other mammalian species animal species which can be used for easy production of hybridomas, such as mice, are preferable.
  • Humanized antibodies can also be converted into ScFv, ScFv-Fc, minibody, dsFV, Fv or the like by genetic engineering techniques as well as chimeric antibodies.
  • the antibodies can be produced by a suitable promoter in microorganisms such as E. coli and yeasts.
  • Techniques for producing humanized antibodies can be applied to produce monoclonal antibodies in animals in which hybridoma production is not established and the produced antibodies can be preferably administered to the animal species.
  • livestock such as bovine, porcine, sheep, goats, chicken and the like
  • pet animals such as dogs and cats can be targets of therapy with humanized antibodies.
  • the antibodies of the present invention are used as pharmaceutical agents, they are preferably monoclonal antibodies, but polyclonal antibodies are also acceptable.
  • the antibodies of the present invention are polyclonal antibodies, the use of hybridomas is not required. Therefore, the animals in which the hybridoma production technique is not established but the transgenic technique is established, preferably ungulates like bovine, can be used for a larger amount of human antibodies at a low cost with similar methods described above (See, for example, Nat. Biotechnol., 20: 889-994 (2002)).
  • the resulting human polyclonal antibodies can be obtained by purification from blood, ascites, milk, eggs, etc., preferably from milk or eggs, of human antibody-producing animals by the combination of the same purification techniques as described above.
  • Another approach for producing complete human antibodies is a method using phage display.
  • mutation by PCR errors may be introduced to sites other than CDR, and, as a result, there is a small number of reports of HAHA production in a clinical stage.
  • the method has some advantages that there is no risk of xenogenic viral infection from host animals, and that the infinite specificity of the antibodies can be expected (antibodies against prohibited clones and sugar chains, etc., can be easily prepared).
  • the method for preparing the phage display human antibody libraries is not particularly limited.
  • the polyclonal antibodies of the present invention can be produced in accordance with known or similar methods. For example, using immunogens (protein or peptide antigens) alone or complexes of immunogens and carrier proteins, the warm-blooded animals are immunized in the same manner as in the production method of the monoclonal antibodies. Then, antibodies against the proteins in the present invention can be produced by the collection of antibody-containing substances from the immunized animals and separation/purification of the antibodies.
  • Polyclonal antibodies are collected from, e.g., blood, and ascites (preferably blood) of warm-blooded animals immunized in the manner described above.
  • Polyclonal antibody titers in antisera can be measured in the same manner as the measurement of the monoclonal antibody titers in the antiserum as described above.
  • Polyclonal antibodies can be separated and purified by the method similar to ones for the separation/purification of monoclonal antibodies as described above.
  • protein of the present invention or partial peptides thereof (hereinafter, simply referred to as “protein of the present invention”), a DNA encoding the protein of the present invention or partial peptides thereof, and applications of the antibodies of the present invention are described below.
  • the protein is a target molecule of the antibodies of the present invention
  • the presence, absence, or degree of expression of the protein of the present invention in patients affected or suspected to be affected by infectious diseases or inflammatory diseases can be used as an index for the estimation of efficacies of the antibodies of the present invention for the prevention/treatment to the diseases.
  • the antibodies of the present invention can be used as medicines for prevention/treatment of infectious diseases or inflammatory diseases.
  • the present invention includes methods for preventing and/or treating infectious diseases or inflammatory diseases by administering an effective amount of the antibodies of the present invention.
  • persons like medical doctors can appropriately determine effective amount by, e.g., the degree of activity of the antibodies, the physical conditions of the patient, the conditions of the disease.
  • Infectious diseases are broadly interpreted and cover a wide variety of infections of foreign agents (for example, infection of various bacteria, viruses, or parasites, and sepsis caused by these pathogens).
  • the inflammatory diseases include intractable vasculitis (for example, idiopathic thrombocytopenic purpura, agammaglobulinemia, Kawasaki disease, guillain-Barre syndrome, Microscopic PolyAngitis (MPA), and Eosinophilic granulomatosis with polyangiitis (EGPA, Churg-Strauss syndrome)), nephritis and/or glomerulonephritis, idiopathic pulmonary fibrosis, etc.
  • intractable vasculitis for example, idiopathic thrombocytopenic purpura, agammaglobulinemia, Kawasaki disease, guillain-Barre syndrome, Microscopic PolyAngitis (MPA), and Eosinophilic granulomatosis with polyangiitis (EGPA, Churg-Strauss syndrome)
  • nephritis and/or glomerulonephritis idiopathic pulmonary
  • Medicines containing the antibodies of the present invention for prevention/treatment of disease has a low toxicity, and can be administered as liquid medicine or medicine in an appropriate dosage forms to human or mammals (e.g., rats, rabbits, sheep, porcine, bovine, cats, dogs, and monkeys) orally or parenterally like intravascularly, subcutaneously, and intramuscularly.
  • human or mammals e.g., rats, rabbits, sheep, porcine, bovine, cats, dogs, and monkeys
  • the antibodies of the present invention may be administered alone, or may be administered as a suitable pharmaceutical composition.
  • the pharmaceutical composition used for administration may contain the antibodies of the present invention and a salt thereof and a pharmacologically acceptable carriers, diluents or excipients.
  • Such pharmaceutical compositions are provided as dosage forms suitable for oral or parenteral administration.
  • compositions for parenteral administration for example, injections, suppositories, etc. are used, and dosage forms for injections may include intravenous, subcutaneous, intradermal, intramuscular, drip injections and the like.
  • Such injections can be prepared according to known methods.
  • the injection can be prepared, for example, by dissolving, suspending or emulsifying the above-mentioned antibodies of the present invention or a salt thereof in a sterile aqueous or oil solutions usually used for injection.
  • aqueous solution for injection for example, physiological saline, isotonic solution containing glucose and other adjuvants, etc.
  • solubilizers such as alcohol (e.g., ethanol), polyalcohol (e.g., propylene glycol, polyethylene glycol), nonionic surfactants (e.g., polysorbate 80, HCO-50 (polyoxyethylene (50 mol) adduct of hydrogenated castor oil)) and the like.
  • alcohol e.g., ethanol
  • polyalcohol e.g., propylene glycol, polyethylene glycol
  • nonionic surfactants e.g., polysorbate 80, HCO-50 (polyoxyethylene (50 mol) adduct of hydrogenated castor oil)
  • an oily liquid for example, sesame oil, soybean oil and the like are used, and benzyl benzoate, benzyl alcohol and the like may be used in combination as a solubilizing agent.
  • the prepared injection solution is preferably filled in suitable ampoules.
  • Suppositories to be used for rectal administration may be prepared by mixing the above-mentione
  • compositions for oral administration may be in solid or liquid dosage forms, specifically tablets (including coated tablets, film coated tablets), pills, granules, powders, capsules (including soft capsules), syrups, emulsions, suspensions and the like.
  • Such compositions are prepared by known methods, and may contain carriers, diluents or excipients commonly used in the field of formulation.
  • carriers, diluents or excipients commonly used in the field of formulation.
