WO2010064456A1 - Anticorps anti-nr10, et leurs utilisations - Google Patents

Anticorps anti-nr10, et leurs utilisations Download PDF

Info

Publication number
WO2010064456A1
WO2010064456A1 PCT/JP2009/054941 JP2009054941W WO2010064456A1 WO 2010064456 A1 WO2010064456 A1 WO 2010064456A1 JP 2009054941 W JP2009054941 W JP 2009054941W WO 2010064456 A1 WO2010064456 A1 WO 2010064456A1
Authority
WO
WIPO (PCT)
Prior art keywords
seq
antibody
variable region
amino acid
chain variable
Prior art date
Application number
PCT/JP2009/054941
Other languages
English (en)
Japanese (ja)
Inventor
太一 倉持
恵子 糟谷
創平 大山
浩行 角田
智之 井川
達彦 橘
宙丈 白岩
江崎圭子
Original Assignee
中外製薬株式会社
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
Priority claimed from PCT/JP2008/072152 external-priority patent/WO2009072604A1/fr
Priority to JP2009054941A priority Critical patent/JP2010210772A/ja
Application filed by 中外製薬株式会社 filed Critical 中外製薬株式会社
Priority to TW098141125A priority patent/TWI432209B/zh
Priority to EP09830463.7A priority patent/EP2354161B1/fr
Priority to KR1020107014700A priority patent/KR101271323B1/ko
Priority to BRPI0906478A priority patent/BRPI0906478B8/pt
Priority to EP15172595.9A priority patent/EP2949672A1/fr
Priority to US12/809,138 priority patent/US8575317B2/en
Priority to PL09830463T priority patent/PL2354161T3/pl
Priority to SI200931302T priority patent/SI2354161T1/sl
Priority to CN200980106176.2A priority patent/CN101952318B/zh
Priority to CA2710264A priority patent/CA2710264C/fr
Priority to MX2010007936A priority patent/MX2010007936A/es
Priority to PT98304637T priority patent/PT2354161E/pt
Priority to AU2009323249A priority patent/AU2009323249B2/en
Priority to RU2010126078/10A priority patent/RU2487136C2/ru
Priority to HUE09830463A priority patent/HUE025958T2/en
Priority to ES09830463.7T priority patent/ES2552690T3/es
Priority to MYPI2010002989A priority patent/MY154715A/en
Priority to DK09830463.7T priority patent/DK2354161T3/en
Priority to PCT/JP2009/070376 priority patent/WO2010064697A1/fr
Priority to JP2010507563A priority patent/JP5139517B2/ja
Publication of WO2010064456A1 publication Critical patent/WO2010064456A1/fr
Priority to HK11105170.8A priority patent/HK1151048A1/xx
Priority to US14/047,316 priority patent/US9399680B2/en
Priority to HRP20151215TT priority patent/HRP20151215T1/hr

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
    • C07K16/28Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants
    • C07K16/2866Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants against receptors for cytokines, lymphokines, interferons
    • 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
    • C07K16/28Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/395Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum
    • 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]
    • 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
    • G01N33/68Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving proteins, peptides or amino acids
    • G01N33/6863Cytokines, i.e. immune system proteins modifying a biological response such as cell growth proliferation or differentiation, e.g. TNF, CNF, GM-CSF, lymphotoxin, MIF or their receptors
    • G01N33/6869Interleukin
    • 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
    • C07K2317/20Immunoglobulins specific features characterized by taxonomic origin
    • C07K2317/24Immunoglobulins specific features characterized by taxonomic origin containing regions, domains or residues from different species, e.g. chimeric, humanized or veneered
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/50Immunoglobulins specific features characterized by immunoglobulin fragments
    • C07K2317/56Immunoglobulins specific features characterized by immunoglobulin fragments variable (Fv) region, i.e. VH and/or VL
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/50Immunoglobulins specific features characterized by immunoglobulin fragments
    • C07K2317/56Immunoglobulins specific features characterized by immunoglobulin fragments variable (Fv) region, i.e. VH and/or VL
    • C07K2317/565Complementarity determining region [CDR]
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/50Immunoglobulins specific features characterized by immunoglobulin fragments
    • C07K2317/56Immunoglobulins specific features characterized by immunoglobulin fragments variable (Fv) region, i.e. VH and/or VL
    • C07K2317/567Framework region [FR]
    • 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
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/90Immunoglobulins specific features characterized by (pharmaco)kinetic aspects or by stability of the immunoglobulin
    • C07K2317/92Affinity (KD), association rate (Ka), dissociation rate (Kd) or EC50 value
    • 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
    • G01N2333/705Assays involving receptors, cell surface antigens or cell surface determinants
    • G01N2333/715Assays involving receptors, cell surface antigens or cell surface determinants for cytokines; for lymphokines; for interferons
    • G01N2333/7155Assays involving receptors, cell surface antigens or cell surface determinants for cytokines; for lymphokines; for interferons for interleukins [IL]

