WO2020009165A1 - Anticorps modifié et procédé pour sa production - Google Patents

Anticorps modifié et procédé pour sa production Download PDF

Info

Publication number
WO2020009165A1
WO2020009165A1 PCT/JP2019/026521 JP2019026521W WO2020009165A1 WO 2020009165 A1 WO2020009165 A1 WO 2020009165A1 JP 2019026521 W JP2019026521 W JP 2019026521W WO 2020009165 A1 WO2020009165 A1 WO 2020009165A1
Authority
WO
WIPO (PCT)
Prior art keywords
residue
antibody
modified
lysine residue
side chain
Prior art date
Application number
PCT/JP2019/026521
Other languages
English (en)
Japanese (ja)
Inventor
山崎 俊介
奈都紀 敷田
和高 新保
松田 豊
公太 井上
康博 三原
セルゲイ ヴァシリエヴィッチ スミルノフ
イリーナ リヴォヴナ トクマコーヴァ
Original Assignee
味の素株式会社
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
Application filed by 味の素株式会社 filed Critical 味の素株式会社
Publication of WO2020009165A1 publication Critical patent/WO2020009165A1/fr

Links

Images

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K19/00Hybrid peptides, i.e. peptides covalently bound to nucleic acids, or non-covalently bound protein-protein complexes
    • 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
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K45/00Medicinal preparations containing active ingredients not provided for in groups A61K31/00 - A61K41/00
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/50Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates
    • A61K47/51Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent
    • A61K47/68Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an antibody, an immunoglobulin or a fragment thereof, e.g. an Fc-fragment
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K49/00Preparations for testing in vivo
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N9/00Enzymes; Proenzymes; Compositions thereof; Processes for preparing, activating, inhibiting, separating or purifying enzymes