  • a carrier or excipient for tablets for example, lactose, starch, sucrose and magnesium stearate are used.
  • parenteral or oral pharmaceutical compositions are conveniently prepared in dosage unit form adapted to the dose of the active ingredient.
  • dosage unit forms include, for example, tablets, pills, capsules, injections (ampules), suppositories.
  • the content of the antibodies is preferably 5 to 500 mg per dose unit dosage form, particularly 5 to 100 mg for an injection, and 10 to 250 mg for the other dosage form.
  • the dosage of the antibodies of the present invention is usually about 0.01 to 20 mg/kg body weight, preferably about 0.1 to 10 mg/kg body weight, more preferably about 0.1 to 5 mg/kg body weight, about 1 to 5 times a day, preferably 1 to 3 times a day, and is preferably administered by intravenous injection. In the case of other parenteral administration and oral administration, similar amounts can be administered. If the condition is particularly severe, the dose may be increased according to the condition.
  • composition described above may contain other active ingredients as long as the formulation with the above antibodies does not cause any undesirable interaction.
  • antibodies of the present invention may be used in combination with other agents.
  • the antibodies and other agents of the invention may be administered to the patient simultaneously or at different times.
  • a polynucleotide encoding the antibody of the present invention which is used as an active ingredient of the above-described prophylactic/therapeutic agent or immunoglobulin fragment composition.
  • the polynucleotide is a polynucleotide encoding a polypeptide containing an amino acid sequence identical or substantially identical to the amino acid sequence represented by SEQ ID NO: 4, and preferably comprising an amino acid sequence identical or substantially identical to the amino acid sequence represented by SEQ ID NO: 4, more preferably a polypeptide with completely the same sequence represented by SEQ ID NO: 4.
  • the polynucleotide encoding the antibody of the present invention may be DNA or RNA, or may be a DNA/RNA chimera, preferably DNA. Also, the polynucleotide may be double-stranded or single-stranded. When double stranded, it may be double stranded DNA, double stranded RNA or a hybrid of DNA: RNA. When single-stranded, it may be the sense strand (i.e., the coding strand) or the antisense strand (i.e., the non-coding strand).
  • Any DNA encoding the antibodies of the present invention contains, for example, a nucleotide sequence that hybridizes with DNA containing the nucleotide sequence shown in SEQ ID NO: 3 under high stringency conditions, and encoding a protein having substantially the same activity as the protein containing the amino acid sequence shown in SEQ ID NO: 4 can be used as DNA encoding the antibodies of the present invention.
  • DNA capable of hybridizing under the high stringency condition with the base sequence represented by SEQ ID NO: 3 for example, DNA with homology (homology) of about 50% or more, preferably about 60% or more, more preferably about 70% or more, much more preferably about 80% or more, particularly preferably about 90% or more, most preferably about 95% or more with the base sequence represented by SEQ ID NO: 3 are used.
  • homology homology of about 50% or more, preferably about 60% or more, more preferably about 70% or more, much more preferably about 80% or more, particularly preferably about 90% or more, most preferably about 95% or more with the base sequence represented by SEQ ID NO: 3 are used.
  • NCBI BLAST National Center for Biotechnology Information Basic Local Alignment Search Tool
  • the hybridization can be carried out according to a method known per se or a method analogous thereto, for example, a method described in Molecular Cloning 2nd ed. (J. Sambrook et al., Cold Spring Harbor Lab. Press, 1989).
  • a method described in Molecular Cloning 2nd ed. J. Sambrook et al., Cold Spring Harbor Lab. Press, 1989.
  • it can be performed according to the method described in the attached instruction manual. More preferably, it can be performed according to high stringency conditions.
  • High stringency conditions include, for example, sodium concentration of from about 19 to about 40 mM, preferably from about 19 to about 20 mM, the temperature is from about 50 to about 70° C. and preferably from about 60 to about 65° C. Particularly, the sodium salt concentration is preferably about 19 mM and the temperature is about 65° C.
  • One skilled in the art can easily adjust the desired stringency by changing the salt concentration of the hybridization solution, the temperature of the hybridization reaction, and the probe concentration, the length of the probe, the number of mismatches, and the time of hybridization reaction, the salt concentration of the washing liquid, the temperature of washing, and the like.
  • the polynucleotides preferably comprise a nucleotide sequence containing at least the 1st to 777th bases of SEQ ID NO: 3 (or a nucleotide sequence having the above-mentioned homology to the sequence).
  • antibodies of the present invention consisting of a polypeptide comprising an amino acid sequence identical or substantially identical (“substantially identical” means the same as described above) to the amino acid sequence comprising the first to 259th amino acids of SEQ ID NO: 4 are also preferred.
  • the invention also provides vectors containing the polynucleotide of the present invention as described above.
  • the vectors may be used as a preventive and/or therapeutic agent for infectious diseases or inflammatory diseases which itself contains as an active ingredient and may be used for producing the antibodies of the present invention.
  • vectors generally, plasmids or viral vectors carrying DNA encoding the antibody of the present invention are common.
  • Persons skilled in the art can appropriately produce vectors having a desired DNA by general genetic engineering techniques.
  • various commercially available vectors can be used.
  • the vectors of the present invention are useful for retaining the polynucleotides of the present invention in host cells or for expressing the antibodies of the present invention.
  • the polynucleotides of the present invention are usually carried (inserted) into suitable vectors and introduced into a host cell.
  • the vectors are not particularly limited as long as they stably hold the inserted DNA.
  • E. coli is used as host cells
  • pBluescript vector manufactured by Stratagene
  • expression vectors are particularly useful.
  • the expression vectors are not particularly limited as long as they are vectors that express polypeptides in test tubes, in E.
  • Insertion of the polynucleotide of the present invention into a vector can be carried out in a conventional manner, for example, by a ligase reaction using restriction sites, or by the In-fusion method (Takara Bio).
  • the host cells are not particularly limited, and various host cells may be used depending on the purpose.
  • Cells for expressing the antibodies of the present invention include bacterial cells (e.g., Streptococcus, Staphylococcus, E. coli, Streptomyces, Bacillus subtilis ), insect cells (e.g., Drosophila S2 and Spodoptera SF9), animal cells (eg, Examples: CHO, COS, HeLa, C127, 3T3, BHK, HEK 293, Bowes melanoma cells) and plant cells can be exemplified.
  • Vector introduction into host cells can be carried out by known methods, for example, calcium phosphate precipitation, electroporation (Current protocols in Molecular Biology edit. Ausubel et al. (1987) Publish. John Wiley & Sons. Sections 9.1-9.9), lipofection (GIBCO-BRL). and microinjection.
  • An appropriate secretion signal can be incorporated into the polypeptides of interest in order to secrete the polypeptides expressed in host cells into the lumen of the endoplasmic reticulum, into the periplasmic space, or into the extracellular environment. These signals may be endogenous or xenogenous to the polypeptides of interest.
  • Recovery of the polypeptides in the above production method involves recovering the culture medium when the polypeptide of the present invention is secreted into the culture medium. If the polypeptides of the present invention are produced intracellularly, the cells are first lysed and then the polypeptides are recovered.