Definitions

  • the present invention relates to an anti-NR10 antibody and a pharmaceutical composition containing the anti-NR10 antibody.
  • cytokines The existence of many cytokines is known as a humoral factor involved in the proliferation and differentiation of various cells or the activation of the functions of differentiated and mature cells.
  • cells stimulated with cytokines produce other cytokines, and a network of a plurality of cytokines is formed.
  • Biological homeostasis is maintained on a delicate balance by the network coordinating with each other.
  • Many immunoinflammatory diseases are thought to be caused by the breakdown of these cytokine networks, and anti-cytokine therapy using monoclonal antibodies has attracted attention.
  • anti-TNF antibodies and anti-IL-6 receptor antibodies have been highly effective clinically.
  • the compensatory pathway works in actual pathological conditions, and if only one cytokine such as IL-4 is blocked, a therapeutic effect cannot be obtained and it fails.
  • NR10 forms a heterodimer with the oncostatin M receptor (OSMR) and functions as a receptor for IL-31 (Non-patent Document 1).
  • OSMR oncostatin M receptor
  • IL-31 it has been reported that transgenic mice overexpressing IL-31 spontaneously develop pruritic dermatitis (Patent Document 2).
  • Antibodies that bind to NR10 and inhibit the binding of IL-31 and NR10 are thought to be effective in the treatment of inflammatory diseases, but in order to use anti-NR10 antibodies clinically, antibodies with low immunogenicity are required. It is said. Further, in order to exert a high therapeutic effect, an antibody having a high binding activity or neutralizing activity to NR10 is desired.
  • WO00 / 75314 WO03 / 060090 IL-31 is associated with cutaneous lymphocyte antigen-positive skin homing T cells in patients with atopic dermatitis., J Allergy Clin Immunol. 2006 Feb; 117 (2): 418-25.
  • the present invention has been made in view of the above background, and an object of the present invention is to provide an anti-NR10 antibody and a pharmaceutical composition containing the anti-NR10 antibody.
  • the present inventors have conducted intensive research to solve the above problems.
  • the present inventors have succeeded in obtaining an anti-NR10 antibody having effective neutralizing activity against NR10.
  • the present inventors succeeded in humanizing an antibody while maintaining the activity of the antibody.
  • the inventors have succeeded in producing antibodies with improved pharmacokinetics, enhanced antigen binding activity, improved stability, and / or reduced risk of immunogenicity.
  • the antibody is useful as a therapeutic agent for inflammatory diseases.
  • the present invention relates to an anti-NR10 antibody and a pharmaceutical composition containing the anti-NR10 antibody, and more specifically includes the following inventions [1] to [15].
  • an antibody having the heavy chain variable region described in SEQ ID NO: 4 (2) an antibody having the heavy chain variable region described in SEQ ID NO: 4; (3) An antibody having a light chain variable region comprising CDR1 having the amino acid sequence set forth in SEQ ID NO: 5, CDR2 having the amino acid sequence set forth in SEQ ID NO: 6, and CDR3 having the amino acid sequence set forth in SEQ ID NO: 7.
  • an antibody having the light chain variable region set forth in SEQ ID NO: 8 an antibody having the heavy chain variable region of (1) and the light chain variable region of (3), (6) an antibody having the heavy chain variable region of (2) and the light chain variable region of (4), (7)
  • the antibody according to any one of (1) to (6), wherein one or more amino acids are substituted, deleted, added and / or inserted, and any one of (1) to (6) An antibody having an activity equivalent to that of the antibody described in 1.
  • the anti-NR10 antibody according to any one of the following (1) to (8); (1) An antibody having a heavy chain variable region comprising CDR1 having the amino acid sequence of SEQ ID NO: 9, CDR2 having the amino acid sequence of SEQ ID NO: 10, and CDR3 having the amino acid sequence of SEQ ID NO: 11.
  • the anti-NR10 antibody according to any one of (1) to (8) below: (1) An antibody having a heavy chain variable region comprising CDR1 having the amino acid sequence set forth in SEQ ID NO: 25, CDR2 having the amino acid sequence set forth in SEQ ID NO: 26, and CDR3 having the amino acid sequence set forth in SEQ ID NO: 27 , (2) an antibody having the heavy chain variable region set forth in SEQ ID NO: 28, (3) An antibody having a light chain variable region comprising CDR1 having the amino acid sequence set forth in SEQ ID NO: 29, CDR2 having the amino acid sequence set forth in SEQ ID NO: 30 and CDR3 having the amino acid sequence set forth in SEQ ID NO: 31 , (4) an antibody having the light chain variable region set forth in SEQ ID NO: 32, (5) an antibody having the heavy chain variable region of (1) and the light chain variable region of (3), (6) an antibody having the heavy chain variable region of (2) and
  • the antibody variable region or antibody according to any one of (1) to (20) below (1) heavy chain variable region (H17) comprising CDR1 of SEQ ID NO: 196, CDR2 of SEQ ID NO: 197, CDR3 of SEQ ID NO: 11; (2) a heavy chain variable region (H19) comprising CDR1 of SEQ ID NO: 176, CDR2 of SEQ ID NO: 197, CDR3 of SEQ ID NO: 11; (3) heavy chain variable region (H28, H42) comprising CDR1 of SEQ ID NO: 196, CDR2 of SEQ ID NO: 197, CDR3 of SEQ ID NO: 184, (4) heavy chain variable region (H30, H44) comprising CDR1 of SEQ ID NO: 9, CDR2 of SEQ ID NO: 197, CDR3 of SEQ ID NO: 184, (5) A heavy chain variable region comprising CDR
  • H34, H46 heavy chain variable region (H57, H78) comprising CDR1 of SEQ ID NO: 9, CDR2 of SEQ ID NO: 198, CDR3 of SEQ ID NO: 184, (7) A heavy chain variable region comprising CDR1 of SEQ ID NO: 176, CDR2 of SEQ ID NO: 198, CDR3 of SEQ ID NO: 184.
  • a pharmaceutical composition comprising the antibody according to any one of [1] to [11].
  • the pharmaceutical composition according to [12] which is a therapeutic agent for inflammatory diseases.
  • a method for treating or preventing an inflammatory disease comprising a step of administering the antibody according to any one of [1] to [11].
  • FIG. 26 is a continuation diagram of FIG. 25-1.
  • FIG. 2 is a schematic diagram of human / mouse wild type and chimeric NR10-ECD. It is the photograph which detected the binding domain using the Western blot.
  • A is a photograph showing the result of detection with a humanized anti-human NR10 antibody
  • B is a photograph showing the result of detection with a mouse anti-human NR10 antibody
  • C is a photograph showing the result of detection with an anti-Myc antibody.
  • binding antigens were detected only at hhh, hhm, and hmm, and binding could not be confirmed for mmm, mmh, and mhm.
  • FIG. 29 is a continuation of FIG. 28-1.
  • FIG. 29 is a continuation of FIG. 28-2.
  • It is a figure which shows the amino acid sequence of each modification of L0 (sequence number: 52).
  • FIG. 30 is a continuation diagram of FIG. 29-1.
  • NR10 NR10 is a protein that forms a heterodimer with the oncostatin M receptor (OSMR) and functions as a receptor for IL-31. Also known as names such as glm-r (J Biol Chem 277, 16831-6, 2002), GPL (J Biol Chem 278, 49850-9, 2003), IL31RA (Nat Immunol 5, 752-60, 2004) Thus, the NR10 of the present invention includes proteins called by such names.
  • OSMR oncostatin M receptor
  • the origin of the NR10 of the present invention is not particularly limited, and includes NR10 derived from humans, mice, monkeys and other mammals, but preferably derived from humans, mice or monkeys NR10, particularly preferably human-derived NR10.
  • NR10 derived from human (WO00 / 075314).
  • NR10.1 consists of 662 amino acids and has a transmembrane region.
  • NR10.2 is a soluble receptor-like protein that does not have a transmembrane region consisting of a 252 amino acid sequence.
  • NR10.3 and IL31RAv3 are known as NR10 splicing variants that function as a transmembrane receptor protein.
  • the human NR10 in the present invention is not particularly limited as long as it forms a heterodimer with the oncostatin M receptor (OSMR) and functions as a receptor for IL-31, but preferred NR10 is NR10. 3 (also called ILRAv4 (Nat Immunol 5, 752-60, 2004)) and IL31RAv3.
  • NR10.3 IL31RAv4 consists of 662 amino acids (WO00 / 075314, Nat Immunol 5, 752-60, 2004), and IL31RAv3 consists of 732 amino acids (GenBank ACCESSION No: NM_139017).
  • mice-derived NR10 include a protein consisting of the amino acid sequence set forth in SEQ ID NO: 81.
  • NR10 derived from cynomolgus monkey for example, a protein having the amino acid sequence set forth in SEQ ID NO: 66 can be mentioned.
  • a preferred embodiment of the anti-NR10 antibody of the present invention includes the anti-NR10 antibody described in any of (1) to (8) in the following (A) to (D).
  • (A) NS18 (1) CDR1 having the amino acid sequence described in SEQ ID NO: 1 (HCDR1), CDR2 having the amino acid sequence described in SEQ ID NO: 2 (HCDR2), CDR3 having the amino acid sequence described in SEQ ID NO: 3 (HCDR3) (2) An antibody having a heavy chain variable region described in SEQ ID NO: 4 (VH) (3) A CDR1 having an amino acid sequence described in SEQ ID NO: 5 (LCDR1), SEQ ID NO: : An antibody having a light chain variable region comprising CDR2 having the amino acid sequence described in 6 (LCDR2) and CDR3 having the amino acid sequence described in SEQ ID NO: 7 (LCDR3) (4) described in SEQ ID NO: 8 (VL) (5) (1) heavy chain variable region and (3) light chain variable region antibody (6) (2) heavy chain variable region and (4) light chain variable Antibody having region (7)
  • one or more amino acids are substituted, deleted, added and / or inserted.
  • (B) NS22 (1) CDR1 having the amino acid sequence set forth in SEQ ID NO: 9 (HCDR1), CDR2 having the amino acid sequence set forth in SEQ ID NO: 10 (HCDR2), CDR3 having the amino acid sequence set forth in SEQ ID NO: 11 (HCDR3) (2) An antibody having a heavy chain variable region described in SEQ ID NO: 12 (VH) (3) A CDR1 having an amino acid sequence described in SEQ ID NO: 13 (LCDR1), SEQ ID NO: : An antibody having a light chain variable region comprising CDR2 having the amino acid sequence described in 14 (LCDR2) and CDR3 having the amino acid sequence described in SEQ ID NO: 15 (LCDR3) (4) described in SEQ ID NO: 16 (VL) (5) (1) heavy chain variable region and (3) light chain variable region antibody (6) (2) heavy chain variable region and (4) light chain variable Antibody having region (7)
  • one or more amino acids are substituted, deleted, added and An antibody having the same activity as the antibody according to
  • the amino acid after substitution is not particularly limited, but preferred examples include Ile.
  • the amino acid after substitution is not particularly limited, but preferred examples include Leu.
  • the amino acid after substitution is not particularly limited, but preferred examples include Ala.
  • the amino acid after substitution is not particularly limited, but preferred examples include Glu.
  • the amino acid after substitution is not particularly limited, but a preferred example is Asp.
  • substitution of 13th Gln to another amino acid in heavy chain CDR2 of SEQ ID NO: 10 is not particularly limited, but a preferred example is Asp.
  • the amino acid after substitution is not particularly limited, but preferred examples include Gln.
  • the amino acid after substitution is not particularly limited, but a preferred example is Asp.
  • the amino acid after substitution is not particularly limited, but preferred examples include substitution of 16th Lys with Gln and 17th Gly with Asp.
  • the amino acid after substitution is not particularly limited, but preferred examples include substitution of Gln at the 14th Lys, Gln at the 16th Lys, and Asp at the 17th Gly.
  • the amino acid after substitution is not particularly limited, and preferred examples include substitution of 13th Gln Asp, 14th Lys Gln, 16th Lys Gln, and 17th Gly Asp.
  • Substitution of the 10th Ser in the heavy chain CDR2 of SEQ ID NO: 10 with another amino acid is not particularly limited, but a preferred example is Asp.
  • substitution of 13th Gln to another amino acid in heavy chain CDR2 of SEQ ID NO: 10 The amino acid after substitution is not particularly limited, but Pro can be mentioned as a preferred example.
  • substitution of the third Tyr to another amino acid in the heavy chain CDR3 of SEQ ID NO: 11 is not particularly limited, but preferred examples include Leu.
  • Substitution of the 10th Met to another amino acid in the heavy chain CDR3 of SEQ ID NO: 11 is not particularly limited, but preferred examples include Leu.
  • the amino acid after substitution is not particularly limited, but preferred examples include Glu.
  • substitution of the 12th Tyr to another amino acid in the heavy chain CDR3 of SEQ ID NO: 11 is not particularly limited, but preferred examples include Thr and Ser.
  • substitution of the 10th Met, 11th Asp and 12th Tyr with another amino acid is not particularly limited, but preferred examples include substitution of Leu at the 10th Met, Glu at the 11th Asp, and Thr at the 12th Tyr.
  • substitution of 11th Asp and 12th Tyr with another amino acid is not particularly limited, but preferred examples include substitution of 11th Asp with Glu and 12th Tyr with Thr.
  • substitution of the 3rd Tyr, 11th Asp and 12th Tyr with another amino acid is not particularly limited, and preferred examples include substitution of Leu at the 3rd Tyr, Glu at the 11th Asp, and Thr or Ser at the 12th Tyr.
  • the amino acid after substitution is not particularly limited, but preferred examples include Gln.
  • amino acid after substitution is not particularly limited, but a preferred example is Asp.
  • the amino acid after the substitution is not particularly limited, but preferred examples include substitution of the first Arg to Gln and the fifth Asn to Asp.
  • Substitution of the eighth Ser in the light chain CDR1 of SEQ ID NO: 13 with another amino acid is not particularly limited, but preferred examples include Arg.
  • the amino acid after substitution is not particularly limited, and preferred examples include substitution of Arg at the 8th Ser and Val at the 10th Leu.
  • the amino acid after substitution is not particularly limited, but preferred examples include Ala or Ser.
  • Substitution of the first Asn in the light chain CDR2 of SEQ ID NO: 14 with another amino acid is not particularly limited, but a preferred example is Asp.
  • the amino acid after substitution is not particularly limited, but preferred examples include Gln.
  • the amino acid after substitution is not particularly limited, but preferred examples include Glu.
  • Substitution of 7th Lys to another amino acid in light chain CDR2 of SEQ ID NO: 14 is not particularly limited, but preferred examples include Gln or Asp.
  • the amino acid after the substitution is not particularly limited, but preferred examples include substitution of the third Lys to Gln, the fifth Leu to Glu, and the seventh Lys to Gln.
  • the amino acid after substitution is not particularly limited, but a preferred example is Asp.
  • Substitution of the sixth Ser in the light chain CDR3 of SEQ ID NO: 15 with another amino acid is not particularly limited, but a preferred example is Asp.
  • the amino acid after substitution is not particularly limited, but preferred examples include Phe.
  • substitutions may be performed alone or a plurality of substitutions may be combined. Further, the above-described substitution may be combined with substitutions other than those described above. These substitutions can improve the pharmacokinetics (retention in plasma) of the antibody, enhance the binding activity to the antigen, improve the stability, and / or reduce the risk of immunogenicity.
  • variable region combining the above substitutions in the present invention include a heavy chain variable region having the amino acid sequence of SEQ ID NO: 167 or a light chain variable region having the amino acid sequence of SEQ ID NO: 168.
  • an antibody combining the above-mentioned substitution an antibody comprising a heavy chain variable region having the amino acid sequence of SEQ ID NO: 167 and a light chain variable region having the amino acid sequence of SEQ ID NO: 168 can be mentioned.
  • variable regions include the following variable regions.
  • (L11) (j) A light chain variable region comprising CDR1 of SEQ ID NO: 201, CDR2 of SEQ ID NO: 170, CDR3 of SEQ ID NO: 193.
  • (L12) (k) A light chain variable region comprising CDR1 of SEQ ID NO: 202, CDR2 of SEQ ID NO: 170, CDR3 of SEQ ID NO: 193.
  • (L17) (l) A light chain variable region comprising CDR1 described in SEQ ID NO: 203, CDR2 described in SEQ ID NO: 170, CDR3 described in SEQ ID NO: 193. (L50)
  • an antibody comprising the heavy chain variable region of (c) and the light chain variable region of (k) (ii) an antibody comprising the heavy chain variable region of (d) and the light chain variable region of (k) (iii) ( e) an antibody comprising the heavy chain variable region of (k) and the light chain variable region of (k) (iv) the heavy chain of the antibody (v) (g) comprising the heavy chain variable region of (f) and the light chain variable region of (k)
  • An antibody comprising a chain variable region and a light chain variable region of (k) (vi) An antibody comprising a heavy chain variable region of (h) and a light chain variable region of (l)
  • C CDR1 having the amino acid sequence of SEQ ID NO: 17 (HCDR1), CDR2 having the amino acid sequence of SEQ ID NO: 18 (HCDR2), CDR3 having the amino acid sequence of SEQ ID NO: 19 (HCDR3) (2) An antibody having a heavy chain variable region described in SEQ ID NO: 20 (VH) (3) A CDR1 having the amino acid sequence described in SEQ ID NO: 21 (LCDR1), SEQ ID NO: : An antibody having a light chain variable region comprising CDR2 having the amino acid sequence described in 22 (LCDR2) and CDR3 having the amino acid sequence described in SEQ ID NO: 23 (LCDR3) (4) described in SEQ ID NO: 24 (VL) (5) (1) heavy chain variable region and (3) light chain variable region antibody (6) (2) heavy chain variable region and (4) light chain variable Antibody having region (7) One or more amino acids in the antibody according to any one of (1) to (6) A substituted, deleted, added and / or inserted antibody having an activity equivalent to that of any of the antibodies described
  • (D) NS33 (1) CDR1 having the amino acid sequence described in SEQ ID NO: 25 (HCDR1), CDR2 having the amino acid sequence described in SEQ ID NO: 26 (HCDR2), CDR3 having the amino acid sequence described in SEQ ID NO: 27 (HCDR3) (2) An antibody having a heavy chain variable region described in SEQ ID NO: 28 (VH) (3) A CDR1 having an amino acid sequence described in SEQ ID NO: 29 (LCDR1), SEQ ID NO: : An antibody having a light chain variable region comprising CDR2 having the amino acid sequence described in 30 (LCDR2) and CDR3 having the amino acid sequence described in SEQ ID NO: 31 (LCDR3) (4) described in SEQ ID NO: 32 (VL) (5) (1) heavy chain variable region and (3) light chain variable region antibody (6) (2) heavy chain variable region and (4) light chain variable Antibody having region (7) One or more amino acids in the antibody according to any one of (1) to (6) A substituted, deleted, added and / or inserted antibody having an activity equivalent to that of any
  • any framework region may be used, but human-derived FR is preferably used.
  • any constant region may be used as the constant region, but a human-derived constant region is preferably used.
  • the amino acid sequence of the FR or constant region used in the antibody of the present invention may be the original FR or constant region amino acid sequence from which it is derived, or may be substituted, deleted, added and / or substituted for one or more amino acids. Alternatively, different amino acid sequences may be used by insertion or the like.
  • the above-mentioned NS18 heavy chain amino acid sequence is shown in SEQ ID NO: 34, and the base sequence encoding the amino acid sequence is shown in SEQ ID NO: 33.
  • the amino acid sequence of the light chain is shown in SEQ ID NO: 36, and the base sequence encoding the amino acid sequence is shown in SEQ ID NO: 35.
  • amino acid sequence of the heavy chain of NS22 is shown in SEQ ID NO: 38, and the base sequence encoding the amino acid sequence is shown in SEQ ID NO: 37.
  • amino acid sequence of the light chain is shown in SEQ ID NO: 40, and the base sequence encoding the amino acid sequence is shown in SEQ ID NO: 39.
  • amino acid sequence of the heavy chain of NS23 is shown in SEQ ID NO: 42, and the base sequence encoding the amino acid sequence is shown in SEQ ID NO: 41.
  • the amino acid sequence of the light chain is shown in SEQ ID NO: 44, and the base sequence encoding the amino acid sequence is shown in SEQ ID NO: 43.
  • the amino acid sequence of the heavy chain of NS33 is shown in SEQ ID NO: 46, and the base sequence encoding the amino acid sequence is shown in SEQ ID NO: 45.
  • the amino acid sequence of the light chain is shown in SEQ ID NO: 48, and the base sequence encoding the amino acid sequence is shown in SEQ ID NO: 47.
  • “activity equivalent to the antibody according to any one of (1) to (6)” means that the binding activity and / or neutralizing activity to NR10 (eg, human NR10) is equivalent.
  • “equivalent” does not necessarily have the same level of activity, and the activity may be enhanced, or the activity may be decreased as long as it has activity.
  • Examples of antibodies with decreased activity include antibodies having 30% or more activity, preferably 50% or more activity, more preferably 80% or more activity compared to the original antibody. .
  • the antibody described in any of (1) to (6) above has one or more amino acid sequences in the variable region (CDR sequence and / or FR sequence) as long as it has binding activity and / or neutralizing activity against NR10.
  • amino acids may be substituted, deleted, added and / or inserted.
  • a method of introducing a mutation into a protein is known.
  • a person skilled in the art can perform site-directed mutagenesis (Hashimoto-Gotoh, T, Mizuno, T, Ogasahara, Y, and Nakagawa, M. (1995) An oligodeoxyribonucleotide-directed dual amber method for site-directed mutagenesis. Gene 152, 271-275, Zoller, MJ, and Smith, M. (1983) Oligonucleotide-directed mutagenesis of DNA fragments cloned into M13 vectors.Methods Enzymol.
  • antibodies may be prepared as functionally equivalent variants with binding activity and / or neutralizing activity against NR10.
  • an antibody having one or more amino acids mutated in the variable region and having binding activity and / or neutralizing activity for NR10 is also included in the antibody of the present invention.
  • amino acid side chain properties include hydrophobic amino acids (A, I, L, M, F, P, W, Y, V), hydrophilic amino acids (R, D, N, C, E, Q, G, H, K, S, T), amino acids having aliphatic side chains (G, A, V, L, I, P), amino acids having hydroxyl group-containing side chains (S, T, Y), sulfur atom-containing side chains Amino acids (C, M) having carboxylic acid and amide-containing side chains (D, N, E, Q), amino acids having base-containing side chains (R, K, H), and aromatic-containing sides Amino acids having a chain (H, F, Y, W) can be mentioned (the parentheses indicate single letter symbols of amino acids).
  • Amino acid substitutions within each of these groups are referred to as conservative substitutions. It is already known that a polypeptide having an amino acid sequence modified by deletion, addition and / or substitution with other amino acids of one or more amino acid residues to a certain amino acid sequence maintains its biological activity. (Mark, D. F. et al., Proc.Natl.Acad.Sci.USA (1984) 81: 5662-6; Zoller, M. J. and Smith, M., Nucleic Acids Res. (1982) 10 : 6487-500; Wang, A. et al., Science (1984) 224: 1431-3; Dalbadie-McFarland, G.
  • variants are at least 70%, more preferably at least 75%, more preferably at least 80%, more preferably at least 80% of the amino acid sequence of the variable region of the present invention (eg, CDR sequence, FR sequence, entire variable region). It has 85%, even more preferably at least 90%, and most preferably at least 95% amino acid sequence identity.
  • sequence identity refers to the amino acid sequence of the original heavy chain variable region or light chain variable region after aligning the sequences as necessary so that the sequence identity is maximized and introducing gaps as appropriate. Defined as the percentage of residues that are identical to the residues. The identity of amino acid sequences can be determined by the method described below.
  • amino acid sequence of the variable region (CDR sequence and / or FR sequence) one or more amino acids are substituted, deleted, added and / or inserted, and have binding activity and / or neutralizing activity against NR10.
  • the amino acid sequence of the variable region can also be obtained from a nucleic acid that hybridizes under stringent conditions to a nucleic acid comprising a base sequence encoding the amino acid sequence of the variable region.
  • Stringent hybridization conditions for isolating a nucleic acid that hybridizes under stringent conditions to a nucleic acid consisting of a base sequence encoding the amino acid sequence of the variable region include 6M urea, 0.4% SDS, 0.5 x SSC.
  • nucleic acids with higher homology can be expected by using conditions with higher stringency, for example, 6M6urea, 0.4% SDS, 0.1 x SSC, and 42 ° C.
  • the sequence of the isolated nucleic acid can be determined by a known method described later.
  • the homology of the isolated nucleic acid is at least 50% or more, more preferably 70% or more, more preferably 90% or more (for example, 95%, 96%, 97%, 98%, 99%) in the entire base sequence. And the like).