Definitions

  • the present invention relates to a modified antibody and a method for producing the same.
  • ADC antibody-drug conjugates
  • ADC is a drug obtained by conjugating a drug (eg, an anticancer agent) to an antibody, and has a direct cell killing activity against cancer cells and the like.
  • a typical ADC is T-DM1 (trade name: Kadcyla (registered trademark)) jointly developed by Immunogene and Roche. Therefore, there is a need to develop a technique that is useful for modifying antibodies with drugs.
  • antibodies are a type of protein. Modification of a protein is useful for modulating the function of the desired protein. As a protein modification, modification using functions of various naturally occurring enzymes has been attempted. For example, phosphopantetheinyl transferase (PPTase), biotin ligase, and lipoic acid protein ligase (Lpl) are known as enzymes capable of modifying proteins.
  • PPTase phosphopantetheinyl transferase
  • biotin ligase biotin ligase
  • Lpl lipoic acid protein ligase
  • PPTase is an enzyme that forms a phosphate bond between a hydroxyl group of a serine residue in a protein and coenzyme A (CoA).
  • H / D S
  • L / I has been reported as a PPTase recognition amino acid sequence, and PPTase is considered to recognize an ⁇ -helix structure around a serine residue.
  • Non-patent Document 1 Since PPTase is an enzyme widely conserved in prokaryotes, it is considered to be suitable for searching for a target protein modifying enzyme.
  • PPTase has a short recognition amino acid sequence consisting of three amino acid residues, and is not strict in recognition of both N-terminal and C-terminal amino acid residues in the recognition amino acid sequence. It is thought that it is not high.
  • Biotin ligase is an enzyme that covalently binds biotin to lysine residues in proteins. Biotin has a very high affinity for avidin (Kd ⁇ 10 ⁇ 15 M), so that biotin ligase can add various drugs to lysine residues of proteins via avidin.
  • E. coli a kind of biotin ligase. It has been reported that E. coli BirA can recognize an amino acid sequence consisting of 15 amino acid residues represented by GLNDIFEAQ [K] IEWHE ([K] indicates a lysine residue to which biotin is added).
  • FIG. A fusion protein of an acceptor peptide having an E. coli BirA-recognizing amino acid sequence with a target protein is prepared. A technique for preparing an avidin-modified fusion protein by treating with E. coli BirA has been reported (Non-Patent Document 2).
  • Lpl is an enzyme that binds lipoic acid to a lysine residue in a protein for acylation.
  • Escherichia coli Escherichia coli LplA exists.
  • Escherichia coli (Escherichia coli) LplA is known to function as a coenzyme for enzymes such as pyruvate dehydrogenase, ⁇ -ketoglutarate dehydrogenase, and glycine cleavage system enzymes (Non-patent Documents 3 and 4).
  • LplA recognizes the amino acid sequence of E2 protein (E2p), which is one of the subunits of these enzymes, and activates E2p by adding lipoic acid (lipoylation) to lysine residues in the recognition sequence. As shown in Table 1, LplA is reported to have a relatively long recognition amino acid sequence consisting of 12 amino acid residues (Non-Patent Document 5).
  • N-terminal amino acid residue and the C-terminal amino acid residue of lysylated lysine residues are aspartic acid residue (D) and It is restricted to amino acid residues having a hydrocarbon group as a side chain [alanine (A), valine (V), or leucine (L)] (Table 1).
  • Patent Document 1 As a modification of a protein with LplA, a method of modifying a target protein with lipoic acid and a lipoic acid analog via the LplA recognition peptide tag in a target protein to which an LplA recognition peptide tag has been added is known (Patent Document 1). . Further, as a modification of a protein (antibody) by LplA, a method of modifying a Fab fragment with lipoic acid via an LplA recognition polypeptide in a Fab fragment to which an LplA recognition polypeptide has been added has been reported (Patent Document 2). .
  • the purpose of the present invention is to develop a technology that allows modification of an antibody.
  • the present inventors have conducted intensive studies on antibody modification by an enzymatic method. As a result, among the tested protein-modifying enzymes, phosphopantetheinyltransferase (PPTase) which has the shortest recognized amino acid sequence and is expected to have low substrate specificity ) And biotin ligase did not have the ability to modify an antibody, but lipoic acid protein ligase (Lpl) could modify an antibody despite having no known Lpl-recognizing amino acid sequence. Was found. Next, the present inventors analyzed the modification site of the antibody with Lpl.
  • PPTase phosphopantetheinyltransferase
  • Lpl lipoic acid protein ligase
  • the present invention is as follows. [1] including reacting the antibody with a lipoic acid analog having a modifying moiety in the presence of lipoic acid protein ligase to produce a modified antibody having a modifying moiety on the side chain of a lysine residue in the constant region. , A method for producing a modified antibody having a modified moiety. [2] The method of [1], wherein the antibody has a natural polypeptide chain structure. [3] The method of [1] or [2], wherein the antibody is a monoclonal antibody.
  • the lipoic acid analog having a modified moiety is a C 4 -C 10 alkyl-carboxylic acid having a modified moiety;
  • a modified antibody having a modification portion in the side chain of a lysine residue in the constant region is an antibody having a C 4 to C 10 alkyl-carbonyl having a modification portion in the side chain of a lysine residue in the constant region, [ Any one of 1) to [7].
  • the lipoic acid analog having a modified moiety is represented by the following formula (I): R—C n H 2n —COOH (I) (In the formula, R is a modifying moiety; n is an integer of 4 to 10.
  • a modified antibody having a modified portion on the side chain of a lysine residue in the constant region is represented by the following formula (II): R—C n H 2n —CO—NH— (II) (In the formula, R and n are the same as in formula (I); NH- is a group present on the side chain of a lysine residue in the constant region.