  • the protein (APOA2) of the present invention is expressed in patients who are successfully treated with the antibody of the present invention.
  • inhibition of APOA2 activity can prevent or treat infectious diseases or inflammatory diseases. Therefore, a compound that inhibits the activity of APOA2 or its salt derivatives can be used as a preventive and/or therapeutic agent for infectious diseases or inflammatory diseases.
  • the protein (APOA2) of the present invention is useful as a reagent for screening compounds that inhibit the activity of the protein or its salt derivatives.
  • screening methods for compounds that inhibit the activity of the protein or its salt derivatives using the proteins of the present invention are provided.
  • screening methods are:
  • the protein of the present invention used in the screening method of the above (a-1) can be isolated and purified using the above-mentioned method of producing the protein of the present invention or its partial peptides.
  • the cells having the ability to produce the proteins of the present invention used in the screening method of the above (a-2) are not particularly limited as long as they are human or other warm-blooded animal cells naturally expressing them or biological samples (blood, tissues, organs, etc.) containing them.
  • biological samples blood, tissues, organs, etc.
  • they may be isolated from a living body and cultured in vitro.
  • a test substance is administered to a living body and then those biological samples are isolated from the body a predetermined time after the administration.
  • Examples of cells having the ability to produce the proteins of the present invention include various transformants produced by the above-mentioned genetic engineering techniques.
  • Test substances include, for example, proteins, peptides, non-peptide compounds, synthetic compounds, fermentation products, cell extracts, plant extracts, animal tissue extracts. These substances may be novel or known.
  • the activity of the protein of the present invention in the screening methods described above (a-1) and (a-2) can be measured by conventional methods.
  • the screening kits of the present invention include the proteins of the present invention or their partial peptides (hereafter, simply referred to as “the proteins of the present invention”).
  • the proteins of the present invention may be isolated/purified using any of the methods described above or may be provided in the form of cells (warm-blooded animal cells) producing them.
  • the screening kits of the present invention can also include the antibodies of the present invention in order to measure the expression amount of the proteins of the present invention in cells producing the proteins.
  • the screening kits can optionally include, in addition to the above, reaction buffers, blocking solutions, washing buffers, labeling reagents, label detection reagents, etc.
  • Substances inhibiting the activity of the protein of the present invention may be useful as low-toxic and safe medicines for preventing/treating infectious diseases or inflammatory diseases.
  • the compounds or their salt derivatives obtained by the screening methods or screening kits of the present invention are used as medicines for preventing/treating diseases described above, they can be formulated according to conventional means.
  • bases, amino acids and the like are indicated by abbreviations, they are based on abbreviations according to IUPAC-IUB Commission on Biochemical Nomenclature or common abbreviations in the field.
  • optical isomer of an amino acid it is assumed to indicate an L-isomer unless otherwise specified.
  • the protein of the present invention (APOA2) is a cause of infectious or inflammatory diseases.
  • inducers for infectious disease or inflammatory disease which comprises apolipoprotein A2 (APOA2) as an active ingredient, are provided.
  • methods of producing animal disease models for infectious diseases or inflammatory diseases comprising the step of administering apolipoprotein A2 (APOA2) to non-human, non-transgenic animals are provided.
  • APOA2 apolipoprotein A2
  • pathological model animals of infectious diseases or inflammatory diseases produced by the above-mentioned production method are also provided.
  • the method for producing pathological model animals of infectious diseases or inflammatory diseases comprises the step of administering apolipoprotein A2 (APOA2) to non-human, non-transgenic animals.
  • APOA2 apolipoprotein A2
  • the non-transgenic animals used in the method for producing pathological model animals according to the present embodiment refers to so-called wild-type animals which are not transgenic animals.
  • transgenic animals are animals into which foreign DNA has been introduced into the genome, and also includes a knockout animal or the like in which the function of a specific gene is not expressed by introducing an artificially engineered gene.
  • transgenic animals include both those with heritable germline DNA changes and those with non-heritable somatic DNA changes. That is, the pathological model animals concerning this form are produced based on wild type animals.
  • pathological animal models for infectious diseases or inflammatory diseases which were derived from wild-type animals.
  • any mammals except human can be used.
  • mice examples include rodents such as guinea pigs, mice, rats, and hamsters, non-human primates such as monkeys, chimpanzees and orangutans, rabbits, bovine, goats, sheep and porcine, but not limited to these. Rodents, particularly mice, are preferable.
  • the administration route of apolipoprotein A2 is not particularly limited. Whether orally or parenterally administered, any conventionally known administration routes, such as oral, intraperitoneal, intravenous, intraarterial, intramuscular, subcutaneous, intradermal, inhalation, intragastric, enteral, transdermal, etc. may be exemplified, but are not limited thereto.
  • Apolipoprotein A2 may be administered alone, or in combination with a suitable pharmaceutically acceptable carrier or diluent as long as the effect of apolipoprotein A2 (APOA2) is not impaired. Therefore, administration components may include various ingredients used for medicines and cosmetics, and it can be prepared and used as a preparation which has various dosage forms.
  • apolipoprotein A2 can be appropriately set according to the type of animal, strain, age, sex difference, etc.
  • mice FVB/N, Balb/c and C57BL/6, which are widely used as experimental animals, are preferable, but not limited thereto.
  • a single administration may be adequate if the desired symptoms develop in a single dose, and administration may be continued until the desired onset occurs.
  • the method for confirming whether or not the produced disease model animal has developed infectious diseases or inflammatory diseases can be performed based on known methods.
  • the onset of vasculitis as shown in examples described later, can be confirmed by examining the presence of pathological changes specific to the above-mentioned diseases by the observation of tissues, such as kidney, spleen and lung of a model animal under the microscope.
  • the onset may also be confirmed by evaluating the animal's Urinary Score or detecting or measuring various components in the blood collected from the animal.
  • pathological model animals produced by the preparation method of the pathological model animal of the infectious disease or inflammatory disease mentioned above are also provided.
  • the pathological model animals of the infectious disease or inflammatory disease provided by the present embodiment are non-transgenic animals, i.e., one produced by administering apolipoprotein A2 (APOA2) to wild-type animals, and indicate symptoms specific to the infectious disease or inflammatory disease. Therefore, the pathological model animals provided by the present embodiment can be used for analysis of the onset and pathogenesis mechanism of an infectious disease or an inflammatory disease, in particular, intractable vasculitis, and for evaluation and screening of treating/preventing methods for these diseases and therapeutic/preventive agents.