  • a gene amplification method using a primer synthesized based on nucleotide sequence information encoding the amino acid sequence of the variable region for example, a polymerase chain reaction (PCR) method is used. It is also possible to isolate a nucleic acid that hybridizes with a nucleic acid comprising a base sequence encoding the amino acid sequence of the region under stringent conditions.
  • PCR polymerase chain reaction
  • the identity of the base sequence and amino acid sequence can be determined by the algorithm BLAST (Proc. Natl. Acad. Sci. USA (1993) 90: 5873-7) by Karlin and Altschul. Based on this algorithm, programs called BLASTN and BLASTX have been developed (Altschul et al., J. Mol. Biol. (1990) 215: 403-10).
  • BLASTN a base sequence
  • BLASTX based on BLAST
  • the present invention also provides an antibody that binds to the same epitope as that to which the antibody according to any one of (1) to (7) binds.
  • Whether or not a certain antibody recognizes the same epitope as another antibody can be confirmed by competition for both epitopes.
  • Competition between antibodies can be evaluated by a competitive binding assay.
  • the means include ELISA, fluorescence energy transfer measurement (FRET), and fluorescence micromeasurement technique (FMAT (registered trademark)).
  • FRET fluorescence energy transfer measurement
  • FMAT fluorescence micromeasurement technique
  • the antibody labeled with an appropriate label and the antibody to be evaluated are simultaneously added to the antigen, and the bound antibody is detected using the label.
  • the amount of the antibody bound to the antigen can be easily measured by labeling the antibody in advance.
  • This labeling is not particularly limited, but a labeling method corresponding to the technique is selected. Specific examples of the labeling method include fluorescent labeling, radiolabeling, enzyme labeling and the like.
  • the fluorescently labeled antibody and the unlabeled antibody or test antibody are simultaneously added to animal cells expressing NR10, and the labeled antibody is detected by a fluorescence micromeasurement technique.
  • an antibody recognizing the same epitope refers to a non-labeled antibody that has a non-labeled antibody against a concentration (IC 50 ) that reduces the binding amount by binding of the unlabeled antibody. Reduce the binding amount of the labeled antibody by at least 50% at a concentration that is usually 100 times, preferably 80 times, more preferably 50 times, more preferably 30 times, more preferably 10 times higher than the IC 50 of the labeled antibody. It is an antibody that can.
  • the antibody that binds to the epitope to which the antibody described in any of (1) to (7) above binds is particularly useful in that it has a high neutralizing activity.
  • the antibody described in any of (1) to (8) above is not particularly limited, but is preferably a humanized antibody.
  • the present invention provides a gene encoding the anti-NR10 antibody according to any one of (1) to (8) in the above (A) to (D).
  • the gene of the present invention may be any gene, for example, DNA or RNA.
  • Antibody (humanized)
  • One preferred embodiment of the antibody in the present invention is a humanized antibody that binds to NR10.
  • Humanized antibodies can be produced using methods known to those skilled in the art.
  • variable region of an antibody is usually composed of three complementarity determining regions (CDRs) sandwiched between four frames (FR).
  • CDRs are regions that substantially determine the binding specificity of an antibody.
  • the amino acid sequence of CDR is rich in diversity.
  • the amino acid sequence constituting FR often shows high homology among antibodies having different binding specificities. Therefore, it is generally said that the binding specificity of one antibody can be transplanted to another antibody by CDR grafting.
  • a humanized antibody is also referred to as a reshaped human antibody, which is a non-human mammal, for example, a mouse antibody CDR grafted to the complementarity determining region of a human antibody, and its general gene Recombination techniques are also known (see European Patent Application Publication Nos. EP 125023 and WO 96/02576).
  • the CDR when the CDR is derived from a mouse antibody, a DNA sequence designed to link the CDR of the mouse antibody and the framework region (FR) of the human antibody is used for both CDR and FR.
  • the oligonucleotide is synthesized by PCR using several oligonucleotides prepared so as to have a portion overlapping with the terminal region (see the method described in WO98 / 13388).
  • the obtained DNA is obtained by ligating with the DNA encoding the human antibody constant region, then incorporating it into an expression vector, introducing it into a host and producing it (European patent application publication number EP 239400, international patent application publication number). (See WO 96/02576).
  • the framework region of the human antibody to be linked to the CDR is selected such that the complementarity determining region forms a favorable antigen binding site.
  • the amino acid of the framework region in the variable region of the antibody may be substituted, deleted, added, and / or inserted so that the complementarity determining region of the reshaped human antibody forms an appropriate antigen-binding site.
  • amino acid sequence mutations can be introduced into FRs by applying the PCR method used for transplantation of mouse CDRs into human FRs.
  • partial nucleotide sequence mutations can be introduced into primers that anneal to the FR.
  • a nucleotide sequence mutation is introduced into the FR synthesized by such a primer.
  • a mutant FR sequence having a desired property can be selected by measuring and evaluating the antigen-binding activity of a mutant antibody substituted with an amino acid by the above method (Sato, K. etal., Cancer Res. (1993) 53, 851-856).
  • C ⁇ 1, C ⁇ 2, C ⁇ 3, C ⁇ 4, C ⁇ , C ⁇ , C ⁇ 1, C ⁇ 2, C ⁇ are used for the H chain
  • C ⁇ , C ⁇ are used for the L chain. be able to.
  • the amino acid sequence of C ⁇ is shown in SEQ ID NO: 58, and the base sequence encoding the amino acid sequence is shown in SEQ ID NO: 57.
  • the amino acid sequence of C ⁇ 1 is shown in SEQ ID NO: 60, and the base sequence encoding the amino acid sequence is shown in SEQ ID NO: 59.
  • the amino acid sequence of C ⁇ 2 is shown in SEQ ID NO: 62, and the base sequence encoding the amino acid sequence is shown in SEQ ID NO: 61.
  • the amino acid sequence of C ⁇ 4 is shown in SEQ ID NO: 64, and the base sequence encoding the amino acid sequence is shown in SEQ ID NO: 63.
  • the human antibody C region may be modified in order to improve the stability of the antibody or its production. Examples of the modified human antibody C region include the C region described later.
  • Human antibodies used for humanization may be human antibodies of any isotype such as IgG, IgM, IgA, IgE, IgD, but IgG is preferably used in the present invention. IgG1, IgG2, IgG3, IgG4, etc. can be used as IgG.
  • amino acids in the variable region eg, CDR, FR
  • constant region e.g., CDR, FR
  • the humanized antibody also includes a humanized antibody having such amino acid substitution.
  • the origin of CDR in a humanized antibody is not particularly limited and may be derived from any animal.
  • sequences of mouse antibody, rat antibody, rabbit antibody, camel antibody and the like can be used, but the CDR sequence of mouse antibody is preferable.
  • humanization of antibodies it is usually difficult to perform humanization while maintaining the binding activity and neutralizing activity of the derived antibody, but in the present invention, the binding is equivalent to that of the derived mouse antibody.
  • humanized anti-NR10 antibody examples include the antibodies described in any of the following (a) to (e).
  • a humanized antibody comprising a heavy chain variable region having the amino acid sequence of SEQ ID NO: 50 (H0-VH) (b) a human comprising a heavy chain variable region having the amino acid sequence of SEQ ID NO: 112 (H1-VH)
  • C a humanized antibody comprising a light chain variable region having the amino acid sequence of SEQ ID NO: 52 (L0-VL)
  • the heavy chain variable region having the amino acid sequence set forth in SEQ ID NO: 50 H0-VH
  • the heavy chain variable region having the amino acid sequence set forth in SEQ ID NO: 112 the sequence described in SEQ ID NO: 52 (L0-VL)
  • one or more amino acids may be substituted, deleted, added and / or inserted. Amino acid substitutions, deletions, additions and / or insertions may be made in the CDR or FR, or in both the CDR and FR.
  • the humanized anti-NR10 antibody in the present invention include, for example, the antibodies described in any of (f) to (j) below.
  • (h) SEQ ID NO: 52 (L0- An antibody comprising a light chain variable region having an amino acid sequence in which one or more amino acids are substituted, deleted, added and / or inserted in the amino acid sequence of (VL)
  • the antibody described in any of (f) to (j) preferably has the same activity as the antibody described in any of (a) to (e).
  • amino acid substitution, deletion, addition and / or insertion is not particularly limited, but as a specific example, for example, the amino acid substitution described above can be performed. More specifically, for example, the following amino acid substitutions can be mentioned.
  • CDR2 (SEQ ID NO: 10) with 17th Gly to Asp in the heavy chain variable region of SEQ ID NO: 50 or SEQ ID NO: 112 (SEQ ID NO: 118). Therefore, in the present invention, in the heavy chain variable region having the amino acid sequence of SEQ ID NO: 50 or SEQ ID NO: 112, CDR2 having the amino acid sequence of SEQ ID NO: 10 is substituted with CDR2 having the amino acid sequence of SEQ ID NO: 118. Provides a heavy chain variable region.
  • CDR3 (SEQ ID NO: 11) is substituted with 10th Met to Leu, 11th Asp to Glu, and 12th Tyr Thr. (SEQ ID NO: 184). Therefore, in the present invention, in the heavy chain variable region having the amino acid sequence of SEQ ID NO: 50 or SEQ ID NO: 112, CDR3 having the amino acid sequence of SEQ ID NO: 11 is substituted with CDR3 having the amino acid sequence of SEQ ID NO: 184. Provides a heavy chain variable region.
  • the present invention relates to a light chain variable region having the amino acid sequence of SEQ ID NO: 52, a light chain variable region in which CDR1 having the amino acid sequence of SEQ ID NO: 13 is replaced with CDR1 having the amino acid sequence of SEQ ID NO: 121. provide.
  • the present invention relates to a light chain variable region having the amino acid sequence of SEQ ID NO: 52, a light chain variable region in which CDR1 having the amino acid sequence of SEQ ID NO: 13 is replaced with CDR1 having the amino acid sequence of SEQ ID NO: 122. provide.
  • the present invention relates to a light chain variable region having the amino acid sequence of SEQ ID NO: 52, a light chain variable region in which CDR1 having the amino acid sequence of SEQ ID NO: 13 is replaced with CDR1 having the amino acid sequence of SEQ ID NO: 188. provide.
  • the present invention relates to a light chain variable region having the amino acid sequence of SEQ ID NO: 52, a light chain variable region in which CDR1 having the amino acid sequence of SEQ ID NO: 13 is replaced with CDR1 having the amino acid sequence of SEQ ID NO: 189. provide.
  • the present invention relates to a light chain variable region having the amino acid sequence of SEQ ID NO: 52, a light chain variable region in which CDR1 having the amino acid sequence of SEQ ID NO: 13 is replaced with CDR1 having the amino acid sequence of SEQ ID NO: 190. provide.
  • the present invention relates to a light chain variable region having the amino acid sequence of SEQ ID NO: 52, a light chain variable region in which CDR1 having the amino acid sequence of SEQ ID NO: 13 is substituted with CDR1 having the amino acid sequence of SEQ ID NO: 191.
  • the present invention relates to a light chain variable region having the amino acid sequence of SEQ ID NO: 52, a light chain variable region in which CDR1 having the amino acid sequence of SEQ ID NO: 13 is replaced with CDR1 having the amino acid sequence of SEQ ID NO: 192. provide.
  • the present invention relates to a light chain variable region having the amino acid sequence of SEQ ID NO: 52, a light chain variable region in which CDR2 having the amino acid sequence of SEQ ID NO: 14 is replaced with CDR2 having the amino acid sequence of SEQ ID NO: 123. provide.
  • the present invention relates to a light chain variable region having the amino acid sequence of SEQ ID NO: 52, a light chain variable region in which CDR2 having the amino acid sequence of SEQ ID NO: 14 is replaced with CDR2 having the amino acid sequence of SEQ ID NO: 124. provide.
  • the present invention relates to a light chain variable region having the amino acid sequence of SEQ ID NO: 52, a light chain variable region in which CDR2 having the amino acid sequence of SEQ ID NO: 14 is replaced with CDR2 having the amino acid sequence of SEQ ID NO: 125. provide.
  • the present invention relates to a light chain variable region having the amino acid sequence of SEQ ID NO: 52, a light chain variable region in which CDR2 having the amino acid sequence of SEQ ID NO: 14 is replaced with CDR2 having the amino acid sequence of SEQ ID NO: 126. provide.
  • the present invention relates to a light chain variable region having the amino acid sequence of SEQ ID NO: 52, a light chain variable region in which CDR2 having the amino acid sequence of SEQ ID NO: 14 is replaced with CDR2 having the amino acid sequence of SEQ ID NO: 127. provide.
  • the present invention relates to a light chain variable region having the amino acid sequence of SEQ ID NO: 52, a light chain variable region in which CDR1 having the amino acid sequence of SEQ ID NO: 13 is replaced with CDR1 having the amino acid sequence of SEQ ID NO: 169. provide.
  • the present invention provides a light chain variable region in which the CDR2 having the amino acid sequence of SEQ ID NO: 14 is replaced with the CDR2 having the amino acid sequence of SEQ ID NO: 170 in the light chain variable region of SEQ ID NO: 52.
  • the present invention provides a light chain variable region in which the CDR3 having the amino acid sequence of SEQ ID NO: 15 is replaced with the CDR3 having the amino acid sequence of SEQ ID NO: 193 in the light chain variable region of SEQ ID NO: 52.
  • the present invention provides a light chain variable region in which the CDR3 having the amino acid sequence of SEQ ID NO: 15 is replaced with the CDR3 having the amino acid sequence of SEQ ID NO: 194 in the light chain variable region of SEQ ID NO: 52.
  • the present invention provides a light chain variable region in which the CDR3 having the amino acid sequence of SEQ ID NO: 15 is replaced with the CDR3 having the amino acid sequence of SEQ ID NO: 195 in the light chain variable region of SEQ ID NO: 52.
  • examples of the substitution other than the amino acid substitution described above include substitution of the third Arg with another amino acid in the heavy chain FR2 having the amino acid sequence of SEQ ID NO: 97.
  • the amino acid after substitution is not particularly limited, but preferred examples include Gln.
  • the 3rd Arg of SEQ ID NO: 97 is substituted with Gln, the 5th Ala can be substituted with Ser to make FR2 a human sequence.
  • the amino acid sequence in which the third Arg is substituted with Gln and the fifth Ala with Ser is shown in SEQ ID NO: 120.
  • FR2 having the amino acid sequence of SEQ ID NO: 97 is substituted with FR2 having the amino acid sequence of SEQ ID NO: 120 in the heavy chain variable region having the amino acid sequence of SEQ ID NO: 50 or SEQ ID NO: 112. Provides a heavy chain variable region.
  • amino acid substitutions described above may be used alone or in combination with the other amino acid substitutions described above. Moreover, you may combine with amino acid substitution other than the above-mentioned.
  • the antibody in which the above substitution has been made include an antibody comprising a heavy chain variable region having the amino acid sequence of SEQ ID NO: 167, an antibody comprising a light chain variable region having the amino acid sequence of SEQ ID NO: 168, sequence And an antibody comprising a heavy chain variable region having the amino acid sequence of No. 167 and a light chain variable region having the amino acid sequence of SEQ ID No. 168.
  • heavy chain variable region having the amino acid sequence set forth in SEQ ID NO: 204 (H17)
  • Heavy chain variable region having the amino acid sequence set forth in SEQ ID NO: 205 (H19)
  • Heavy chain variable region having the amino acid sequence set forth in SEQ ID NO: 206 (H28)
  • Heavy chain variable region (H30) having the amino acid sequence set forth in SEQ ID NO: 207
  • Heavy chain variable region having the amino acid sequence set forth in SEQ ID NO: 208 (H34)
  • Heavy chain variable region having the amino acid sequence set forth in SEQ ID NO: 209 H42
  • Heavy chain variable region having the amino acid sequence set forth in SEQ ID NO: 210 (H44)
  • Heavy chain variable region having the amino acid sequence set forth in SEQ ID NO: 211 (H46)
  • Heavy chain variable region having the amino acid sequence set forth in SEQ ID NO: 212 (H57)
  • Heavy chain variable region having the amino acid sequence set forth in SEQ ID NO: 212 (H57)
  • Heavy chain variable region having the
  • the constant region used in the humanized antibody of the present invention may be any constant region derived from a human antibody.
  • Preferable examples of constant regions derived from human antibodies include constant regions derived from IgG1 or IgG2.
  • a constant region in which one or more amino acids are substituted, deleted, added and / or inserted in a constant region derived from a human antibody may be used.
  • the constant region in which one or more amino acids are substituted, deleted, added and / or inserted in the constant region derived from a human antibody is not particularly limited, and examples thereof include the following constant regions.
  • the heavy chain or antibody using the above-described constant region include the following heavy chains or antibodies.
  • a heavy chain comprising a variable region having the amino acid sequence of SEQ ID NO: 167 and a constant region having the amino acid sequence of SEQ ID NO: 128
  • the heavy chain of (1) has the amino acid sequence of SEQ ID NO: 171
  • a heavy chain of (2) heavy chain (3) (1) in which CDR2 is replaced by CDR2 having the amino acid sequence of SEQ ID NO: 172 and a heavy chain of light chain having the amino acid sequence of SEQ ID NO: 152 (4)
  • An antibody comprising a chain and a light chain having the amino acid sequence of SEQ ID NO: 152
  • humanized anti-NR10 antibody of the present invention include, for example, the antibodies described in any of (k) to (o) below.
  • n A heavy chain having the amino acid sequence of SEQ ID NO: 54 (H0-VH + constant region), and SEQ ID NO: 56
  • An antibody comprising a light chain having the amino acid sequence of (L0-VL + constant region) (o) a heavy chain having the amino acid sequence of SEQ ID NO: 130 (H1-VH + constant region), and SEQ ID NO: 56 (L0-VL + constant region)
  • the heavy chain having the amino acid sequence set forth in SEQ ID NO: 54 (H0-VH + constant region) and the light chain having the amino acid sequence set forth in SEQ ID NO: 56 (L0-VL + constant region) are one or more amino acids. May be substituted, deleted, added and / or inserted. Amino acid substitutions, deletions, additions and / or insertions may be made in the variable region or constant region, or in both the variable region and constant region.
  • the present invention provides the antibody described in any of (p) to (t) below.
  • the antibody described in any of (p) to (t) preferably has the same activity as the antibody described in any of (k) to (o).
  • amino acid substitution, deletion, addition and / or insertion are not particularly limited, for example, the above-described amino acid substitution can be performed as a specific example.
  • the base sequence encoding the amino acid sequence (SEQ ID NO: 50) of the above-mentioned humanized heavy chain variable region is shown in SEQ ID NO: 49, and the base sequence encoding the amino acid sequence of the humanized light chain variable region (SEQ ID NO: 52).
  • SEQ ID NO: 51 the base sequence encoding the humanized heavy chain amino acid sequence (SEQ ID NO: 54) is SEQ ID NO: 53, and the humanized light chain amino acid sequence (SEQ ID NO: 56) is the base sequence.
  • the present invention provides an antibody that recognizes the same epitope as the epitope recognized by the antibody described in any of (a) to (t) above. The binding to the same epitope has already been described.
  • the present invention provides the antibody described in any of (u) to (w) below.
  • the present invention provides a heavy chain, light chain, or antibody described in any of the following.
  • Heavy chain (H17) having the amino acid sequence set forth in SEQ ID NO: 222 (2) Heavy chain (H19) having the amino acid sequence set forth in SEQ ID NO: 223 (3) Heavy chain having the amino acid sequence set forth in SEQ ID NO: 224 (H28) (4) Heavy chain (H30) having the amino acid sequence set forth in SEQ ID NO: 225 (5) Heavy chain having the amino acid sequence set forth in SEQ ID NO: 226 (H34) (6) Heavy chain having the amino acid sequence set forth in SEQ ID NO: 227 (H42) (7) Heavy chain having the amino acid sequence set forth in SEQ ID NO: 228 (H44) (8) Heavy chain having the amino acid sequence set forth in SEQ ID NO: 229 (H46) (9) Heavy chain having the amino acid sequence set forth in SEQ ID NO: 230 (H57) (10) Heavy chain having the amino acid sequence set forth in SEQ ID NO: 231 (H71) (11) Heavy chain having
  • Amino acid substitution, deletion, addition and / or insertion are as described above.
  • An antibody that recognizes the same epitope as that recognized by a certain antibody is as described above.
  • the present invention provides a gene encoding the variable region of the present invention, the heavy chain of the present invention, the light chain of the present invention or the antibody of the present invention.
  • the present invention provides a vector containing the above gene.
  • the present invention provides a host cell transformed with the above vector.
  • the present invention relates to a method for producing the variable region of the present invention, the heavy chain of the present invention, the light chain of the present invention or the antibody of the present invention, which comprises the step of culturing the above host cell.
  • the vector, host cell, and host cell culture are as described later.
  • domain 1 refers to the region (LPAKP to LENIA) from the 21st amino acid to the 120th amino acid in the number containing the signal peptide in the amino acid sequence of human NR10 described in SEQ ID NO: 76.
  • domain 2 is a region from amino acid 121 to amino acid 227 in the amino acid sequence of human NR10 described in SEQ ID NO: 76 in the state containing a signal peptide (KTEPP to EEEAP).
  • Such an antibody is not particularly limited, but is usually an antibody having neutralizing activity, preferably a humanized antibody.
  • An example of a preferable antibody in the present invention is an antibody that recognizes domain 1.
  • An antibody that recognizes domain 1 has a high neutralizing activity and is particularly useful as a pharmaceutical product.
  • the present invention further provides an anti-NR10 antibody having neutralizing activity.
  • the neutralizing activity against NR10 is an activity that inhibits the binding between NR10 and its ligand IL-31, and preferably an activity that suppresses physiological activity based on NR10.
  • Selection of an antibody having NR10 neutralizing activity can be performed, for example, by a method of observing the growth inhibitory effect of the IL-31-dependent cell line when a candidate antibody is added to the IL-31-dependent cell line. Is possible.
  • the antibody that suppresses the growth of the IL-31-dependent cell line in the above method is judged to be an antibody having neutralizing activity against NR10.
  • the antibody of the present invention is not limited in its origin, and may be an antibody derived from any animal such as a human antibody, a mouse antibody, or a rat antibody. Further, it may be a recombinant antibody such as a chimeric antibody or a humanized antibody. As mentioned above, humanized antibodies can be mentioned as preferred antibodies of the present invention.
  • Chimeric antibodies are non-human mammals, for example, antibodies consisting of mouse antibody heavy and light chain variable regions and human antibody heavy and light chain constant regions. Chimeric antibodies can be produced using known methods.
  • the antibody gene can be cloned from a hybridoma, incorporated into an appropriate vector, and introduced into a host (for example, Carl, A. K. Borrebaeck, James, W. Larrick, THERAPEUTIC MONOCLONAL ANTIBODIES, “Published” in “the” United “Kingdom” by “MACMILLAN” PUBLISHERS “LTD,” 1990).
  • cDNA of the variable region (V region) of the antibody is synthesized from the hybridoma mRNA using reverse transcriptase.
  • DNA encoding the V region of the target antibody is obtained, it is ligated with DNA encoding the desired human antibody constant region (C region) and incorporated into an expression vector.
  • DNA encoding the V region of the antibody may be incorporated into an expression vector containing DNA of the human antibody C region. It is incorporated into an expression vector so as to be expressed under the control of an expression control region such as an enhancer or promoter.
  • host cells can be transformed with this expression vector to express the chimeric antibody.
  • a method for obtaining a human antibody is also known.
  • human lymphocytes are sensitized with a desired antigen or cells expressing the desired antigen in vitro, and the sensitized lymphocytes are fused with human myeloma cells, such as U266, to have a desired human antibody having an antigen-binding activity.