  • APOA2 apolipoprotein A2
  • the screening method of the preventive and/or therapeutic agent of an infectious disease or an inflammatory disease is also provided.
  • a test substance which is a candidate for a preventive and/or therapeutic agent for an infectious disease or an inflammatory disease can be administered to the above-described pathological model animal to evaluate improvement and elimination of symptoms.
  • any test substances may be used as long as they are candidate substances for infectious disease or inflammatory disease, regardless of whether they are in vivo substances, genetically modified substances, chemically synthesized compounds, etc., and the test substances are not limited to these.
  • they may be administered alone, but may be formulated and administered in combination with a suitable pharmaceutically acceptable carrier or diluent.
  • Administration of the test substances may be performed according to any known administration routes and may be either oral or parenteral administration. Therefore, oral, intraperitoneal, intravenous, intraarterial, intramuscular, subcutaneous, intradermal, inhalation, intragastric, enteral, transdermal, etc., can be exemplified, but not limited thereto. It may be set in consideration of the properties of the test substances, particularly pharmacokinetic properties and solubility. The dose, frequency and period of administration of the test substances can be appropriately set according to the type of animal, strain, age, sex, etc., and the type of test substances.
  • Evaluation of the improvement and elimination of symptoms in the method for screening a preventive and/or therapeutic agent for an infectious disease or an inflammatory disease in this embodiment can be performed in the same way as those employed in the various indexes described above and the examples described later. Then, when the administration of the test substance improves or eliminates the symptoms, the test substance can be evaluated as a preventive and/or therapeutic agent for infectious disease or inflammatory disease.
  • a negative control a pathological model animal to which a formulation having the same composition except the test substance is administered or no test substance is administered can be used. The negative control animals can be compared to the animals receiving the test substance for improvement and elimination of symptoms. And, for example, when the symptom in the animal to which the test substance is administered is improved over the negative control animal to which the test substance is not administered, the test substance can be evaluated as an agent for preventing and/or treating infectious disease or inflammatory disease.
  • VasSF recombinant human ScFv
  • the present inventors searched for the target antigen of VasSF by using each of the following three techniques, and finally, the present inventors succeeded in identifying target antigens by narrowing down candidate substances using tosylactivated Dynabeads® and MS/MS method.
  • the present inventors prepared plasma from healthy individuals as a sample, treated it with ProteoSeekTM Albumin/IgG Removal Kit (PIERCE), and then attempted to identify the target antigen by SDS-PAGE and Western blotting using the following primary to quaternary antibodies.
  • PIERCE ProteoSeekTM Albumin/IgG Removal Kit
  • FIG. 1B The photograph on the left of FIG. 1B shows the result of Coomassie brilliant blue (CBB) staining, and the photograph on the right shows the result of Western blotting. Although the reaction was found at the circled site on the right side of the photograph, a large number of target antigen candidate spots overlapped. Therefore, this experiment did not lead to the identification of the target antigen.
  • CBB Coomassie brilliant blue
  • HiTrap NHS-activated HP column (GE Healthcare) with a bed volume of 1 mL was prepared. Five milliliters (2 mg) of a 0.4 mg/mL solution of VasSF was weighed out and concentrated about 40-fold using Amicon 10K. It was then diluted with coupling buffer and allowed to bind to the column prepared above.
  • FIG. 1C The staining results of the sample-bound column are shown in FIG. 1C .
  • CBB staining left in FIG. 1C
  • silver staining middle in FIG. 1C
  • Western blotting right in FIG. 1C
  • a thick band was observed around 10 kDa.
  • the identification of the target antigen was not achieved even in this experiment.
  • ligand-coated beads were prepared by mixing and reacting Dynabeads Tosylactivated (25 mg) in 0.1 M sodium borate buffer using 1 mg of VasSF as a ligand. The coated beads were then washed 5 times with PBS (containing 0.05% Tween 20).
  • This sample was mixed with 25 mg of the above coated beads and allowed to react at room temperature with rotation for 1 hour. After removing plasma, the beads were washed three times with PBS (containing 0.05% Tween 20).
  • the fraction bound to the coated beads was then eluted with elution buffer (0.1 M glycine-HCl, pH 2.7) and then neutralized with 1/10 volume of neutralization buffer (1 M Tris HCl, pH 9.0).
  • elution buffer 0.1 M glycine-HCl, pH 2.7
  • neutralization buffer 1 M Tris HCl, pH 9.0
  • the fraction eluted in this manner was subjected to SDS-PAGE.
  • a photograph showing the result of silver staining of this SDS-PAGE is shown in FIG. 2 .
  • the band (spot) of about 10 kDa was cut out.
  • IgG was separated from plasma using protein G column.
  • the obtained polyclonal antibodies were administered to spontaneous onset model mice (SCG/Kj mice) of intractable vasculitis to determine the therapeutic effect of each antibody.
  • the anti-APOA2 polyclonal antibody that showed remarkable therapeutic effect in embodiment 3 described above was used by dissolving it in 1.5% D-mannitol solution containing 0.45% glycine (Gly) and 0.9% sodium chloride.
  • anti-APOA2 polyclonal antibodies are administered intraperitoneally to a 10-week-old SCG/Kj mice at a prescription of 10-40 mg/kg/day for 5 days by intraperitoneal administration (ip), and when the mice became 13-week-old, they were euthanatized with CO 2 gas.
  • MPO-ANCA myeloperoxidase-specific anti-neutrophil cytotoxic antibody
  • serum which is an index of vasculitis, spleen weight, leukocyte count in peripheral blood, lymphocyte count, monocyte count, granulocyte count, the number of neutrophils and the number of platelets were measured, and the treatment effect was determined based on the results.
  • SCG/Kj mice are kept in an environment set to a light/dark cycle of 14L10D (light period 5:00-19:00, dark period 19:00-5:00), temperature 23 ⁇ 1° C., and humidity 50 ⁇ 5%.
  • Feeds were radiation-sterilized CRF1 for small rodents (for special breeding, Oriental Yeast Co., Ltd.) until the experiment, and radiation-sterilized FR-2 for small rodents (for general breeding, Funabashi Farm) from the day before administration.
  • Drinking water was clean water from a reverse osmosis (RO) drinking water treatment apparatus and supplemented with chlorine at a concentration of 0.3 to 2.0 ppm. Feeds and water were given ad libitum.
  • the animal facility was supplied with clean air through HEPA filters and was limited to SPF animals only.
  • mice were subjected to a weekly urinalysis test from about 3 weeks ago, and divided into experimental groups based on the weight and the value of the urine occult blood and urine score on the day of administration.
  • the sample was injected ip at 0.1 to 400 mg/kg for 5 days
  • Solvent stabilizer: D mannitol, glycine, PBS
  • the dose was calculated from the final measured body weight and adjusted to 0.33 mL per animal. After administration, weight measurement and urinalysis were performed twice a week.
  • the residual blood was centrifuged and the serum was stored at ⁇ 80° C. Then, it was sent to A-CLIP Research Institute, Inc. (for MPO-ANCA and cytokine (Bio-Plex) measurement).
  • Kidney After taking pictures, 5 mm squares were transferred into RNA Later for RNA, and the remaining tissue was divided and immersed into 10% neutral buffer formalin solution.
  • the main target tissues of intractable vasculitis which is an autoimmune disease, are kidney, lung and spleen.
  • kidney, lung and spleen are kidney, lung and spleen.