  • a desired human antibody can be obtained by immunizing a transgenic animal having all repertoires of human antibody genes with a desired antigen (International Patent Application Publication Nos. WO 93/12227, WO 92/03918, WO 94/02602, WO 94/25585, WO 96/34096, WO 96/33735).
  • variable region of a human antibody is expressed as a single chain antibody (scFv) on the surface of the phage by the phage display method, and a phage that binds to the antigen can be selected.
  • scFv single chain antibody
  • the DNA sequence encoding the variable region of the human antibody that binds to the antigen can be determined. If the DNA sequence of scFv that binds to the antigen is clarified, an appropriate expression vector having the sequence can be prepared and a human antibody can be obtained.
  • the antibody of the present invention is not only a bivalent antibody represented by IgG, but also a monovalent antibody, or a multivalent antibody represented by IgM, or different. Bispecific antibodies that can bind to an antigen are also included.
  • the multivalent antibodies of the present invention include multivalent antibodies that all have the same antigen-binding site, or multivalent antibodies that have some or all different antigen-binding sites.
  • the antibody of the present invention is not limited to the full-length molecule of the antibody, and may be a low molecular weight antibody or a modified product thereof as long as it binds to the NR10 protein.
  • the antibody in the present invention may be a low molecular weight antibody.
  • the low molecular weight antibody is an antibody containing an antibody fragment in which a part of a full-length antibody (whole antibody, such as whole IgG) is deleted, and is not particularly limited as long as it has a binding activity to NR10 and / or a neutralizing activity.
  • the low molecular weight antibody is not particularly limited as long as it includes a part of the full-length antibody, but preferably includes a heavy chain variable region (VH) or a light chain variable region (VL), and particularly preferably VH and VL. It is a low molecular weight antibody containing both.
  • Another preferred example of the low molecular weight antibody of the present invention is a low molecular weight antibody containing the CDR of the antibody.
  • the CDR included in the low molecular weight antibody may include all six CDRs of the antibody or may include a part of the CDRs.
  • the low molecular weight antibody in the present invention preferably has a smaller molecular weight than the full-length antibody.
  • it may form a multimer such as a dimer, trimer or tetramer, and the molecular weight is larger than that of the full-length antibody. There is also.
  • antibody fragments include Fab, Fab ′, F (ab ′) 2, and Fv.
  • specific examples of the low molecular weight antibody include, for example, Fab, Fab ′, F (ab ′) 2, Fv, scFv (single chain Fv), Diabody, sc (Fv) 2 (single chain (Fv) 2) And so on. Multimers of these antibodies (eg, dimer, trimer, tetramer, polymer) are also included in the low molecular weight antibody of the present invention.
  • Antibody fragments can be obtained, for example, by treating an antibody with an enzyme to generate antibody fragments.
  • enzymes that produce antibody fragments include, for example, papain, pepsin, and plasmin.
  • genes encoding these antibody fragments can be constructed, introduced into an expression vector, and then expressed in an appropriate host cell (for example, Co, MS et al., J. Immunol. (1994) 152). , 2968-2976, Better, M. & Horwitz, A. H. Methods in Enzymology (1989) 178, 476-496, Plueckthun, A. & Skerra, A.
  • the digestive enzyme cleaves a specific position of the antibody fragment to give an antibody fragment having the following specific structure. If a genetic engineering technique is used for such an enzymatically obtained antibody fragment, any part of the antibody can be deleted.
  • the antibody fragments obtained when the above digestive enzymes are used are as follows. Papain digestion: F (ab) 2 or Fab Pepsin digestion: F (ab ') 2 or Fab' Plasmin digestion: Facb
  • the low molecular weight antibody in the present invention can contain an antibody fragment lacking any region as long as it has binding activity to NR10 and / or neutralizing activity.
  • Diabody refers to a bivalent antibody fragment constructed by gene fusion (Holliger Pet et al., Proc. Natl. Acad. Sci. USA 90: 6444-6448 (1993), EP 404,097, WO93 / 11161 etc.).
  • Diabodies are dimers composed of two polypeptide chains. Usually, in the polypeptide chain constituting the dimer, VL and VH are connected by a linker in the same chain. The linker in the diabody is generally so short that VL and VH cannot bind to each other. Specifically, the amino acid residues constituting the linker are, for example, about 5 residues. Therefore, VL and VH encoded on the same polypeptide chain cannot form a single chain variable region fragment but form a dimer with another single chain variable region fragment. As a result, the diabody has two antigen binding sites.
  • the scFv antibody is an antibody in which a heavy chain variable region ([VH]) and a light chain variable region ([VL]) are combined with a linker or the like to form a single chain polypeptide (Huston, J. S. et al. , Proc. Natl. Acad. Sci. USA 1988 (1988) 85, 5879-5883, Plickthun ⁇ The Pharmacology of Monoclonal Antibodies '' Vol. 113, Resenburg and Moore, Springer Verlag, New York, pp.269-315, (1994 )).
  • the H chain V region and L chain V region in scFv may be derived from any of the antibodies described herein.
  • peptide linker which connects V area
  • any single chain peptide consisting of about 3 to 25 residues can be used as a linker.
  • a peptide linker described later can be used.
  • V regions of both strands can be linked by, for example, the PCR method as described above.
  • the DNA encoding the desired partial amino acid sequence is used as a template.
  • the DNAs encoding the V region of the H chain and the L chain are each amplified by PCR using a pair of primers having sequences corresponding to the sequences at both ends of the DNA to be amplified.
  • DNA encoding a peptide linker portion is prepared.
  • DNA encoding a peptide linker can also be synthesized using PCR.
  • a base sequence that can be linked to the amplification product of each V region synthesized separately is added to the 5 ′ side of the primer to be used.
  • PCR reaction is performed using each DNA of [H chain V region DNA]-[peptide linker DNA]-[L chain V region DNA] and assembly PCR primers.
  • the primer for assembly PCR consists of a combination of a primer that anneals to the 5 ′ side of [H chain V region DNA] and a primer that anneals to the 3 ′ side of [L chain V region DNA]. That is, the assembly PCR primer is a primer set that can amplify DNA encoding the full-length sequence of scFv to be synthesized. On the other hand, a base sequence that can be linked to each V region DNA is added to [peptide linker DNA]. As a result, these DNAs are ligated, and the full length of scFv is finally produced as an amplification product by the primers for assembly PCR.
  • an expression vector containing them and a recombinant cell transformed with the expression vector can be obtained according to a conventional method. Further, the scFv can be obtained by culturing the resulting recombinant cells and expressing the DNA encoding the scFv.
  • the order of the heavy chain variable region and the light chain variable region to be combined is not particularly limited, and may be arranged in any order, and examples thereof include the following arrangements.
  • Sc (Fv) 2 is a low molecular weight antibody in which two VHs and two VLs are combined with a linker or the like to form a single chain (Hudson et al., J Immunol. Methods Methods 1999; 231: 177-189).
  • sc (Fv) 2 can be prepared, for example, by linking scFv with a linker.
  • VHs and two VLs are arranged in the order of VH, VL, VH, and VL ([VH] linker [VL] linker [VH] linker [VL]) starting from the N-terminal side of the single-chain polypeptide.
  • the order of the two VHs and the two VLs is not particularly limited to the above arrangement, and may be arranged in any order. For example, the following arrangements can also be mentioned.
  • the amino acid sequence of the heavy chain variable region or light chain variable region in the low molecular antibody may be substituted, deleted, added and / or inserted. Furthermore, when the heavy chain variable region and the light chain variable region are associated, a part may be deleted or another polypeptide may be added as long as it has antigen binding activity.
  • the variable region may be chimerized or humanized.
  • the linker that binds the variable region of the antibody is any peptide linker that can be introduced by genetic engineering, or a synthetic compound linker, for example, a linker disclosed in Protein Engineering, 9 (3), 299-305, 1996 Can be used.
  • a preferred linker in the present invention is a peptide linker.
  • the length of the peptide linker is not particularly limited, and can be appropriately selected by those skilled in the art according to the purpose, but is usually 1 to 100 amino acids, preferably 3 to 50 amino acids, more preferably 5 to 30 amino acids, Particularly preferred is 12 to 18 amino acids (for example, 15 amino acids).
  • amino acid sequence of the peptide linker examples include the following sequences. Ser Gly ⁇ Ser Gly ⁇ Gly ⁇ Ser Ser ⁇ Gly ⁇ Gly Gly, Gly, Gly, Ser (SEQ ID NO: 82) Ser, Gly, Gly, Gly (SEQ ID NO: 83) Gly, Gly, Gly, Gly, Ser (SEQ ID NO: 84) Ser, Gly, Gly, Gly, Gly (SEQ ID NO: 85) Gly, Gly, Gly, Gly, Gly, Ser (SEQ ID NO: 86) Ser, Gly, Gly, Gly, Gly (SEQ ID NO: 87) Gly, Gly, Gly, Gly, Gly, Gly, Ser (SEQ ID NO: 88) Ser, Gly, Gly, Gly, Gly, Gly, Gly, Gly (SEQ ID NO: 89) (Gly, Gly, Gly, Gly, Ser (SEQ ID NO: 84))
  • n which determines the length of the above peptide linker is usually 1 to 5, preferably 1 to 3, more preferably 1 or 2.
  • Synthetic compound linkers are commonly used for cross-linking peptides such as N-hydroxysuccinimide (NHS) disuccinimidyl suberate (DSS) and bis (sulfosuccinimidyl) suberate.
  • NHS N-hydroxysuccinimide
  • DSS disuccinimidyl suberate
  • SSS bis (sulfosuccinimidyl) suberate
  • BS3 dithiobis (succinimidylpropionate) (DSP), dithiobis (sulfosuccinimidylpropionate) (DTSSP), ethylene glycol bis (succinimidyl succinate) (EGS), ethylene glycol Bis (sulfosuccinimidyl succinate) (sulfo-EGS), disuccinimidyl tartrate (DST), disulfosuccinimidyl tartrate (sulfo-DST), bis [2- (succinimideoxycarbonyloxy) Ethyl] sulfone (BSOCOES), bis [2- (sulfosuccinimidooxycarbonyloxy) ethyl ] And the like sulfone (sulfo-BSOCOES), These crosslinking agents are commercially available.
  • linkers When linking 4 antibody variable regions, usually 3 linkers are required. A plurality of linkers may be the same or different linkers may be used.
  • the antibody of the present invention includes an antibody in which one or more amino acid residues are added to the amino acid sequence of the antibody of the present invention. Also included are fusion proteins in which these antibodies are fused with other peptides or proteins. In the method for producing a fusion protein, a polynucleotide encoding the antibody of the present invention and a polynucleotide encoding another peptide or polypeptide are linked so that the frames coincide with each other, introduced into an expression vector, and expressed in a host. Any technique known to those skilled in the art can be used. Other peptides or polypeptides to be subjected to fusion with the antibody of the present invention include, for example, FLAG (Hopp, T. P.
  • polypeptides to be subjected to fusion with the antibody of the present invention examples include GST (glutathione-S-transferase), HA (influenza agglutinin), immunoglobulin constant region, ⁇ -galactosidase, MBP (maltose). Binding protein) and the like. Preparing a fusion polypeptide by fusing a commercially available polynucleotide encoding the peptide or polypeptide with a polynucleotide encoding the antibody of the present invention and expressing the fusion polynucleotide prepared thereby. Can do.
  • the antibody of the present invention may be a conjugated antibody bound to various molecules such as polyethylene glycol (PEG), hyaluronic acid and other high molecular substances, radioactive substances, fluorescent substances, luminescent substances, enzymes, and toxins.
  • PEG polyethylene glycol
  • hyaluronic acid and other high molecular substances
  • radioactive substances such as radioactive substances, fluorescent substances, luminescent substances, enzymes, and toxins.
  • fluorescent substances such as fluorescent substances, luminescent substances, enzymes, and toxins.
  • enzymes such as enzymes, and toxins.
  • a conjugated antibody can be obtained by chemically modifying the obtained antibody.
  • the modification method of an antibody has already been established in this field (for example, US5057313, US5156840).
  • the “antibody” in the present invention includes these conjugated antibodies.
  • the antibody used in the present invention may be a bispecific antibody.
  • Bispecific antibodies refer to antibodies that have variable regions that recognize different epitopes within the same antibody molecule.
  • the bispecific antibody may be a bispecific antibody that recognizes different epitopes on the NR10 molecule, or one antigen binding site recognizes NR10 and the other antigen binding site is the other. Bispecific antibodies that recognize substances can also be used.
  • bispecific antibodies can be produced by combining two types of antibodies with different recognition antigens.
  • the antibody to be bound may be a 1 ⁇ 2 molecule each having an H chain and an L chain, or may be a 1 ⁇ 4 molecule consisting only of an H chain.
  • bispecific antibody-producing fused cells can be prepared by fusing hybridomas that produce different monoclonal antibodies.
  • bispecific antibodies can be produced by genetic engineering techniques.
  • the antibody of the present invention may differ in amino acid sequence, molecular weight, isoelectric point, presence / absence of sugar chain, form, etc., depending on the antibody-producing cell, host or purification method described below. However, as long as the obtained antibody has a function equivalent to the antibody of the present invention, it is included in the present invention. For example, when the antibody of the present invention is expressed in prokaryotic cells such as E. coli, a methionine residue is added to the N-terminus of the original antibody amino acid sequence. The antibody of the present invention also includes such an antibody.
  • the antibody of the present invention may be a polyclonal antibody or a monoclonal antibody.
  • a monoclonal antibody having binding activity and / or neutralizing activity for NR10 is prepared by preparing an anti-NR10 monoclonal antibody by a known method using, for example, NR10 derived from a mammal such as a human or a mouse or a fragment peptide thereof as an immunogen. It can be obtained by selecting an antibody having NR10 binding activity and / or neutralizing activity from the obtained anti-NR10 monoclonal antibody. That is, a desired antigen or a cell expressing the desired antigen is used as a sensitizing antigen and immunized according to a normal immunization method.
  • anti-NR10 monoclonal antibodies by fusing the obtained immune cells with known parental cells by ordinary cell fusion methods and screening monoclonal antibody-producing cells (hybridomas) by ordinary screening methods.
  • animals to be immunized include mammals such as mice, rats, rabbits, sheep, monkeys, goats, donkeys, cows, horses, and pigs.
  • the antigen can be prepared using a known NR10 gene sequence according to a known method, for example, a method using a baculovirus (WO98 / 46777 etc.).
  • Hybridoma can be prepared according to, for example, the method of Milstein et al. (Kohler. G. Milstein, C., Methods Enzymol. (1981) 73: 3-46).
  • immunization may be performed by binding to an immunogenic macromolecule such as albumin.
  • An embodiment of the antibody having binding activity and / or neutralizing activity for NR10 of the present invention includes a monoclonal antibody having binding activity and / or neutralizing activity for human NR10.
  • the immunogen for producing a monoclonal antibody having binding activity and / or neutralizing activity against human NR10 is not particularly limited as long as an antibody having binding activity and / or neutralizing activity against human NR10 can be produced.
  • human NR10 is known to have a plurality of variants, but any variant may be used as an immunogen as long as an antibody having binding activity and / or neutralizing activity against human NR10 can be produced.
  • an NR10 fragment peptide or a natural NR10 sequence with an artificial mutation may be used as an immunogen.
  • Human NR10.3 is one of the preferred immunogens for producing antibodies having binding activity and / or neutralizing activity for NR10 of the present invention.
  • the measurement of the binding activity and / or neutralizing activity of the antibody against NR10 can be performed, for example, by the method for observing the growth inhibitory effect of the IL-31-dependent cell line described in the Examples.
  • DNA immunization refers to immunization by administering a vector DNA constructed in such a manner that a gene encoding an antigen protein can be expressed in an immunized animal, and expressing the immunizing antigen in the body of the immunized animal. It is a method of giving a stimulus.
  • DNA immunization can be expected to have the following advantages. -The structure of the membrane protein can be maintained to provide immune stimulation-There is no need to purify the immune antigen. However, in DNA immunization, it is difficult to combine with immune stimulation means such as an adjuvant.
  • DNA encoding NR10 is administered to an immunized animal.
  • DNA encoding NR10 can be synthesized by a known method such as PCR.
  • the obtained DNA is inserted into an appropriate expression vector and administered to an immunized animal.
  • the expression vector for example, a commercially available expression vector such as pcDNA3.1 can be used.
  • a method of administering the vector to a living body a generally used method can be used.
  • DNA immunization can be performed by implanting gold particles adsorbed with an expression vector into cells with a gene gun. Boosting with NR10-expressing cells after DNA immunization is a preferred method for obtaining monoclonal antibodies.
  • immune cells are collected from the mammal and subjected to cell fusion.
  • spleen cells can be used.
  • Mammalian myeloma cells are used as cells to be fused with the above immune cells.
  • the myeloma cell is preferably provided with an appropriate selection marker for screening.
  • a selectable marker refers to a trait that can (or cannot) survive under certain culture conditions.
  • Known selection markers include hypoxanthine-guanine-phosphoribosyltransferase deficiency (hereinafter abbreviated as HGPRT deficiency) or thymidine kinase deficiency (hereinafter abbreviated as TK deficiency).
  • HGPRT deficiency hypoxanthine-guanine-phosphoribosyltransferase deficiency
  • TK deficiency thymidine kinase deficiency
  • Cells having HGPRT or TK deficiency have hypoxanthine-aminopterin-thymidine sensitivity (hereinafter abbreviated as HAT sensitivity).
  • HGPRT-deficient or TK-deficient cells can be selected in a medium containing 6 thioguanine, 8 azaguanine (hereinafter abbreviated as 8AG), or 5 'bromodeoxyuridine, respectively.
  • 8AG 8 azaguanine
  • Normal cells die because they incorporate these pyrimidine analogs into the DNA, but cells deficient in these enzymes cannot survive these pyrimidine analogs and can survive in selective media.
  • a selectable marker called G418 resistance confers resistance to 2-deoxystreptamine antibiotics (gentamicin analogs) with a neomycin resistance gene.
  • gentamicin analogs gentamicin analogs
  • immune cells and myeloma cells according to known methods such as the method of Kohler and Milstein et al. (Kohler. Ler G. and Milstein, C., Methods Enzymol. (1981) 73, 3-46) And cell fusion.
  • cell fusion can be carried out in a normal nutrient culture medium in the presence of a cell fusion promoter.
  • a cell fusion promoter for example, polyethylene glycol (PEG), Sendai virus (HVJ) or the like can be used.
  • an auxiliary agent such as dimethyl sulfoxide can be added as desired in order to increase the fusion efficiency.
  • the usage ratio of immune cells and myeloma cells can be set arbitrarily.
  • the number of immune cells is preferably 1 to 10 times that of myeloma cells.
  • the culture solution used for cell fusion for example, RPMI1640 culture solution suitable for growth of myeloma cell line, MEM culture solution, and other normal culture solutions used for this type of cell culture can be used.
  • serum supplements such as fetal calf serum (FCS) can be added to the culture medium.
  • a predetermined amount of immune cells and myeloma cells are mixed well in a culture solution, and a target PEG (hybridoma) is formed by mixing a PEG solution preheated to about 37 ° C.
  • a target PEG hybrida
  • PEG having an average molecular weight of about 1000 to 6000 can be usually added at a concentration of 30 to 60% (w / v).
  • cell fusion agents and the like that are undesirable for the growth of hybridomas are removed by sequentially adding the appropriate culture medium listed above, and then centrifuging to remove the supernatant.
  • the hybridoma obtained in this manner can be selected by using a selective culture solution corresponding to the selection marker possessed by the myeloma used for cell fusion.
  • a selective culture solution corresponding to the selection marker possessed by the myeloma used for cell fusion.
  • cells having HGPRT or TK deficiency can be selected by culturing in a HAT culture solution (a culture solution containing hypoxanthine, aminopterin and thymidine). That is, when HAT-sensitive myeloma cells are used for cell fusion, cells that have succeeded in cell fusion with normal cells can be selectively proliferated in the HAT culture solution.
  • the culture using the HAT culture solution is continued for a time sufficient for cells other than the target hybridoma (non-fusion cells) to die.
  • the target hybridoma can be selected by culturing for several days to several weeks. Subsequently, by carrying out the usual limiting dilution method, screening and single cloning of the hybridoma producing the target antibody can be performed.
  • an antibody that recognizes NR10 can be produced by the method described in International Publication WO03 / 104453.
  • Screening and single cloning of the target antibody can be suitably performed by a screening method based on a known antigen-antibody reaction.
  • the antigen is bound to a carrier such as beads made of polystyrene or the like, or a commercially available 96-well microtiter plate, and reacted with the culture supernatant of the hybridoma.
  • a secondary antibody labeled with an enzyme is reacted. If the culture supernatant contains an antibody of interest that reacts with the sensitizing antigen, the secondary antibody binds to the carrier via this antibody. By detecting the secondary antibody that finally binds to the carrier, it can be determined whether the antibody of interest is present in the culture supernatant.
  • substantially the same NR10 protein can be preferably used as the antigen, including those used for immunization.
  • a cell line expressing NR10, an extracellular domain of NR10, or an oligopeptide consisting of a partial amino acid sequence constituting the region can be used as an antigen.
  • a target antibody can be obtained by sensitizing human lymphocytes with an antigen.
  • human lymphocytes are first sensitized with NR10 protein in vitro.
  • the immunized lymphocytes are then fused with an appropriate fusion partner.
  • the fusion partner for example, a myeloma cell derived from human and having a permanent division ability can be used (see Japanese Patent Publication No. 1-59878).
  • the antibody obtained by this method is a human antibody having binding activity to the NR10 protein.
  • the base sequence and amino acid sequence encoding the anti-NR10 antibody obtained by the above method can be obtained by methods known to those skilled in the art.
  • the amino acids contained in the amino acid sequences described in the present invention are modified after translation (for example, modification to pyroglutamic acid by pyroglutamylation of N-terminal glutamine is a modification well known to those skilled in the art). In some cases, even if the amino acid is post-translationally modified as such, it is naturally included in the amino acid sequence described in the present invention.
  • a polynucleotide encoding the antibody is constructed based on the sequence of the antibody recognizing NR10, introduced into an expression vector, and then expressed in an appropriate host cell (for example, Co, M . S. et al., J. Immunol. (1994) 152, 2968-2976; Better, M. and Horwitz, A. H., Methods Enzymol. (1989) 178, 476-496; Pluckthun, A. and Skerra , A., Methods Enzymol.
  • vectors examples include M13 vectors, pUC vectors, pBR322, pBluescript, and pCR-Script.
  • pGEM-T for the purpose of subcloning and excision of cDNA
  • An expression vector is particularly useful when a vector is used for the purpose of producing the antibody of the present invention.
  • the host is E. coli such as JM109, DH5 ⁇ , HB101, XL1-Blue, etc.
  • promoters that can be expressed efficiently in E.
  • coli such as the lacZ promoter (Ward et al., Nature (1989) 341, 544-546; FASEB J. (1992) 6, 2422-2427), araB promoter (Better et al. , (Science) (1988) (240), (1041-1043), or T7 promoter is essential.
  • lacZ promoter Ward et al., Nature (1989) 341, 544-546; FASEB J. (1992) 6, 2422-2427