  • FIG. 4A a microscopic image in which Bowman's cavity of kidney glomerulus was formed is shown.
  • FIG. 4B microscopic images of kidney glomeruli of the untreated (Solvent-administered) group of the model mouse SCG/Kj formed crescents and renal dysfunction occurred (circled in FIG. 4B ).
  • FIG. 4C The same situation was found for negative control administration
  • SCG/Kj model mice in the non-treated (Solvent administration) group had heavier spleens than C57BL/6 healthy mice.
  • administration of the anti-APOA2 polyclonal antibody to SCG/Kj model mice reduced the spleen weight, although it did not fall to the value of the spleen weight of healthy mice.
  • Administration of a molecule different from VasSF (Different Mol) did not reduce the spleen weight.
  • IL-1alpha Out of IL-1alpha, IL-1beta, IL-2, IL-3, IL-4, IL-5, IL-6, IL-9, IL-10, IL-12p40, IL-12p70, IL-13, IL-17, Eotaxin, G-CSF, GM-CSF, IFN-gamma, KC, MCP-1, MIP-1 alpha, MIP-1 beta, RANTES, TNF-alpha, IL-15, IL-18, FGF-basic, LIF, M-CSF, MIG, MIP-2, PDGF-bb, VEGF, IL6sR, and IL-23, almost all cytokine/chemokine levels increased in non-treated (Solvent administration) group of SCG/Kj model mice.
  • IL-1a interleukin-1 alpha IL-1b: interleukin-1 beta IL-2: interleukin-2 IL-3: interleukin-3 IL-4: interleukin-4 IL-5: interleukin-5 IL-6: interleukin-6 IL-9: interleukin-9 (interleave-9) IL-10: interleukin-10 (interleave-10) IL-12p40: interleukin-12 subunit p40 IL-12p70: interleukin-12 subunit p70 IL-13: interleukin-13 IL-17: interleukin-17 EOTAXIN: eotaxin G-CSF: granulocyte colony stimulating factor GM-CSF: granulocyte/macrophage colony stimulating factor IFN-g: interferon gamma Kc: keratinocytes derived chemokines MCP-1: monocyte chemotaxis protein-1 MIP-1a: macrophage inflammatory protein-1 alpha MIP-1b: macrophage
  • Intractable vasculitis is an autoimmune disease, and SCG/Kj model mice had splenomegaly and increased serum levels of autoantibodies MPO-ANCA, anti-moesin antibodies, inflammatory cytokines and chemokines.
  • anti-APOA2 Polyclonal antibodies directed these towards normalization. From these results, the influence on leukocytes in peripheral blood was examined. There were no changes in the total white blood cell count (WBC), the number of lymphocytes (LYM), and the number of monocytes (MON) ( FIGS. 10A, and 10B ). However, the neutrophil count (GRA) improved to almost the value of healthy mice by administration of the anti-APOA2 polyclonal antibody ( FIG. 10B ).
  • vasculitis is an autoimmune disease
  • microscopic images of lungs which are affected organs as well as kidneys, were examined.
  • vasculitis, hemorrhage, and granulation appeared in part in the lung tissue ( FIGS. 11B and 11C ).
  • administration of the anti-APOA2 polyclonal antibody improved the lung tissue close to normal ( FIGS. 11A and 11D ).
  • the pET-based vector which is a vector generally used for the purpose of strong expression of recombinant proteins in E. coli , can be up-regulated by the T7 promoter under the control of the LacZ operon. Therefore, in this experiment, the present inventors attempted to introduce the URq01_OptEcoli sequence into a pET-based vector.
  • the insert sequence (coding region: region of SEQ ID NO: 3) was PCR-amplified from the plasmid pTAC2-URq01_OptEcoli constructed above to prepare an integration fragment.
  • the nucleotide sequences of the primers used for this preparation are as follows (underlined part is a sequence homologous to the end of the cloning site of the pET32 vector).
  • the vector was amplified by performing inverse PCR.
  • the nucleotide sequence of the primers used in this process are as follows (underlined part is a sequence homologous to the His-tag end of the insert sequence).
  • the integration fragment and the linearized pET32 vector amplified above were fused using the In-Fusion method (Takara Bio) to integrate the integration fragment at the integration site of the T7 promoter control region of the pET32 vector ( FIG. 14 ).
  • a transformant was prepared by introducing the above vector into E. coli host DE32 using a heat shock method.
  • the insertion sequence was confirmed from 10 colonies by PCR amplification of the insertion sequence using a primer set in the frame T7 promoter region of the pET32 vector and a primer in the T7 terminator region.
  • the nucleotide sequences of the primers used in this process are as follows.
  • the recombinant protein (composed of SEQ ID NO: 4, also referred to herein as “VasAP”) was purified from the culture of host E. coli DE32 by the following procedure.
  • protein purification can be carried out using a general purification method. Examples of the purification method include immobilized metal ion affinity chromatography, fractionation with an ion exchange column, chromatography with a positive ion exchange resin such as DEAE, and gel filtration.
  • E. coli containing a target clone was cultured at 10° C. for 18 to 48 hours.
  • FIGS. 15A and 15B are photographs showing the results of gel filtration HPLC profiles
  • FIG. 15C is a photograph showing the results of electrophoresis and Western blotting.
  • FIG. 16 administration of the VasAP antibody improved crescent formation in the SCG/Kj model mice ( FIG. 16A ), and urinary findings of antibody-treated mice showed lower values compared to ones of Different Mol treated mice ( FIG. 16B ).
  • VasAP antibody decreased the serum levels of TNF-alpha, G-CSF, RANTES, IFN-gamma, IL-5, and IL-10, indicating the efficacy.
  • VasAP antibody did not significantly improve whole white blood cell counts (WBC), lymphocytes (LYM) ( FIG. 20A ), but the numbers of monocytes (MON) and neutrophils (GRA) reduced to nearly the value of the healthy mice ( FIG. 20B ).
  • mice Four female BALB/cCR mice (10-week old) were prepared. Three of them were assigned to experimental group and one to control group.
  • APOA2 protein (BTI: BT-928, lot: 9280413) was prepared for this embodiment.
  • the protein (lmg) was dissolved in 1 ml of saline (#412190, produced by Nippon Pharmaceutical Industry Co. Ltd.). After addition of lml of the saline solution, the protein solution was uniformly mixed to prepare 0.5 mg/mL test solution.
  • test solution prepared above were intraperitoneally administered to three mice of the experimental group (0.2 ml per mice; the dose of the test protein was 0.1 mg per mouse). On the other hand, in the control group, 0.2 ml of saline (solvent) was administered.
  • mice of the experimental group administered with the above-described test solution were evaluated by urinary score up to 44 days after the administration of test solution (containing a test protein).
  • the results are shown in FIG. 21 .
  • the graph shown in FIG. 21 is a relative value when the value on the day of administration (day 0) is 100 as the change in urinary scores (vertical axis; arithmetic mean value of 3 animals) with respect to days after administration (horizontal axis).
  • the spleen tissue and lung tissue of each mouse were observed in the same manner as in Embodiment 4 described above. The results are shown in FIG. 23 .
  • the crescent formation rate in the kidney glomeruli was significantly increased as compared to the mice of the control group.
  • the microscopic image shown in FIG. 23 the microscopic image of the kidney glomeruli of the mice of the experimental group formed crescents, and renal dysfunction occurred.
  • the “enlarged view” in the lower part of FIG. 23 is an enlarged view of a square-enclosed portion of the microscope image in the upper part.
  • an inflammatory disease here, vasculitis
  • APOA2 protein a wild-type non-human animal
  • sequence id numbers (SEQ ID NO.) in the sequence table of the present specification show the following sequences.
  • the nucleotide sequence of DNA encoding human APOA2 (containing CDS and termination codon).
  • the nucleotide sequence of DNA encoding one (VasAP) of the antibodies in the present invention (including CDS and a termination codon).
  • the nucleotide sequence of a PCR primer (pET32_Rq01OptEc_infF(Tm)).
  • the nucleotide sequence of a PCR primer (Rq01_OptEc_cds_6HisStpcmpR).
  • the nucleotide sequence of a PCR primer (His6StpCmp_pET32_110-129F).
  • the nucleotide sequence of a PCR primer (pET32vbRev693-712).
  • the nucleotide sequence of a PCR primer (Rq01_OptEc_cds_F).