  • araB promoter Better et al. , (Science) (1988) (240), (1041-1043
  • T7 promoter is essential.
  • examples of such vectors include pGEX-5X-1 (Pharmacia), “QIAexpress system” (Qiagen), pEGFP, or pET (in this case, the host expresses T7 RNA polymerase BL21). are preferred).
  • the vector may also contain a signal sequence for antibody secretion.
  • a signal sequence for antibody secretion a pelB signal sequence (Lei, S. P. et al J. Bacteriol. (1987) 169, 4379) may be used when the periplasm of E. coli is produced.
  • Introduction of a vector into a host cell can be performed using, for example, a calcium chloride method or an electroporation method.
  • vectors for producing the antibody of the present invention include mammalian-derived expression vectors (for example, pcDNA3 (manufactured by Invitrogen)), pEF-BOS® (Nucleic® Acids.® Res.
  • insect cell-derived expression vectors eg, “Bac-to-BAC baculovairus expression system” (manufactured by Gibco BRL), pBacPAK8), plant-derived expression vectors (eg, pMH1, pMH2) Animal virus-derived expression vectors (for example, pHSV, pMV, pAdexLcw), retrovirus-derived expression vectors (for example, pZIPneo), yeast-derived expression vectors (for example, “Pichia® Expression® Kit” (manufactured by Invitrogen), pNV11, SP-Q01), and an expression vector derived from Bacillus subtilis (for example, pPL608, pKTH50).
  • Bacillus subtilis for example, pPL608, pKTH50.
  • promoters necessary for expression in cells such as the SV40 promoter (Mulligan et al., Nature (1979) 277, 108), It is essential to have the MMLV-LTR promoter, EF1 ⁇ promoter (Mizushima et al., Nucleic Acids Res. (1990) 18, 5322), CMV promoter, etc., and genes for selecting transformation into cells (for example, More preferably, it has a drug resistance gene that can be discriminated by a drug (neomycin, G418, etc.). Examples of such a vector include pMAM, pDR2, pBK-RSV, pBK-CMV, pOPRSV, and pOP13.
  • a vector having a DHFR gene complementary to CHO cells lacking the nucleic acid synthesis pathway is introduced and amplified with methotrexate (MTX), and for the purpose of transient gene expression
  • MTX methotrexate
  • the expression vectors are selectable markers: aminoglycoside transferase (APH) gene, thymidine kinase (TK) gene, E. coli xanthine guanine phosphoribosyltransferase (Ecogpt) gene, dihydrofolate reductase ( dhfr) gene and the like.
  • APH aminoglycoside transferase
  • TK thymidine kinase
  • Ecogpt E. coli xanthine guanine phosphoribosyltransferase
  • dhfr dihydrofolate reductase
  • the present invention is encoded by a polypeptide of the present invention or a gene encoding a polypeptide of the present invention, comprising the step of culturing a host cell containing a vector into which a polynucleotide encoding the polypeptide of the present invention has been introduced.
  • a method for producing a polypeptide is provided. More specifically, a method for producing the polypeptide of the present invention comprising the following steps is provided. (a) culturing a host cell containing a vector into which a gene encoding the polypeptide of the present invention has been introduced, (b) A step of obtaining a polypeptide encoded by the gene.
  • the antibody of the present invention thus obtained can be isolated from the inside of the host cell or outside the cell (medium etc.) and purified as a substantially pure and homogeneous antibody. Separation and purification of antibodies may be carried out using separation and purification methods used in normal antibody purification, and are not limited in any way. For example, chromatography column, filter, ultrafiltration, salting out, solvent precipitation, solvent extraction, distillation, immunoprecipitation, SDS-polyacrylamide gel electrophoresis, isoelectric focusing, dialysis, recrystallization, etc. are appropriately selected, When combined, antibodies can be separated and purified.
  • chromatography examples include affinity chromatography, ion exchange chromatography, hydrophobic chromatography, gel filtration, reverse phase chromatography, adsorption chromatography, etc. (Strategies for Protein Purification and Characterization: A Laboratory Course Manual. Ed Daniel R. Marshak et al., Cold Spring Harbor Laboratory Press, 1996). These chromatography can be performed using liquid phase chromatography, for example, liquid phase chromatography such as HPLC and FPLC.
  • liquid phase chromatography such as HPLC and FPLC.
  • the column used for affinity chromatography include a protein A column and a protein G column. Examples of the column using protein A include Hyper D, POROS, Sepharose F (GE Amersham Biosciences), and the like.
  • the present invention also encompasses antibodies highly purified using these purification methods.
  • Measurement of the binding activity of the obtained antibody to NR10 can be performed by methods known to those skilled in the art.
  • ELISA enzyme-linked immunosorbent assay
  • EIA enzyme immunoassay
  • RIA radioimmunoassay
  • fluorescent antibody method can be used as a method for measuring the antigen-binding activity of an antibody.
  • an enzyme immunoassay a sample containing an antibody, for example, a culture supernatant of an antibody-producing cell or a purified antibody is added to a plate coated with an antigen.
  • compositions and this invention provide the pharmaceutical composition which contains the above-mentioned antibody as an active ingredient. Furthermore, this invention provides the therapeutic agent of the inflammatory disease which uses the above-mentioned antibody as an active ingredient.
  • an inflammatory disease is a pathological condition related to cytological / histological reactions occurring in affected blood vessels and adjacent tissues due to damage or abnormal stimulation by a physical, chemical, or biological agent. This refers to a disease with findings (Stedman Medical Dictionary 5th edition, Medical Review Inc., 2005).
  • inflammatory diseases are dermatitis (atopic dermatitis, chronic dermatitis, etc.), inflammatory bowel disease (colitis, etc.), asthma, arthritis (rheumatoid arthritis, osteoarthritis, etc.), bronchitis , Th-2 autoimmune disease, systemic lupus erythematosus, myasthenia gravis, chronic GVHD, Crohn's disease, osteoarthritis spondylitis, low back pain, gout, post-traumatic inflammation, swelling relief, neuralgia, sore throat, bladder
  • hepatitis nonalcoholic steatohepatitis, alcoholic hepatitis, etc.
  • hepatitis B hepatitis C
  • arteriosclerosis pruritus.
  • Preferred examples of the inflammatory disease that is the subject of the present invention include atopic dermatitis, chronic dermatitis, rheumatism, osteoarthritis, chronic asthma, and pruritus.
  • “Containing anti-NR10 antibody as an active ingredient” means that the anti-NR10 antibody is contained as at least one active ingredient, and does not limit the content ratio.
  • the therapeutic agent for inflammatory diseases of the present invention may contain a component that promotes treatment of other inflammatory diseases in combination with the anti-NR10 antibody described above.
  • the therapeutic agent of the present invention may be used for preventive purposes.
  • the anti-NR10 antibody of the present invention can be formulated according to a conventional method (for example, Remington's Pharmaceutical Science, Latest Edition, Mark Publishing Company, Easton, USA). Further, if necessary, a pharmaceutically acceptable carrier and / or additive may be included. For example, surfactants (PEG, Tween, etc.), excipients, antioxidants (ascorbic acid, etc.), coloring agents, flavoring agents, preservatives, stabilizers, buffering agents (phosphoric acid, citric acid, other organic acids) Etc.), chelating agents (EDTA, etc.), suspending agents, tonicity agents, binders, disintegrants, lubricants, fluidity promoters, flavoring agents, and the like.
  • a pharmaceutically acceptable carrier and / or additive may be included.
  • surfactants PEG, Tween, etc.
  • excipients antioxidants (ascorbic acid, etc.)
  • coloring agents for example, flavoring agents, preservatives, stabilizers, buffering agents (
  • the preventive or therapeutic agent for inflammatory diseases of the present invention is not limited to these, and may contain other conventional carriers as appropriate.
  • the anti-NR10 antibody may contain other low molecular weight polypeptides, proteins such as serum albumin, gelatin and immunoglobulin, and amino acids such as glycine, glutamine, asparagine, arginine and lysine.
  • an aqueous solution for injection the anti-NR10 antibody is dissolved in an isotonic solution containing, for example, physiological saline, glucose or other adjuvants.
  • adjuvants examples include D-sorbitol, D-mannose, D-mannitol, sodium chloride, and further suitable solubilizers such as alcohol (ethanol, etc.), polyalcohol (propylene glycol, PEG, etc.), A nonionic surfactant (polysorbate 80, HCO-50) may be used in combination.
  • solubilizers such as alcohol (ethanol, etc.), polyalcohol (propylene glycol, PEG, etc.), A nonionic surfactant (polysorbate 80, HCO-50) may be used in combination.
  • anti-NR10 antibody can be encapsulated in microcapsules (microcapsules such as hydroxymethylcellulose, gelatin, poly [methylmethacrylic acid]) or colloid drug delivery systems (liposomes, albumin microspheres, microemulsions, nanoparticles and (See, for example, Remington's Pharmaceutical Science 16th edition & Oslo Ed. (1980)).
  • a method of making a drug a sustained-release drug is also known and can be applied to the antibody NR10 antibody (Langer et al., J. Biomed. Mater. Res. (1981) 15: 167-277; Langer, Chem Tech. (1982) 12:; 98-105; U.S. Patent 3,773,919; European Patent Application Publication (EP) 58,481; Sidman et al., Biopolymers (1983) 22: 547-56; EP 133,988).
  • microcapsules such as hydroxymethylcellulose, gelatin, poly [methylmethacrylic acid]
  • the pharmaceutical composition of the present invention can be administered either orally or parenterally, but is preferably administered parenterally. Specifically, it is administered to a patient by injection and transdermal administration. As an example of the injection form, it can be administered systemically or locally by, for example, intravenous injection, intramuscular injection or subcutaneous injection. Local injection, particularly intramuscular injection, may be performed at or around the site where inflammation is to be suppressed.
  • the administration method can be appropriately selected depending on the age and symptoms of the patient.
  • the dosage can be selected, for example, within the range of 0.0001 mg to 100 mg of active ingredient per kg of body weight per time.
  • the active ingredient when administered to a human patient, can be selected within a range of 0.001 to 1000 mg / kg ⁇ body ⁇ weight per patient.
  • the antibody of the present invention is 0.01
  • An amount of about 50 mg / kg ⁇ body ⁇ weight is preferably included.
  • the preventive or therapeutic agent for inflammatory diseases of the present invention is not limited to these doses.
  • Hybridoma production 1.1 Preparation of DNA immunization human NR10 plasmid and cynomolgus monkey NR10 plasmid 1.1.1. Preparation of hNR10 and cynNR10 Expression Vector Human NR10 (base sequence SEQ ID NO: 75, amino acid sequence SEQ ID NO: 76) is expressed in vector pMacII (WO2005 / 054467) that expresses protein under the control of mouse ⁇ -actin promoter. was used as an integrated hNR10 expression vector. Similarly, a cynNR10 expression vector was constructed from cynomolgus monkey NR10 (base sequence SEQ ID NO: 65, amino acid sequence SEQ ID NO: 66).
  • 1.2. Production of anti-human NR10 antibody-producing hybridoma 1.2.1. Hybridoma production using human NR10 and cynomolgus monkey NR10 immunized mice 10 Balb / c mice (female, 6 weeks old at the start of immunization, Charles River, Japan) were immunized with human NR10 or cynomolgus monkey NR10 as follows. did. As a first immunization, a DNA cartridge prepared with the hNR10 expression vector was immunized using the Herios Gene Gun system (BIO-RAD). The second immunization was performed 1 week later by administering a DNA cartridge prepared with the cynNR10 expression vector using the Herios Gene Gun system.
  • BIO-RAD Herios Gene Gun system
  • HAT selection medium (10% FBS / RPMI1640, 2 vol% HAT 50x concentrate (Dainippon Pharmaceutical), 5 vol% BM-Condimed H1 (Roche Diagnostics) ) was inoculated in a 96-well plate with 1 colony per well. After culturing for 3 to 4 days, the culture supernatant of each well was collected, and the mouse IgG concentration in the culture supernatant was measured.
  • the culture supernatant in which mouse IgG was confirmed was evaluated by neutralizing activity using human IL-31-dependent cell lines (hNR10 / hOSMR / BaF3 cells; Reference Example 2), and some of them have high NR10 neutralizing activity. Clones were obtained (FIG. 3). A clone that suppresses cell growth induced by human IL-31 in a concentration-dependent manner and a cell that suppresses cell growth induced by cynomolgus monkey IL-31 (cynNR10 / cynOSMR / BaF3 cells; Reference Example 2) in a concentration-dependent manner were obtained. (FIG. 4).
  • FIG. 1 shows the H chain variable region
  • FIG. 2 shows the L chain variable region of the amino acid sequences of the obtained mouse antibodies NS18, NS22, NS23 and NS33.
  • PCR was performed with PrimeSTAR HS DNA Polymerase (TaKaRa) using the primer combinations shown in Table 1, and the obtained amplified fragments were converted into constant regions (human ⁇ 1 or It was combined with ⁇ 2, human ⁇ ) and inserted into an animal cell expression vector.
  • the base sequence of each DNA fragment was determined using the BigDye
  • Kit Applied
  • Biosystems) according to the method of an attached description.
  • CHO-S-SFMII medium to 700 ⁇ L to the prepared plasmid DNA mixture (13.8 ⁇ g total), add 10.7 ⁇ L / mL Polyethylenimine (Polysciences Inc.) 20.7 ⁇ L, mix and leave for 10 minutes at room temperature. Cells were loaded and incubated for 4-5 hours in a CO 2 incubator (5% CO 2 at 37 ° C.). Then, 6.9 mL of CHO-S-SFMII (Invitrogen) medium was added and cultured in a CO 2 incubator for 3-4 days.
  • the cells were removed by centrifugation (about 2000 g, 5 minutes, room temperature), and further passed through a 0.22 ⁇ m filter MILLEX (registered trademark) -GV (Millipore). Each sample was stored at 4 ° C until use.
  • the antibody was purified from the supernatant using Protein G Sepharose (Amersham Biosciences). The purified antibody was concentrated using Amicon Ultra 15 (Millipore), and the solvent was replaced with PBS ( ⁇ ) containing 0.05% NaN 3 using PD-10 Desalting columns (Amersham Biosciences). Absorbance at 280 nm was measured with an ND-1000 Spectrophotometer (NanoDrop), and the concentration was calculated by the method of Pace et al. (Protein Science (1995) 4: 2411-2423).
  • hNR10 / hOSMR / BaF3 cells were prepared in RPMI1640 medium (GIBCO) containing 10% FBS (MOREGATE) and 1% Penicillin-Streptomycin (Invitrogen) so as to be 1.5 ⁇ 10 5 cells / mL. A part of this was added and hIL-31 (R & D Systems) was added to 4 ng / mL (IL-31 (+), final conc .; 2 ng / mL). The remaining cell suspension was designated IL-31 (-).
  • the purified NS22 was adjusted to 2 ⁇ g / mL with a medium, and a dilution ratio of 3 and a total of 8 series of dilutions were prepared (final conc.; ⁇ 1 ⁇ g / mL).
  • Each well of a 96-well flat bottom plate (CORNING) was seeded with 50 ⁇ L of the cell suspension and a diluted solution of chimeric NS22 (human ⁇ 1, ⁇ ), and cultured for 2 days in a 37 ° C., 5% CO 2 incubator.
  • a mixed solution of Cell Counting Kit-8 (Dojindo) and PBS in equal amounts was added to each well, and the absorbance (450 nm / 620 nm) was measured (TECAN, SUNRISE CLASSIC). After reacting for 2 hours in a 37 ° C., 5% CO 2 incubator, the absorbance was measured again.
  • the neutralization activity of NS22 was expressed as an inhibition rate using a value obtained by subtracting the 0 hour value from the 2 hour value.
  • NS22 was found to inhibit cell growth induced by IL-31 stimulation in a concentration-dependent manner in the hNR10 / hOSMR / BaF3 cell line, and proved to have neutralizing activity against human IL-31 signaling. (FIG. 5).
  • DU145 cell line human prostate cancer cell line showing IL-6 production induction by IL-31 stimulation
  • IL-31 neutralizing activity was evaluated as shown below.
  • a total of 6 dilutions were prepared, each was mixed 1: 1 with 100 ng / mL human interleukin-31 (R & D systems), and 50 ⁇ L was added to each well.
  • the IL-6 concentration in the culture supernatant after culturing at 37 ° C. under 5% CO 2 for 2 days was measured using DuoSet ELISA Development kit (R & D systems).
  • NS22 neutralization activity was evaluated as an inhibition rate (%).
  • IL-6 concentration (A) in the absence of IL-31 is the maximum inhibitory activity (100% inhibition)
  • IL-6 concentration (B) in the absence of IL-31 and NS22 is inhibited
  • the IL-6 concentration (C) under the addition of IL-31 and NS22 was calculated from the following formula.
  • Inhibition rate (%) (BC) / (BA) x 100
  • NS22 was found to suppress IL-6 production by IL-31 stimulation in a concentration-dependent manner in the DU145 cell line, and proved to have neutralizing activity against human IL-31 signaling. (FIG. 6).
  • IL-31 / NR10 binding inhibitory activity of the antibodies was evaluated as shown below.
  • NS22 and NA633 (constant regions are ⁇ 1, ⁇ , respectively) with assay buffer (10 mM HEPES, 140 mM NaCl, 2.5 mM CaCl 2 , 3 mM MgCl 2 , 2% FBS, 0.01% NaN 3 ) Dilution was further carried out, and a total of 7 dilutions with a dilution ratio of 2 was prepared and added to a plate (96-Well FMAT Plates, Applied Biosystems) at 40 ⁇ L / well. Subsequently, FMAT Blue-labeled hIL-31 was diluted 400 times with Assay buffer and added at 20 ⁇ L / well.
  • assay buffer 10 mM HEPES, 140 mM NaCl, 2.5 mM CaCl 2 , 3 mM MgCl 2 , 2% FBS, 0.01% NaN 3
  • Example 3 Competition of anti-NR10 antibody against NR10 NS22 antibody purified from the hybridoma culture supernatant was labeled with FMAT Blue (Applied Biosystems, 4328853). 17 ⁇ L 1 M NaHCO 3 solution and 3.4 ⁇ L of 17 nmoles FMAT Blue dissolved in DMSO were added to 170 ⁇ L of NS22 prepared in 1 mg / ml-PBS, vortexed, and allowed to stand at room temperature for 30 minutes.
  • FMAT Blue Applied Biosystems, 4328853
  • Example 4 Humanization of NS22 antibody Selection of each framework sequence The variable region of mouse NS22 antibody was compared with the human germline sequence. Among them, FR sequences used for humanization are summarized in Table 2. CDR and FR were determined according to Kabat numbering. As humanized variable regions, the H chain is the sequence consisting of FR1, FR2, FR3_1, FR4 listed in Table 2 as H0-VH (SEQ ID NO: 50), and the sequence consisting of FR1, FR2, FR3_2, FR4 is H1. -VH (SEQ ID NO: 112). In addition, the L chain has a sequence composed of FR1, FR2, FR3 and FR4 as L0 (SEQ ID NO: 52).
  • variable region of humanized NS22 H0L0 In order to create a variable region of humanized NS22 in which the CDR region of NS22 was transplanted into the FR region used for humanization, a synthetic oligo DNA was designed for each of the H chain and L chain. Each synthetic oligo DNA was mixed, and a gene encoding the variable region of humanized NS22 was prepared by assembly PCR. Assemble PCR was performed using KOD-Plus (TOYOBO), and PCR was performed according to the following conditions. The reaction mixture consisting of the attached PCR Buffer, dNTPs, MgSO 4 , KOD-Plus and 10 pmol synthetic oligo DNA was heated at 94 ° C.
  • H0-SKSC SEQ ID NO: 54
  • L0 SEQ ID NO: 56
  • the base sequence of each DNA fragment was determined using the BigDye Terminator Cycle Sequencing Kit (Applied Biosystems) with the DNA sequencer ABI PRISM 3730xL DNA Sequencer or ABI PRISM 3700 DNA Sequencer (Applied Biosystems) according to the method described in the attached instructions.
  • H1-SKSC Preparation of variable region of humanized NS22 H1
  • H1-SKSC SEQ ID NO: 130
  • E 73rd glutamine
  • K lysine
  • the mutant was prepared using a commercially available QuikChange Site-Directed Mutagenesis Kit (Stratagene), and the mutant was prepared according to the method described in the attached instructions.
  • HEK293H derived from human fetal kidney cancer cells is suspended in DMEM medium (Invitrogen) containing 10% Fetal Bovine Serum (Invitrogen), and the dish (diameter) has a cell density of 5-6 ⁇ 10 5 cells / mL. 10 mL to each dish of 10 cm, CORNING) After incubating overnight in a CO 2 incubator (37 ° C, 5% CO 2 ), the medium was removed by suction and 6.9 mL of CHO-S-SFMII (Invitrogen) medium was added.
  • the prepared plasmid DNA mixture (13.8 ⁇ g in total) was mixed with 20.7 ⁇ L of 1 ⁇ g / mL Polyethylenimine (Polysciences Inc.) and 690 ⁇ L of CHO-S-SFMII medium and allowed to stand for 10 minutes at room temperature. Incubated in a CO 2 incubator (5% CO 2 at 37 ° C.) for 4-5 hours. Then, 6.9 mL of CHO-S-SFMII (Invitrogen) medium was added and cultured in a CO 2 incubator for 3 days.
  • IgG antibody 50 ⁇ L of rProtein A Sepharose TM Fast Flow (Amersham Biosciences) suspended in TBS was added to the obtained culture supernatant and mixed by inverting at 4 ° C. for 4 hours or more.
  • the solution was transferred to a 0.22 ⁇ m filter cup Ultrafree (registered trademark) -MC (Millipore), washed 3 times with 500 ⁇ L of TBS, and then suspended in 100 ⁇ L of 50 mM sodium acetate aqueous solution, pH 3.3 in rProtein A Sepharose TM resin. After standing for 3 minutes, the antibody was eluted. Immediately, 6.7 ⁇ L of 1.5 M Tris-HCl, pH 7.8 was added for neutralization.
  • NS22 chimeric antibody and NS22_H0L0 H chain H0-SKSC / SEQ ID NO: 54, L chain L0 / SEQ ID NO: 56
  • assay buffer 10 mM HEPES, 140 mM NaCl, 2.5 mM CaCl 2 , 3 mM MgCl 2 , 2% FBS, 0.01% NaN 3 , pH 7.4
  • a dilution ratio of 2, and a total of 8 series of dilutions are prepared, and the plate (96-Well FMAT Plates, Applied Biosystems).
  • FMAT Blue-labeled hIL-31 was diluted 400 times with Assay buffer and added at 20 ⁇ L / well.
  • the cell suspension adjusted to 2.5 ⁇ 10 5 / mL with Assay buffer was added at 40 ⁇ L / well (final 1 ⁇ 10 4 / well).
  • fluorescence FL1 was measured with 8200 Cellular Detection System (Applied Biosystems).
  • humanized NS22 antibody H0L0 (H chain H0-SKSC / SEQ ID NO: 54, L chain L0 / SEQ ID NO: 56), H1L0 (H chain H1-SKSC / SEQ ID NO: 130, L chain L0 / SEQ ID NO: : 56) showed almost the same competitive activity as the chimeric antibody as shown in FIG. 9, and thus can be said to be a H0L0, H1L0 humanized anti-IL-31 receptor antibody. Further, it is considered that both FRs used for H0L0 and H1L0 can be used as FRs during humanization.
  • Example 5 Heterogeneity reducing effect by novel constant regions M14 and M58 in humanized anti-IL31 receptor antibody As shown in Reference Examples 7 to 9, in the huPM1 antibody which is a humanized anti-IL-6 receptor antibody, the constant region is It was confirmed that by converting from IgG2 to M14 or M58, heterogeneity derived from the hinge region of IgG2 can be reduced without reducing stability. Thus, it was examined whether the heterogeneity of humanized anti-IL-31 receptor antibody can be reduced by converting the constant region from wild-type IgG2 to M14 or M58.
  • Example 4 including IgG1 (SEQ ID NO: 60) and IgG2 (SEQ ID NO: 132) containing M14 (SEQ ID NO: 129) and M58 (SEQ ID NO: 128) prepared in Reference Examples 8 and 9 as H chains.
  • Evaluation by cation exchange chromatography was performed as a method for evaluating heterogeneity.
  • the heterogeneity of the prepared antibody was evaluated by using ProPac WCX-10 (Dionex) as a column, 20 m Sodium Acetate, pH5.0 as mobile phase A, 20 m Sodium Acetate, 1 MNaCl, pH5 as mobile phase B 0.0 was used with the appropriate flow rate and gradient.
  • the results of evaluation by cation exchange chromatography (IEC) are shown in FIG.
  • the heterogeneity is increased by converting the constant region from IgG1 to IgG2, and the constant region is converted to M14 or M58. It was also confirmed that heterogeneity can be reduced.
  • Example 6 Pharmacokinetic improvement effect of novel constant region M58 in anti-IL-31 receptor antibody As shown in Reference Example 9, in the huPM1 antibody which is an anti-IL-6 receptor antibody, the constant region is converted from IgG1 to M58. Thus, it was found that the binding to human FcRn was improved and the pharmacokinetics was improved in human FcRn transgenic mice. Therefore, it was investigated whether anti-IL-31 receptor antibody can improve pharmacokinetics by converting the constant region to M58.
  • H0L0-IgG1 H chain H0-IgG1 / SEQ ID NO: 133, L chain L0 / SEQ ID NO: 56
  • H0L0-M58 H chain H0-M58 / SEQ ID NO: 136
  • the binding to human FcRn was evaluated for the L chain ⁇ L0 / SEQ ID NO: 56) by the method shown in Reference Example 9. The results are shown in Table 3.
  • Example 7 Identification of mutation sites that lower the isoelectric point Preparation of mutants Each mutant was prepared by a method according to Example 4 or by performing Assemble PCR using PCR. In the method using Assemble PCR, oligo DNAs designed based on the normal and reverse strand sequences including the modification site are synthesized.
  • a reverse-strand oligo DNA that binds to a normal-chain oligo DNA containing the modification site and a vector in which the gene to be modified is inserted, or a reverse-strand oligo DNA that contains the modification site and a vector in which the gene to be modified is inserted By combining the respective normal strand oligo DNAs to be bound and performing PCR using PrimeSTAR (TAKARA), two fragments containing the modified site were prepared on the 5 ′ end side and the 3 ′ end side. Each mutant was generated by joining the two fragments by Assemble PCR. The prepared mutant was inserted into an expression vector that enables expression of the inserted gene in animal cells, and the base sequence of the obtained expression vector was determined by a method known to those skilled in the art. Antibody production and purification were performed according to the method of Example 4.
  • H0L0 H chain H0-SKSC / SEQ ID NO: 54, L chain L0 / SEQ ID NO: 56
  • the modified site that can reduce the isoelectric point of the variable region Study was carried out.
  • HIL-31 / hNR10 binding inhibitory activity of each variant was evaluated using FMAT. The method was performed according to the method of Example 4. As shown in FIG. 11, the competitive activity of each variant did not show a significant decrease compared to that of H0L0.
  • * indicates a modified part that is not related to the isoelectric point in order to obtain a human sequence.
  • humanized NS22 antibodies with a reduced isoelectric point combining these modifications include Hp3Lp15 (H chain Hp3-SKSC / SEQ ID NO: 151, L chain Lp15 / SEQ ID NO: 152).
  • Hp3Lp15 H chain Hp3-SKSC / SEQ ID NO: 151, L chain Lp15 / SEQ ID NO: 1512.
  • the affinity of Hp3Lp15 for NR10, isoelectric point, and plasma retention in mice were compared to H0L0.
  • Phast-Gel Dry IEF (Amersham Biosciences) gel was swollen for about 30 min with the following swelling solution using Phastsystem Cassette (Amersham Biosciences). Milli-Q water 1.5 mL Pharmalyte 5-8 for IEF (Amersham Biosciences) 100 ⁇ L
  • Step 1 2000 V 2.5 mA 3.5 W 15 ° C 75 Vh
  • Step 2 200 V 2.5 mA 3.5 W 15 ° C 15 Vh
  • Step 3 2000 V 2.5 mA 3.5 W 15 ° C 410 Vh
  • the gel after electrophoresis was fixed with 20% TCA, and then silver staining was performed using Silver-staining Kit, protein (Amersham Biosciences) according to the protocol attached to the kit. After staining, the isoelectric point of the sample (full-length antibody) was calculated from the known isoelectric point of the pI marker.
  • the isoelectric point of H0L0 is approximately 7.8, and the isoelectric point of Hp3Lp15 is approximately 5.5. Therefore, the isoelectric point of Hp3Lp15 is the isoelectric point of H0L0. It was about 2.3 lower than The theoretical isoelectric point of variable region VH / VL was calculated by GENETYX (GENETYX CORPORATION). The theoretical isoelectric point of variable region of H0L0 was 7.76, and the theoretical isoelectric point of variable region of Hp3Lp15 was 4.63. The theoretical isoelectric point of Hp3Lp15 decreased by 3.13 compared with H0L0.
  • H0L0 and Hp3Lp15 were administered to mice (C57BL / 6J, Nihon Charles River) as a single intravenous dose of 1 mg / kg and plasma concentrations were compared. Plasma concentration was measured by ELISA.
  • An immunoplate (Nunc-Immuno Plate, MaxiSorp (manufactured by Nalge nunc International)) in which a calibration curve sample and a plasma measurement sample of an appropriate concentration were immobilized on an anti-human IgG (Fc-specific) antibody (manufactured by Sigma)
  • the solution was dispensed and allowed to stand at room temperature for 1 hour.
  • Goat Anti-Human IgG-ALP manufactured by Sigma
  • color reaction was performed using BluePhos Microwell Phosphatase Substrates System (manufactured by Kirkegaard & Perry Laboratories) as a substrate, and 650 using a microplate reader.
  • the absorbance at nm was measured.
  • the plasma concentration was calculated from the absorbance of the calibration curve using analysis software SOFTmax PRO (Molecular Devices).
  • Pharmacokinetic parameters (AUC, whole body clearance (CL)) were calculated from the obtained plasma concentration transition data using pharmacokinetic analysis software WinNonlin (Pharsight) and shown in Table 6. Compared with H0L0, AUC after intravenous administration of Hp3Lp15 decreased by about 14% and clearance decreased by about 12%. Thus, it was found that Hp3Lp15 having a reduced isoelectric point of H0L0 has improved pharmacokinetics.
  • H chain the constant region was SKSC (SEQ ID NO: 62) prepared in Reference Examples 7 and 9, and the M58 (SEQ ID NO: 128) variable region was variable region Hp3 (Hp3-VH / SEQ ID NO: prepared in Example 7). 167) were combined to produce Hp3-M58 (SEQ ID NO: 240) and Hp3-SKSC (SEQ ID NO: 151). Hp3Lp15-SKSC (H chain Hp3-SKSC / SEQ ID NO: 151, L chain Lp15 / SEQ ID NO: 152) and Hp3Lp15 are combined with the prepared H chain and L chain Lp15 (Lp15 / SEQ ID NO: 152) prepared in Example 7. -M58 (H chain Hp3-M58 / SEQ ID NO: 240, L chain Lp15 / SEQ ID NO: 152) was prepared. Each antibody was expressed and purified by the method described in Example 4.
  • H0L0-SKSC H chain H0-SKSC / SEQ ID NO: 54, L chain L0 / SEQ ID NO: 56
  • H0L0-M58 H chain H0-M58 / SEQ ID NO: 136, L chain L0 / SEQ ID NO: 56
  • H0L0-IgG2 H chain H0-IgG2 / SEQ ID NO: 134, L chain L0 prepared in Example 5
  • biological activity using BaF / NR10 was carried out by the method described in Example 2, and the results are summarized in FIG.
  • H0L0 H chain H0-SKSC / SEQ ID NO: 54, L chain L0 / SEQ ID NO: 56 thermal acceleration product was prepared by the following method.
  • FIG. 19 shows the chromatogram results of the sample before and after the thermal acceleration of H0L0.
  • the basic peak In the sample after thermal acceleration of H0L0, there was a tendency for the basic peak to increase.
  • Ha355, Ha356, Ha360, Ha362 were found, and Ha355L0 (H chain Ha355-SKSC / SEQ ID NO: 242 L chain L0) in which these H chain variants and L0 were combined.
  • Ha356L0 H chain Ha356-SKSC / SEQ ID NO: 243, L chain L0 / SEQ ID NO: 56
  • Ha360L0 H chain Ha360-SKSC / SEQ ID NO: 244, L chain L0 / SEQ ID NO: 56
  • Ha362L0 H chain Ha362-SKSC / SEQ ID NO: 245, L chain L0 / SEQ ID NO: 56
  • each prepared antibody was prepared as a heat-accelerated product and analyzed by cation exchange chromatography. The results are shown in FIG.
  • the modification of Ha355, Ha356, Ha360, and Ha362 is effective in suppressing degradation products generated by thermal acceleration and enhancing the stability of the antibody.
  • Example 10 Identification of mutation sites that increase affinity In order to improve the affinity of H0L0 to NR10, a library in which mutations were introduced into CDR sequences was prepared and examined. As a result of screening libraries in which mutations were introduced into CDRs, mutations that improved the affinity for NR10 were found and are summarized in Table 8.
  • Respective heavy chain variants Ha101-SKSC SEQ ID NO: 246), Ha103-SKSC (SEQ ID NO: 247), Ha111-SKSC (SEQ ID NO: 248), Ha204-SKSC (SEQ ID NO: 249), Ha219-SKSC ( SEQ ID NO: 250) and L0 (L0 / SEQ ID NO: 56), the respective L chain variants La134 (SEQ ID NO: 251), La130 (SEQ ID NO: 252), La303 (SEQ ID NO: 253), La328 (SEQ ID NO: : 254) and H0 (H0-SKSC / SEQ ID NO: 54) were combined to prepare each antibody. Production and purification of each variant was carried out by the method described in Example 4.
  • Ha401La402 H chain Ha401-SKSC / SEQ ID NO: 255, L chain La402 / SEQ ID NO: 256
  • H17L11 H chain H17-M58 / SEQ ID NO: 222, L chain L11 / SEQ ID NO: 236
  • Affinity for NR10 of Ha401La402 H chain Ha401-SKSC / SEQ ID NO: 255, L chain La402 / SEQ ID NO: 256
  • biological activity using BaF / NR10 were carried out by the methods described in Reference Example 10 and Example 2. Comparison was made with H0L0 (H chain H0-SKSC / SEQ ID NO: 54, L chain L0 / SEQ ID NO: 56). The results of the affinity measured are shown in Table 10, and the biological activity using BaF / NR10 is shown in FIG. It was found that both affinity and biological activity were improved from that of H0L0 (H chain H0-SKSC / SEQ ID NO: 54, L chain L0 / SEQ ID NO: 56).
  • the affinity for NR10 of H17L11 (H chain H17-M58 / SEQ ID NO: 222, L chain L11 / SEQ ID NO: 236) and biological activity using BaF / NR10 were determined by the methods described in Example 7 and Example 2. And compared with H0L0 (H chain H0-M58 / SEQ ID NO: 136, L chain L0 / SEQ ID NO: 56). The measured affinity results are shown in Table 11, and the biological activity using BaF / NR10 is shown in FIG. It was found that both affinity and biological activity were improved from that of H0L0 (H chain H0-M58 / SEQ ID NO: 136, L chain L0 / SEQ ID NO: 56).
  • Example 11 Identification of mutation sites for reducing the risk of immunogenicity Reduction of immunogenicity risk of H chain CDR1
  • the T-cell epitope present in the variable region sequence of H0L0 was analyzed using TEPITOPE (Methods. 2004 Dec; 34 (4): 468-75). As a result, it was predicted that there are many T-cell epitopes that bind to HLA in the H chain CDR1 (a sequence with a high risk of immunogenicity exists). Therefore, we examined modifications that reduce the risk of immunogenicity of H chain CDR1 in TEPITOPE analysis.By replacing kabat numbering 33rd isoleucine (I) with alanine (A), the risk of immunogenicity is increased.
  • I isoleucine
  • A alanine
  • H19 H19-M58 / SEQ ID NO: 2283 was prepared by adding this modification to H17 prepared in Example 10. The prepared H19 was combined with L12 to prepare H19L12 (H chain H19-M58 / SEQ ID NO: 223, L chain L12 / SEQ ID NO: 237). Each variant was prepared and purified by the method described in Example 4.
  • Affinity to NR10 and biological activity using BaF / NR10 were carried out by the methods described in Reference Example 10 and Example 2, and H0L0 (H chain H0-M58 / SEQ ID NO: 136, L chain L0 / SEQ ID NO: 56) and Compared.
  • the results of the affinity measured are shown in Table 13, and the biological activity using BaF / NR10 is shown in FIG. Both affinity and biological activity were shown to be almost the same as that of H0L0.
  • Example 12 Preparation of fully humanized NS22 antibody Modifications that reduce the pI, modifications that increase affinity, modifications that suppress the degradation of the heavy chain, and modifications that reduce the risk of immunogenicity found in the above examples
  • Various variable regions of NS22 variants were created for H0 (H0-M58 / SEQ ID NO: 136), H1 (H1-M58 / SEQ ID NO: 257), or L0 (L0 / SEQ ID NO: 56).
  • H28L17 H chain H28-M58 / SEQ ID NO: 224, L chain L17 / SEQ ID NO: 238)
  • H30L17 H chain H30-M58 / SEQ ID NO: 225, L chain L17 / SEQ ID NO: 238)
  • H34L17 H chain H34-M58 / SEQ ID NO: 226, L chain L17 / SEQ ID NO: 238)
  • H42L17 H chain H42-M58 / SEQ ID NO: 227, L chain L17 / SEQ ID NO: 238)
  • H44L17 H chain H44-M58 / SEQ ID NO: 228, L chain L17 / SEQ ID NO: 238)
  • H46L17 H chain H46-M58 / SEQ ID NO: 229, L chain L17 / SEQ ID NO: 238)
  • H57L17 H chain H57-M58 / SEQ ID NO: 230, L chain L17 / SEQ ID NO:
  • Example 13 Analysis of binding domain of anti-NR10 neutralizing antibody (1) Preparation of human / mouse wild type and chimeric antigens Human and mouse wild type and chimeric NR10 extracellular region (hhh (SEQ ID NO: 258), mmm (SEQ ID NO: 259), hhm (SEQ ID NO: 260), mmh (SEQ ID NO: 261), hmm (SEQ ID NO: 262), mhm (SEQ ID NO: 263), mhh (SEQ ID NO: 264)) The gene was inserted into an animal cell expression vector with a His tag and Myc tag (HHHHHHEQKLISEEDL / SEQ ID NO: 287) at the C-terminus, and transiently expressed with the FreeStyle 293 Expression System (invitrogen TM ). A schematic diagram of these human / mouse wild type and chimeric NR10-ECD is shown in FIG.
  • Ni-NTA-Superflow column chromatography Purification of human / mouse wild type and chimeric antigens (hhh, mmm, hhm, mmh, hmm, mhm, mhh) from the culture supernatant was performed by Ni-NTA-Superflow column chromatography. Specifically, 1 mL of Ni-NTA-Superflow (QIAGEN) is packed into a polyprep empty column (BioRad), each 30 mL of culture supernatant is added, and D-PBS (Dulbecco's Phosphate containing 150 mM sodium chloride and 20 mM mM imidazole is added.
  • D-PBS Dulbecco's Phosphate containing 150 mM sodium chloride and 20 mM mM imidazole is added.
  • One (humanized anti-human NR10 antibody detection system) is 5 ⁇ g / mL H44M58L17
  • one (mouse anti-human NR10 antibody detection system) is 5 ⁇ g / mL ND41
  • one (Myc tag detection system) is The mixture was reacted with an anti-Myc antibody (SantaCruz, Cat. # Sc-789) diluted 500-fold with TBS containing 5% SkimMilk at room temperature for 1 hour.
  • Alkaline phosphatase-labeled goat anti-human IgG ⁇ (BIOSOURCE, Cat. # AHI0305) is used for the humanized anti-human NR10 antibody detection system, and alkaline phosphatase-labeled goat anti-mouse IgG (SantaCruz, Cat. # Sc) for the mouse anti-human NR10 antibody detection system.
  • alkaline phosphatase-labeled goat anti-rabbit IgG was used for the Myc tag detection system and reacted at room temperature for 1 hour.
  • TBS Tris-Buffered Saline
  • TBS Tris-Buffered-Saline
  • the cynomolgus monkey full-length NR10 and the cynomolgus OSMR gene were inserted into the expression vector pCOS1, respectively, and expressed in Ba / F3 cells to express the cynomolgus IL-31-dependent cell line ( cynNR10 / cynOSMR / BaF3 cells) were established.
  • amino acid sequence encoded by the nucleotide sequence of the intracellular region-deficient human NR10 gene is represented by SEQ ID NO: 74, and the nucleotide sequence of the intracellular region-deleted cynomolgus monkey NR10 gene (SEQ ID NO: 71).
  • the encoded amino acid sequence is shown in SEQ ID NO: 72.
  • DNA immunization was performed using a He gas injection gene gun, and a hybridoma was prepared by a conventional method.
  • the culture supernatant of these hybridomas was evaluated by neutralizing activity using the human IL-31-dependent cell line (hNR10 / hOSMR / BaF3 cells) shown in Reference Example 2 to obtain ND41 having NR10 neutralizing activity. It was.
  • the physical properties of the protein, especially homogeneity and stability, are extremely important, and it has been reported that the IgG2 isotype has a large amount of heterogeneity derived from disulfide bonds in the hinge region. (J Biol Chem. 2008 Jun 6; 283 (23): 16206-15.). It is not easy to manufacture a large amount as a pharmaceutical product while maintaining the heterogeneity of the target substance / related substance derived from this, and it is not easy and leads to an increase in cost. Therefore, in developing an IgG2 isotype antibody as a pharmaceutical, it is desirable that the heterogeneity derived from disulfide bonds is reduced without reducing stability.
  • SC SEQ ID NO: 153
  • CS SEQ ID NO: 153
  • each constant region prepared above, IgG1 (SEQ ID NO: 60), IgG2 (SEQ ID NO: 132), and variable region of humanized anti-IL-6 receptor antibody H chain variable region huPM1-VH / sequence
  • IgG1 SEQ ID NO: 60
  • IgG2 SEQ ID NO: 132
  • variable region of humanized anti-IL-6 receptor antibody H chain variable region huPM1-VH / sequence
  • No. 155 L chain variable region huPM1-VL / SEQ ID No. 156
  • HuPM1-SC (SEQ ID NO: 157), huPM1- CS (SEQ ID NO: 158), huPM1-IgG1 (SEQ ID NO: 159), huPM1-IgG2 (SEQ ID NO: 160) and huPM1-SKSC (SEQ ID NO: 161) are used, and huPM1-L (SEQ ID NO: : 162) to prepare each antibody.
  • Each antibody was expressed and purified by the method described in Example 4.
  • the heterogeneity of each antibody was compared.
  • evaluation by cation exchange chromatography was performed.
  • ProPac WCX-10 (Dionex) is used as the column, 20 mM Sodium Acetate, pH5.0 as mobile phase A, 20 ⁇ mM Sodium Acetate, 1 M NaCl, pH5.0 as mobile phase B, This was carried out using a gradient.
  • the results of evaluation by cation exchange chromatography are shown in FIG.
  • the heterogeneity increased by converting the constant region from IgG1 to IgG2, but the heterogeneity was greatly reduced by converting the constant region to SKSC.
  • the constant region was SC
  • the heterogeneity was greatly reduced as in the case where the constant region was SKSC.
  • the constant region was CS, the heterogeneity was not sufficiently improved.
  • Tm value thermal denaturation intermediate temperature
  • DSC differential scanning calorimetry
  • huPM1-IgG1, huPM1-IgG2, huPM1-SC, huPM1-CS, and huPM1-SKSC were dialyzed (EasySEP, TOMY) against a solution of 20 msodium acetate, 150 mM NaCl, pH6.0, and about 0.1 DSC measurement was performed at a temperature increase rate of 1 ° C./min from 40 ° C. to 100 ° C. at a protein concentration of mg / mL.
  • the resulting DSC denaturation curve is shown in FIG. 13 and the Tm value of the Fab portion is shown in Table 17 below.
  • the Tm values of huPM1-IgG1 and huPM1-IgG2 were about the same, about 94 ° C (IgG2 was about 1 ° C lower), whereas the Tm values of huPM1-SC and huPM1-CS were about 86 ° C. Yes, the Tm value was significantly reduced compared to huPM1-IgG1 and huPM1-IgG2. On the other hand, the Tm value of huPM1-SKSC was about 94 ° C., which was almost equivalent to huPM1-IgG1 and huPM1-IgG2.
  • huPM1-SC and huPM1-CS are significantly less stable than IgG2, it was considered that huPM1-SKSC in which the cysteine of the CH1 domain was also changed to serine was preferable for development as a pharmaceutical product.
  • the reason why the Tm values of huPM1-SC and huPM1-CS were significantly reduced compared to IgG2 was considered to be because huPM1-SC and huPM1-CS took a different form from the disulfide bond pattern of IgG2.
  • the denaturation peaks of the Fab portion of huPM1-IgG1 and huPM1-SKSC were sharp, whereas huPM1-SC and huPM1-CS were compared with these,
  • the denaturation peak was broad, and huPM1-IgG2 had a shoulder peak on the low temperature side of the denaturation peak of the Fab portion.
  • the DSC denaturation peak usually shows a sharp denaturation peak in the case of a single component, but it is considered that the denaturation peak becomes broad when there are multiple components having different Tm (that is, heterogeneity).
  • huPM1-IgG2, huPM1-SC, and huPM1-CS have a plurality of components, suggesting that huPM1-SC and huPM1-CS may not have sufficiently reduced the heterogeneity of natural IgG2.
  • the natural IgG2 heterogeneity involves not only the cysteine in the hinge part but also the cysteine present in the CH1 domain.
  • the hinge It was considered necessary to modify not only the partial cysteine but also the cysteine of the CH1 domain.
  • SC and CS which are constant regions in which only the cysteine in the hinge portion is replaced with serine, are insufficient in terms of heterogeneity and stability. It is considered that the heterogeneity can be significantly reduced while maintaining the same stability as IgG2 for the first time by replacing the cysteine at the 131st EU numbering in the CH1 domain with serine. It was issued.
  • An example of such a constant region was SKSC.
  • IgG2 EU numbering 327, 330, 331 amino acids are naturally changed to IgG4 sequence. A new peptide sequence of 9 amino acids that can be a non-existing T-cell epitope peptide appears, resulting in the risk of immunogenicity.
  • IgG2 EU numbering it is possible to prevent the appearance of a new peptide sequence by modifying the 339th threonine to alanine.
  • a mutation was introduced from the 397th methionine to valine in the EU numbering of IgG2, which improves the stability of IgG2 under acidic conditions.
  • SKSC SEQ ID NO: 62
  • SKSC that improves the heterogeneity derived from the disulfide bond in the hinge region prepared in Reference Example 7 is a T-cell epitope peptide that does not exist in nature with the 131st and 133rd mutations. Since a new peptide sequence of 9 amino acids that could become an immunological risk arises, by introducing the EU numbering mutation 137th glutamic acid to glycine and the 138th serine to glycine mutation, The peptide sequence from the 131st to the 139th region was the same as IgG1. A constant region sequence M14 (SEQ ID NO: 129) into which all of these mutations were introduced was prepared.
  • the main component is a sequence in which the lysine of the C-terminal amino acid present on the base sequence is deleted by post-translational modification, but the C-terminal amino group due to the loss of both glycine and lysine as a minor component remaining in lysine
  • the amidated subcomponent of is also present as a heterogeneity. It is not easy to manufacture a large amount as a pharmaceutical product while maintaining the heterogeneity of the target substance / related substance, leading to increased costs, and it is desirable that it be a single substance as much as possible. For development, it is desirable that these heterogeneities are reduced. Therefore, it is desirable that there is no H chain C-terminal heterogeneity when developing as a pharmaceutical product. In order to reduce the dose of the antibody, it is desirable to increase the plasma half-life of the antibody.
  • EU numbering 137th glutamic acid and 138th serine glycine against huPM1-SKSC with high stability and reduced heterogeneity related to IgG2 isotype constant region antibody.
  • 268th histidine is replaced with glutamine
  • 355th arginine is replaced with glutamine
  • 419th glutamine is replaced with glutamic acid
  • 446th glycine and HuPM1-M58 was found.
  • Expression and purification of huPM1-M58 using huPM1-M58 as the H chain and huPM1-L (SEQ ID NO: 162) as the L chain were carried out by the method described in Example 4.
  • Table 18 shows the DSC results. Further, as shown in FIGS. 13 and 16, also in huPM1-M58, similar to huPM1-SKSC, heterogeneity is reduced without impairing stability.
  • IgG1, IgG2, IgG3, and IgG4 isotypes are known as IgG type antibodies, but the plasma half-life in these humans is about 36 days for IgG1, IgG2, about 29 days for IgG3, and 16 for IgG4. (Nat ⁇ Biotechnol. 2007 Dec; 25 (12): 1369-72.), And IgG1 and IgG2 are considered to have the longest retention in plasma.
  • antibody drug isotypes are IgG1, IgG2, and IgG4.
  • the binding to the above-mentioned human FcRn is improved by modifying the sequence of the constant region of IgG. (J Biol Chem. 2007 Jan 19; 282 (3): 1709-17, J Immunol. 2006 Jan 1; 176 (1): 346-56).
  • FcRn is a complex of FcRn and ⁇ 2-microglobulin. Oligo DNA primers were prepared based on the published human FcRn gene sequence (J. Exp. Med. 180 (6), 2377-2381 (1994)). A human cDNA (Human Placenta Marathon-Ready cDNA, Clontech) was used as a template, and a DNA fragment encoding the entire gene length was prepared by PCR using the prepared primer.
  • a DNA fragment encoding an extracellular region (Met1-Leu290) including a signal region was amplified by PCR and inserted into an animal cell expression vector (human FcRn amino acid sequence / SEQ ID NO: 165). ).
  • oligo DNA primers were prepared based on the published human ⁇ 2-microglobulin gene sequence (Proc. Natl. Acad. Sci. USA 99 (26), 16899-16903 (2002)).
  • a human cDNA Human-Placenta Marathon-Ready cDNA, CLONTECH
  • a DNA fragment encoding the full length of the gene was prepared by PCR using the prepared primer.
  • a DNA fragment encoding ⁇ 2-microglobulin full length (Met1-Met119) including the signal region was amplified by PCR and inserted into an animal cell expression vector (human ⁇ 2-microglobulin amino acid sequence / sequence) Number: 166).
  • Soluble human FcRn is expressed in human fetal kidney cancer cell-derived HEK293H strain (Invitrogen) using 10% Fetal Bovine Serum (Invitrogen), and the prepared human FcRn and human ⁇ 2-microglobulin plasmids are introduced into the cells by the lipofection method. did. After collecting the obtained culture supernatant, purification was performed using IgG ⁇ Sepharose 6 Fast Flow (Amersham Biosciences) according to the method of (J Immunol. 2002 Nov 1; 169 (9): 5171-80.). Thereafter, purification was performed using HiTrap Q HP (GE Healthcare).
  • Biacore® 3000 was used for evaluation of binding to human FcRn, and human FcRn was interacted with human FcRn as an analyte to an antibody bound to Protein® L or rabbit anti-human IgG® Kappa® chain antibody immobilized on the sensor chip.
  • the affinity (KD) was calculated from the amount of binding.
  • Protein L was immobilized on sensor chip CM5 (BIACORE) by amine coupling method using 50 mM Na-phosphate buffer, pH 6.0 containing 150 m ⁇ ⁇ ⁇ ⁇ NaCl as a running buffer. Thereafter, the antibody was diluted with a running buffer containing 0.02% Tween 20 and injected to bind the antibody to the chip. Then, human FcRn was injected and the binding of the antibody to human FcRn was evaluated.
  • association rate constant k a (1 / Ms) and dissociation rate constant k d (1 / s), which are kinetic parameters, are calculated from the sensorgram obtained by the measurement, and K D (M ) was calculated.
  • K D (M ) was calculated.
  • Each parameter was calculated using Biacore T100 Evaluation Software version 1.1 (GE Healthcare Bioscience).
  • Protein A immobilized on the sensor chip Protein A was immobilized on all flow cells of the sensor chip CM5 (GE Healthcare Bioscience) by the amine coupling method.
  • HBS-EP + (10 mM HEPES, 0.15 M NaCl, 3 mM EDTA, 0.05% v / v Surfactant P20) was used as a running buffer, and experiments were performed at a flow rate of 10 ⁇ L / min.
  • the carboxyl group of carboxymethyldextran on the sensor chip is 100 ⁇ L of a 1: 1 mixture of 75 mg / mL EDC (N-ethyl-N '-(3-dimethylaminopropyl) carbodiimide hydrochloride) and 11.5 mg / mL NHS (N-hydroxysuccinimide). Then, Protein A prepared at 50 ⁇ g / mL with 10 mM acetate buffer (pH 4.5) was allowed to flow therethrough for reaction. Thereafter, 100 ⁇ L of 1 M ethanolamine hydrochloride (pH 8.5) was flowed to inactivate unreacted active groups. Finally, about 4000-5000 RU was immobilized. All experiments were performed at 25 ° C.
  • HBS-EP + was used as an affinity measurement running buffer for antigen-antibody reaction between antibody captured by Protein A and rhNR10 .
  • Each antibody was prepared to bind to 0.25 ⁇ g / mL or about 100 RU of Protein A.
  • RhNR10 used as the analyte was prepared to 0, 38.5, 77.0, 154 nM, or 0, 19.25, 77.01 nM using HBS-EP +.
  • the antibody solution was first captured by Protein A, and the analyte solution was reacted for 3 min at a flow rate of 20 ⁇ L / min, then switched to HBS-EP + and the 5 min dissociation phase was measured.
  • the sensor chip was regenerated by washing with 10 mM glycine-HCl (pH 1.5). From the obtained sensorgram, kinetic analysis was performed using Biacore T100 Evaluation Software Version 1.1, which is Biacore's dedicated data analysis software.
  • the anti-NR10 antibody obtained by the present inventor exhibits an effective neutralizing activity against NR10, and is useful, for example, as a therapeutic agent for inflammatory diseases.