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Engineering & Computer Science (AREA)
  • General Health & Medical Sciences (AREA)
  • Organic Chemistry (AREA)
  • Biomedical Technology (AREA)
  • Immunology (AREA)
  • Medicinal Chemistry (AREA)
  • Molecular Biology (AREA)
  • Animal Behavior & Ethology (AREA)
  • Public Health (AREA)
  • Veterinary Medicine (AREA)
  • Urology & Nephrology (AREA)
  • Biochemistry (AREA)
  • Genetics & Genomics (AREA)
  • Biophysics (AREA)
  • Zoology (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • General Chemical & Material Sciences (AREA)
  • Pathology (AREA)
  • Hematology (AREA)
  • Rheumatology (AREA)
  • Biotechnology (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Endocrinology (AREA)
  • Cell Biology (AREA)
  • Toxicology (AREA)
  • Physics & Mathematics (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Microbiology (AREA)
  • General Physics & Mathematics (AREA)
  • Epidemiology (AREA)
  • Food Science & Technology (AREA)
  • Environmental Sciences (AREA)
  • Diabetes (AREA)
  • Gastroenterology & Hepatology (AREA)
  • Analytical Chemistry (AREA)
US16/481,241 2017-01-27 2018-01-26 Prophylactic and/or therapeutic agent of infectious diseases or inflammatory diseases Abandoned US20190367600A1 (en)