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Immunology (AREA)
  • Medicinal Chemistry (AREA)
  • Molecular Biology (AREA)
  • General Health & Medical Sciences (AREA)
  • Organic Chemistry (AREA)
  • Engineering & Computer Science (AREA)
  • Biochemistry (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Biomedical Technology (AREA)
  • Microbiology (AREA)
  • Urology & Nephrology (AREA)
  • Hematology (AREA)
  • Cell Biology (AREA)
  • Animal Behavior & Ethology (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Biophysics (AREA)
  • Public Health (AREA)
  • Veterinary Medicine (AREA)
  • Genetics & Genomics (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Pathology (AREA)
  • Mycology (AREA)
  • Epidemiology (AREA)
  • General Physics & Mathematics (AREA)
  • Analytical Chemistry (AREA)
  • Biotechnology (AREA)
  • Physics & Mathematics (AREA)
  • Food Science & Technology (AREA)
  • Rheumatology (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Pain & Pain Management (AREA)
  • General Chemical & Material Sciences (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Peptides Or Proteins (AREA)
  • Medicines Containing Antibodies Or Antigens For Use As Internal Diagnostic Agents (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)

Abstract

La présente invention concerne l'obtention réussie d'anticorps anti-NR10 présentant une activité de neutralisation effective sur NR10. Les anticorps anti-NR10 ainsi obtenus conviennent comme médicaments pour le traitement ou la prévention notamment de maladies inflammatoires.
PCT/JP2009/054941 2007-12-05 2009-03-13 Anticorps anti-nr10, et leurs utilisations WO2010064456A1 (fr)