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
JP2017013486 2017-01-27
JP2017-013486 2017-01-27
PCT/JP2018/002576 WO2018139608A1 (ja) 2017-01-27 2018-01-26 感染性疾患または炎症性疾患の予防および/または治療剤

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
PCT/JP2018/002576 A-371-Of-International WO2018139608A1 (ja) 2017-01-27 2018-01-26 感染性疾患または炎症性疾患の予防および/または治療剤

Related Child Applications (1)

Application Number Title Priority Date Filing Date
US17/655,687 Division US20220213180A1 (en) 2017-01-27 2022-03-21 Prophylactic and/or therapeutic agent of infectious diseases or inflammatory diseases

Publications (1)

Publication Number Publication Date
US20190367600A1 true US20190367600A1 (en) 2019-12-05

Family

ID=62978497

Family Applications (2)

Application Number Title Priority Date Filing Date
US16/481,241 Abandoned US20190367600A1 (en) 2017-01-27 2018-01-26 Prophylactic and/or therapeutic agent of infectious diseases or inflammatory diseases
US17/655,687 Pending US20220213180A1 (en) 2017-01-27 2022-03-21 Prophylactic and/or therapeutic agent of infectious diseases or inflammatory diseases

Family Applications After (1)

Application Number Title Priority Date Filing Date
US17/655,687 Pending US20220213180A1 (en) 2017-01-27 2022-03-21 Prophylactic and/or therapeutic agent of infectious diseases or inflammatory diseases

Country Status (9)

Country Link
US (2) US20190367600A1 (ko)
EP (2) EP3581204A4 (ko)
JP (1) JP7126658B2 (ko)
KR (1) KR102581165B1 (ko)
CN (1) CN110267680B (ko)
AU (1) AU2018213716A1 (ko)
CA (1) CA3051826A1 (ko)
SG (2) SG10202108219QA (ko)
WO (1) WO2018139608A1 (ko)

Families Citing this family (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20230144545A1 (en) * 2019-10-30 2023-05-11 National University Corporation Okayama University Prophylactic and/or therapeutic agent for inflammatory pulmonary disease
WO2024004580A1 (ja) * 2022-06-30 2024-01-04 国立大学法人千葉大学 血管炎への罹患状態またはcovid-19の重症化リスクを検査する方法、検査用キット、コンパニオン診断薬、および検査用マーカー

Family Cites Families (27)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US164290A (en) 1875-06-08 Improvement in bedsteads
US4816567A (en) 1983-04-08 1989-03-28 Genentech, Inc. Recombinant immunoglobin preparations
US5225539A (en) 1986-03-27 1993-07-06 Medical Research Council Recombinant altered antibodies and methods of making altered antibodies
US5530101A (en) 1988-12-28 1996-06-25 Protein Design Labs, Inc. Humanized immunoglobulins
CA2078539C (en) 1991-09-18 2005-08-02 Kenya Shitara Process for producing humanized chimera antibody
JP2000014383A (ja) 1998-07-03 2000-01-18 Igaku Seibutsugaku Kenkyusho:Kk ヒト化抗体
FR2801407B1 (fr) 1999-11-22 2002-04-12 Roger Felix Sejalon Bracelet porte etiquette inviolable a tracabilite continue
JP2004081199A (ja) 2002-06-25 2004-03-18 Sekisui Chem Co Ltd 組換え抗体の製造方法
US9028822B2 (en) * 2002-06-28 2015-05-12 Domantis Limited Antagonists against TNFR1 and methods of use therefor
JP3965147B2 (ja) 2003-11-04 2007-08-29 ヤンマー株式会社 ロータリ耕耘装置の爪軸脱着構造
WO2005055810A2 (en) * 2003-12-05 2005-06-23 The Cleveland Clinic Foundation Risk markers for cardiovascular disease
GB0701626D0 (en) 2007-01-22 2007-03-07 Cambridge Entpr Ltd Methods and biomarkers for diagnosing and monitoring psychotic disorders
AU2008229385A1 (en) * 2007-03-19 2008-09-25 Sirtris Pharmaceuticals, Inc. Biomarkers of sirtuin activity and methods of use thereof
WO2009044723A1 (ja) 2007-10-04 2009-04-09 Biomarker Science Co., Ltd. インターフェロン療法の効果予測方法及び予測用キット
GB0720976D0 (en) * 2007-10-26 2007-12-05 Medical Res Council Treatment of inflammatory disease
US8225183B2 (en) 2008-09-30 2012-07-17 Lsi Corporation Methods and apparatus for selective data retention decoding in a hard disk drive
WO2011143308A2 (en) * 2010-05-11 2011-11-17 The General Hospital Corporation Biomarkers of hemorrhagic shock
US9244079B2 (en) 2010-09-22 2016-01-26 National University Corporation Chiba University Testing method and testing reagent for angiitis
US10208131B2 (en) 2011-12-19 2019-02-19 National University Corporation Chiba University Artificially produced polyclonal immunoglobulin preparation
US10196457B2 (en) * 2011-12-19 2019-02-05 National University Corporation Chiba University Artificial immunoglobulin fragment composition
JP6519056B2 (ja) * 2013-10-01 2019-05-29 東レ株式会社 膵腫瘍の検出方法、抗体及び膵腫瘍検出用キット
JP5685672B1 (ja) 2014-07-10 2015-03-18 株式会社山岡製作所 樹脂成形方法
JP6677655B2 (ja) * 2015-01-26 2020-04-08 東レ株式会社 胆道癌の検出方法及び胆道癌検出用キット
JP6670129B2 (ja) 2015-02-27 2020-03-18 国立大学法人千葉大学 人工免疫グロブリン断片組成物
JP6797107B2 (ja) * 2015-03-02 2020-12-09 東レ株式会社 膵臓機能異常の検出方法及び検出用キット
US9895848B2 (en) 2015-04-22 2018-02-20 The Boeing Company Systems and tooling for manufacturing composite parts and related methods
JP6453199B2 (ja) 2015-10-16 2019-01-16 光洋機械工業株式会社 センタレス研削盤

Also Published As

Publication number Publication date
EP3581204A4 (en) 2020-12-16
WO2018139608A1 (ja) 2018-08-02
EP3581204A1 (en) 2019-12-18
SG10202108219QA (en) 2021-08-30
JP7126658B2 (ja) 2022-08-29
CN110267680B (zh) 2023-12-19
US20220213180A1 (en) 2022-07-07
KR102581165B1 (ko) 2023-09-21
EP4317431A3 (en) 2024-05-22
CA3051826A1 (en) 2018-08-02
KR20190108152A (ko) 2019-09-23
SG11201906968YA (en) 2019-08-27
CN110267680A (zh) 2019-09-20
AU2018213716A1 (en) 2019-08-15
JPWO2018139608A1 (ja) 2020-01-09
EP4317431A2 (en) 2024-02-07

Similar Documents

Publication Publication Date Title
US20220213180A1 (en) Prophylactic and/or therapeutic agent of infectious diseases or inflammatory diseases
US7122374B1 (en) Amyloid beta-protein 3(pE)-42 antibodies and uses thereof
NZ586600A (en) Anti macrophage migration inhibitory factor antibodies
US9733244B2 (en) Means and methods for diagnosing and treating multiple sclerosis
JP2016507524A (ja) 免疫グロブリンaのレベルを増大するための方法
US10975144B2 (en) CDNA encoding a monoclonal anti-IL-32 antibody and methods for the production of the antibody
JP2021519088A (ja) 抗cd27抗体、その抗原結合フラグメント及びその医学的使用
KR20160027171A (ko) 인간 항-ifn-알파 항체
EP1924605B1 (en) Compositions and methods for diagnosing and treating an inflammation
EP4293360A1 (en) Adult still's disease inspection method and inspection kit
KR101836756B1 (ko) Il-17a를 표적화하는 백신
JP6962514B2 (ja) 酵母由来組換えヒトgm−csfに特異的に結合するモノクローナル抗体
NZ717188B2 (en) Means and methods for diagnosing and treating multiple sclerosis
NZ615912B2 (en) Means and methods for diagnosing and treating multiple sclerosis

Legal Events

Date Code Title Description
AS Assignment

Owner name: NATIONAL INSTITUTES OF BIOMEDICAL INNOVATION, HEAL

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:SUZUKI, KAZUO;KAMEOKA, YOSUKE;YAMAKAWA, YOSHIO;AND OTHERS;SIGNING DATES FROM 20190711 TO 20190719;REEL/FRAME:049871/0522

Owner name: A-CLIP INSTITUTE, CO., LTD., JAPAN

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:SUZUKI, KAZUO;KAMEOKA, YOSUKE;YAMAKAWA, YOSHIO;AND OTHERS;SIGNING DATES FROM 20190711 TO 20190719;REEL/FRAME:049871/0522

STPP Information on status: patent application and granting procedure in general

Free format text: NON FINAL ACTION MAILED

STPP Information on status: patent application and granting procedure in general

Free format text: RESPONSE TO NON-FINAL OFFICE ACTION ENTERED AND FORWARDED TO EXAMINER

STPP Information on status: patent application and granting procedure in general

Free format text: FINAL REJECTION MAILED

STPP Information on status: patent application and granting procedure in general

Free format text: DOCKETED NEW CASE - READY FOR EXAMINATION

STPP Information on status: patent application and granting procedure in general

Free format text: NON FINAL ACTION MAILED

STCB Information on status: application discontinuation

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