Priority Applications (24)

Application Number Priority Date Filing Date Title
JP2009054941A JP2010210772A (ja) 2009-03-13 2009-03-09 液晶表示装置の製造方法
TW098141125A TWI432209B (zh) 2008-12-05 2009-12-02 Anti-NR10 antibody and its use
JP2010507563A JP5139517B2 (ja) 2008-12-05 2009-12-04 抗nr10抗体、およびその利用
CA2710264A CA2710264C (fr) 2007-12-05 2009-12-04 Anticorps anti-nr10, et utilisation correspondante
AU2009323249A AU2009323249B2 (en) 2007-12-05 2009-12-04 Anti-NR10 antibody, and use thereof
BRPI0906478A BRPI0906478B8 (pt) 2008-12-05 2009-12-04 anticorpo anti-nr10, seu uso e composição farmacêutica que o compreende
EP15172595.9A EP2949672A1 (fr) 2008-12-05 2009-12-04 Anticorps anti-nr10, et utilisation correspondante
US12/809,138 US8575317B2 (en) 2007-12-05 2009-12-04 Anti-NR10 antibody and use thereof
PL09830463T PL2354161T3 (pl) 2008-12-05 2009-12-04 Przeciwciało anty -NR10 i jego zastosowanie
SI200931302T SI2354161T1 (sl) 2008-12-05 2009-12-04 Anti-NR10 protitelo in njegova uporaba
CN200980106176.2A CN101952318B (zh) 2007-12-05 2009-12-04 抗nr10抗体及其应用
EP09830463.7A EP2354161B1 (fr) 2008-12-05 2009-12-04 Anticorps anti-nr10, et utilisation correspondante
MX2010007936A MX2010007936A (es) 2007-12-05 2009-12-04 Anticuerpo anti-nr-10 y su uso.
PT98304637T PT2354161E (pt) 2008-12-05 2009-12-04 Anticorpo anti-nr10, e a sua utilização
KR1020107014700A KR101271323B1 (ko) 2008-12-05 2009-12-04 항nr10 항체 및 그의 이용
RU2010126078/10A RU2487136C2 (ru) 2008-12-05 2009-12-04 Антитело против nr10 и его применение
HUE09830463A HUE025958T2 (en) 2007-12-05 2009-12-04 Anti-NR10 antibody and application
ES09830463.7T ES2552690T3 (es) 2008-12-05 2009-12-04 Anticuerpo anti-NR10 y uso del mismo
MYPI2010002989A MY154715A (en) 2008-12-05 2009-12-04 Anti-nr10 antibody and use thereof
DK09830463.7T DK2354161T3 (en) 2008-12-05 2009-12-04 ANTI-NR10 ANTIBODY AND USE THEREOF.
PCT/JP2009/070376 WO2010064697A1 (fr) 2008-12-05 2009-12-04 Anticorps anti-nr10, et utilisation correspondante
HK11105170.8A HK1151048A1 (en) 2008-12-05 2011-05-24 Anti-nr 10 antibody, and use thereof nr10
US14/047,316 US9399680B2 (en) 2007-12-05 2013-10-07 Nucleic acids encoding anti-NR10 antibodies
HRP20151215TT HRP20151215T1 (hr) 2007-12-05 2015-11-12 Anti-nr10 protutijela i njihova uporaba

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
JPPCT/JP2008/072152 2008-12-05
PCT/JP2008/072152 WO2009072604A1 (fr) 2007-12-05 2008-12-05 Anticorps anti-nr10 et son utilisation

Publications (1)

Publication Number Publication Date
WO2010064456A1 true WO2010064456A1 (fr) 2010-06-10

Family

ID=42234508

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/JP2009/054941 WO2010064456A1 (fr) 2007-12-05 2009-03-13 Anticorps anti-nr10, et leurs utilisations

Country Status (5)

Country Link
KR (1) KR20160062207A (fr)
MY (1) MY154715A (fr)
SI (1) SI2354161T1 (fr)
TW (1) TWI432209B (fr)
WO (1) WO2010064456A1 (fr)

Cited By (8)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
JP5697119B1 (ja) * 2013-06-28 2015-04-08 中外製薬株式会社 そう痒を伴う疾患に罹患した患者のil−31アンタゴニストによる治療に対する応答を予測する方法
US9688762B2 (en) 2007-09-26 2017-06-27 Chugai Sciyaku Kabushiki Kaisha Modified antibody constant region
US9828429B2 (en) 2007-09-26 2017-11-28 Chugai Seiyaku Kabushiki Kaisha Method of modifying isoelectric point of antibody via amino acid substitution in CDR
US10066018B2 (en) 2009-03-19 2018-09-04 Chugai Seiyaku Kabushiki Kaisha Antibody constant region variant
US10253091B2 (en) 2009-03-19 2019-04-09 Chugai Seiyaku Kabushiki Kaisha Antibody constant region variant
US10544227B2 (en) 2015-04-14 2020-01-28 Chugai Seiyaku Kabushiki Kaisha Pharmaceutical composition for prevention and/or treatment of atopic dermatitis comprising IL-31 antagonist as active ingredient
US11046784B2 (en) 2006-03-31 2021-06-29 Chugai Seiyaku Kabushiki Kaisha Methods for controlling blood pharmacokinetics of antibodies
US11260125B2 (en) 2019-11-20 2022-03-01 Chugai Seiyaku Kabushiki Kaisha Anti-IL31RA antibody-containing formulations

Citations (17)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1994012215A1 (fr) * 1992-12-01 1994-06-09 Protein Design Labs, Inc. Anticorps humanises reagissant avec l-selectine
WO1996023071A2 (fr) * 1995-01-26 1996-08-01 Bristol-Myers Squibb Company ANTICORPS MONOCLONAUX SPECIFIQUES DE DIFFERENTS EPITOPES DE gp39 HUMAINE ET PROCEDES CONCERNANT LEUR UTILISATION A DES FINS DE DIAGNOSTIC ET DE THERAPIE
WO1997010354A1 (fr) * 1995-09-11 1997-03-20 Kyowa Hakko Kogyo Co., Ltd. Anticorps de la chaine alpha du recepteur de l'interleukine 5 humaine
WO2000075314A1 (fr) * 1999-06-02 2000-12-14 Chugai Research Institute For Molecular Medicine, Inc. Proteine receptrice d'hemopoietine
WO2004091543A2 (fr) * 2003-03-04 2004-10-28 Alexion Pharmaceuticals, Inc. Procede de traitement de maladies auto-immunes en suscitant une presentation d'antigenes par des cellules presentant des antigenes inductrices de tolerance
US20040223970A1 (en) * 2003-02-28 2004-11-11 Daniel Afar Antibodies against SLC15A2 and uses thereof
WO2006070286A2 (fr) * 2004-12-28 2006-07-06 Innate Pharma S.A. Anticorps monoclonaux contre le nkg2a
WO2006088855A1 (fr) * 2005-02-14 2006-08-24 Zymogenetics, Inc. Méthodes de traitement d’affections de la peau en employant un antagoniste de il-31ra
WO2006119062A2 (fr) * 2005-05-03 2006-11-09 Ucb Pharma S.A. Epitopes
WO2006118959A2 (fr) * 2005-04-29 2006-11-09 Rinat Neuroscience Corp. Anticorps diriges contre un peptide amyloide-beta et procedes d'utilisation de ceux-ci
WO2007142325A1 (fr) * 2006-06-08 2007-12-13 Chugai Seiyaku Kabushiki Kaisha Prévention ou traitement d'une maladie inflammatoire
US20080125579A1 (en) * 2006-06-15 2008-05-29 Board Of Trustees Of The University Of Arkansas Monoclonal antibodies that selectively recognize methamphetamine and methamphetamine like compounds
WO2008103432A1 (fr) * 2007-02-23 2008-08-28 Schering Corporation Anticorps obtenus par génie génétique dirigés contre l'il-23p19
US20080219971A1 (en) * 2007-02-14 2008-09-11 Vaccinex, Inc. Human anti-cd100 antibodies
WO2008132453A1 (fr) * 2007-04-25 2008-11-06 Medella Therapeutics Limited Anticorps
WO2009041621A1 (fr) * 2007-09-26 2009-04-02 Chugai Seiyaku Kabushiki Kaisha Anticorps du récepteur anti-il-6
WO2009041643A1 (fr) * 2007-09-26 2009-04-02 Chugai Seiyaku Kabushiki Kaisha Procédé permettant de modifier le point isoélectrique d'un anticorps par substitution d'acide aminé dans une région déterminant la complémentarité (cdr)

Patent Citations (17)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1994012215A1 (fr) * 1992-12-01 1994-06-09 Protein Design Labs, Inc. Anticorps humanises reagissant avec l-selectine
WO1996023071A2 (fr) * 1995-01-26 1996-08-01 Bristol-Myers Squibb Company ANTICORPS MONOCLONAUX SPECIFIQUES DE DIFFERENTS EPITOPES DE gp39 HUMAINE ET PROCEDES CONCERNANT LEUR UTILISATION A DES FINS DE DIAGNOSTIC ET DE THERAPIE
WO1997010354A1 (fr) * 1995-09-11 1997-03-20 Kyowa Hakko Kogyo Co., Ltd. Anticorps de la chaine alpha du recepteur de l'interleukine 5 humaine
WO2000075314A1 (fr) * 1999-06-02 2000-12-14 Chugai Research Institute For Molecular Medicine, Inc. Proteine receptrice d'hemopoietine
US20040223970A1 (en) * 2003-02-28 2004-11-11 Daniel Afar Antibodies against SLC15A2 and uses thereof
WO2004091543A2 (fr) * 2003-03-04 2004-10-28 Alexion Pharmaceuticals, Inc. Procede de traitement de maladies auto-immunes en suscitant une presentation d'antigenes par des cellules presentant des antigenes inductrices de tolerance
WO2006070286A2 (fr) * 2004-12-28 2006-07-06 Innate Pharma S.A. Anticorps monoclonaux contre le nkg2a
WO2006088855A1 (fr) * 2005-02-14 2006-08-24 Zymogenetics, Inc. Méthodes de traitement d’affections de la peau en employant un antagoniste de il-31ra
WO2006118959A2 (fr) * 2005-04-29 2006-11-09 Rinat Neuroscience Corp. Anticorps diriges contre un peptide amyloide-beta et procedes d'utilisation de ceux-ci
WO2006119062A2 (fr) * 2005-05-03 2006-11-09 Ucb Pharma S.A. Epitopes
WO2007142325A1 (fr) * 2006-06-08 2007-12-13 Chugai Seiyaku Kabushiki Kaisha Prévention ou traitement d'une maladie inflammatoire
US20080125579A1 (en) * 2006-06-15 2008-05-29 Board Of Trustees Of The University Of Arkansas Monoclonal antibodies that selectively recognize methamphetamine and methamphetamine like compounds
US20080219971A1 (en) * 2007-02-14 2008-09-11 Vaccinex, Inc. Human anti-cd100 antibodies
WO2008103432A1 (fr) * 2007-02-23 2008-08-28 Schering Corporation Anticorps obtenus par génie génétique dirigés contre l'il-23p19
WO2008132453A1 (fr) * 2007-04-25 2008-11-06 Medella Therapeutics Limited Anticorps
WO2009041621A1 (fr) * 2007-09-26 2009-04-02 Chugai Seiyaku Kabushiki Kaisha Anticorps du récepteur anti-il-6
WO2009041643A1 (fr) * 2007-09-26 2009-04-02 Chugai Seiyaku Kabushiki Kaisha Procédé permettant de modifier le point isoélectrique d'un anticorps par substitution d'acide aminé dans une région déterminant la complémentarité (cdr)

Non-Patent Citations (7)

* Cited by examiner, † Cited by third party
Title
CHIRINO J. ARTHUR ET AL.: "Minimizing the immnugenicity of protein therapeutics", DRUG DISCOVERY TODAY, vol. 9, no. 2, 2004, pages 82 - 90 *
DILLON R. STACEY ET AL.: "Interleukin 31, a cytokine produced by activated T cells, induces dermatitis in mice", NATURE IMMUNOLOGY, vol. 5, no. 7, 2004, pages 752 - 760 *
DIVEU CAROLINE ET AL.: "GPL, a Novel Cytokine Receptor Related to GP130 and Leukemia Inhibitory Factor Receptor", J. BIOL. CHEM., vol. 278, no. 50, 2003, pages 49850 - 49859 *
EWERT STEFAN ET AL.: "Stability improvement of antibodies for extracellular and intracellular applications: CDR grafting to stable frameworks and structure-based framework engineering", METHODS, vol. 34, 2004, pages 184 - 199 *
HANES JOZEF ET AL.: "Picomolar affinity antibodies from a fully synthetic naive library selected and evolved by ribosome display", NATURE BIOTECHNOLOGY, vol. 18, 2000, pages 1287 - 1292 *
TAN H. PHILIP ET AL.: "Engineering the isoelectric point of a renal cell carcinoma targeting antibody greatly enhances scFv solubility", IMMUNOTECHNOLOGY, vol. 4, 1998, pages 107 - 114 *
YAGI YUHKI ET AL.: "Interleukin-31 stimulates production of inflammatory mediators from human colonic subepithelial myofibroblasts", INT. J. MOL. MED., vol. 19, 2007, pages 941 - 946 *

Cited By (12)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US11046784B2 (en) 2006-03-31 2021-06-29 Chugai Seiyaku Kabushiki Kaisha Methods for controlling blood pharmacokinetics of antibodies
US9688762B2 (en) 2007-09-26 2017-06-27 Chugai Sciyaku Kabushiki Kaisha Modified antibody constant region
US9828429B2 (en) 2007-09-26 2017-11-28 Chugai Seiyaku Kabushiki Kaisha Method of modifying isoelectric point of antibody via amino acid substitution in CDR
US11248053B2 (en) 2007-09-26 2022-02-15 Chugai Seiyaku Kabushiki Kaisha Method of modifying isoelectric point of antibody via amino acid substitution in CDR
US11332533B2 (en) 2007-09-26 2022-05-17 Chugai Seiyaku Kabushiki Kaisha Modified antibody constant region
US10066018B2 (en) 2009-03-19 2018-09-04 Chugai Seiyaku Kabushiki Kaisha Antibody constant region variant
US10253091B2 (en) 2009-03-19 2019-04-09 Chugai Seiyaku Kabushiki Kaisha Antibody constant region variant
JP5697119B1 (ja) * 2013-06-28 2015-04-08 中外製薬株式会社 そう痒を伴う疾患に罹患した患者のil−31アンタゴニストによる治療に対する応答を予測する方法
US10544227B2 (en) 2015-04-14 2020-01-28 Chugai Seiyaku Kabushiki Kaisha Pharmaceutical composition for prevention and/or treatment of atopic dermatitis comprising IL-31 antagonist as active ingredient
US11773173B2 (en) 2015-04-14 2023-10-03 Chugai Seiyaku Kabushiki Kaisha Pharmaceutical composition for prevention and/or treatment of atopic dermatitis comprising IL-31 antagonist as active ingredient
US11260125B2 (en) 2019-11-20 2022-03-01 Chugai Seiyaku Kabushiki Kaisha Anti-IL31RA antibody-containing formulations
US11723976B2 (en) 2019-11-20 2023-08-15 Chugai Seiyaku Kabushiki Kaisha Methods of administering anti-IL31A antibody-containing formulations

Also Published As

Publication number Publication date
SI2354161T1 (sl) 2015-12-31
TWI432209B (zh) 2014-04-01
MY154715A (en) 2015-07-10
TW201028165A (en) 2010-08-01
KR20160062207A (ko) 2016-06-01

Similar Documents

Publication Publication Date Title
US20230183363A1 (en) Anti-nr10 antibody and use thereof
WO2010064697A1 (fr) Anticorps anti-nr10, et utilisation correspondante
KR101557603B1 (ko) 염증성 질환의 예방 또는 치료제
US8431127B2 (en) Method for treating pruritus comprising administering an NR10 antagonist
WO2010064456A1 (fr) Anticorps anti-nr10, et leurs utilisations
JP5139517B2 (ja) 抗nr10抗体、およびその利用
JP2011219380A (ja) 抗vegf−d抗体、およびその利用

Legal Events

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

Ref document number: 09830226

Country of ref document: EP

Kind code of ref document: A1

NENP Non-entry into the national phase

Ref country code: DE

122 Ep: pct application non-entry in european phase

Ref document number: 09830226

Country of ref document: EP

Kind code of ref document: A1

NENP Non-entry into the national phase

Ref country code: JP