  • Is a modified antibody having a portion represented by the side chain of a lysine residue in the constant region The method according to any one of [1] to [8]. [10] The method of [9], wherein n is 7.
  • Lysine residues present in both the CH1 region of the heavy chain and the CL region of the light chain are a lysine residue at position 133 in a human IgG heavy chain and a lysine residue at position 169 in a human IgG light chain. , [13].
  • the lipoic acid protein ligase is derived from a microorganism; [16] the lipoic acid protein ligase is derived from a bacterium belonging to the genus Escherichia, a bacterium belonging to the genus Bacillus, a bacterium belonging to the genus Corynebacterium, or a bacterium belonging to the genus Staphylococcus; 15].
  • the lipoic acid protein ligase is obtained from Escherichia coli, Bacillus subtilis, Corynebacterium glutamicum (Corynebacterium glutamicum, or Staphylococcus epsimid sp.). Any one of 1) to [16].
  • the lipoic acid protein ligase is a protein selected from the group consisting of the following (A) to (C): (A) a protein comprising an amino acid sequence selected from the group consisting of SEQ ID NOs: 2, 4, 6, and 8; (B) an amino acid sequence selected from the group consisting of SEQ ID NOs: 2, 4, 6, and 8, including an amino acid sequence containing substitution, deletion, insertion, or addition of one or several amino acids, and lipoic acid A protein having protein ligase activity; and (C) a lipoic acid protein comprising an amino acid sequence having 90% or more identity to an amino acid sequence selected from the group consisting of SEQ ID NOs: 2, 4, 6, and 8.
  • a protein having ligase activity [19] The method of any one of [1] to [17], wherein the lipoic acid protein ligase is a protein selected from the group consisting of the following (A ′) to (C ′): (A ′) a protein having an amino acid sequence having one or more mutations selected from the group consisting of the following (i) to (vii) in the amino acid sequence of SEQ ID NO: 2, and having lipoic acid protein ligase activity: (I) substitution with an arginine residue at position 121, an alanine residue or a threonine residue; (Ii) substitution of a serine residue at position 136 with a leucine residue; (Iii) substitution with a tyrosine residue at position 140, an alanine residue or a valine residue; (Iv) substitution at position 142 of a glutamic acid serine residue, threonine residue, or valine residue; (V) deletion of histidine residue at position 149;
  • a bioorthogonal functional group is an azide residue, aldehyde residue, thiol residue, alkyne residue, alkene residue, halogen residue, tetrazine residue, nitrone residue, hydroxylamine residue, nitrile residue , Hydrazine residue, ketone residue, boronic acid residue, cyanobenzothiazole residue, allyl residue, phosphine residue, maleimide residue, disulfide residue, thioester residue, ⁇ -halocarbonyl residue, isonitrile residue [20] the method of [20], wherein the method is selected from the group consisting of: [22] A method for producing a modified antibody having a functional substance, comprising the following (1) and (2): (1) An antibody is reacted with a lipoic acid analog having a modified portion containing a bioorthogonal functional group.
  • the lipoic acid analog having a modified moiety containing a bioorthogonal functional group is a C 4 -C 10 alkyl-carboxylic acid having a modified moiety containing a bioorthogonal functional group;
  • a modified antibody having a modified moiety containing a bioorthogonal functional group on the side chain of a lysine residue in the constant region is used to convert a C 4 -C 10 alkyl-carbonyl having a modified moiety containing a bioorthogonal functional group into the constant region.
  • An antibody having a side chain of a lysine residue of A modified antibody having a functional substance on the side chain of a lysine residue in the constant region is characterized by a C 4 -C 10 alkyl-carbonyl having a functional substance and a modified moiety containing a bioorthogonal functional group reacted therewith, [20]
  • a lipoic acid analog having a modified moiety containing a bioorthogonal functional group is represented by the following formula (I): R—C n H 2n —COOH (I) (In the formula, R is a modifying moiety containing a bioorthogonal functional group, n is an integer of 4 to 10.
  • a modified antibody having a modified portion containing a bioorthogonal functional group on the side chain of a lysine residue in the constant region is represented by the following formula (II): R—C n H 2n —CO—NH— (II) (In the formula, R and n are the same as in formula (I); NH- is a group present on the side chain of a lysine residue in the constant region.
  • a modified antibody having a portion represented by a side chain of a lysine residue in the constant region is represented by the following formula (III): F-R'-C n H 2n -CO-NH- (III) (In the formula, n is the same as that of formula (I); NH- is the same as that of formula (II), F is a functional substance, R ′ is a divalent group including a moiety generated by a reaction between a functional substance and a bioorthogonal functional group.
  • a modified antibody having a modified portion on the side chain of a lysine residue in the constant region is represented by the following formula (II): R—C n H 2n —CO—NH— (II) (In the formula, R is a modifying moiety; n is an integer of 4 to 10, NH- is a group present on the side chain of a lysine residue in the constant region.
  • [28] The modified antibody according to [28], which has a portion represented by the formula (1) only in the side chain of a lysine residue specific to the antibody.
  • [30] the modified antibody of [28] or [29], wherein n is 7;
  • [32] the modified antibody of any of [28] to [31], wherein the lysine residue in the constant region is a lysine residue in the CH1 region;
  • [33] The modified antibody of any of [28] to [32], wherein the lysine residue in the constant region is a lysine residue present in both the CH1 region of the heavy chain and the CL region of the light chain.
  • Lysine residues present in both the CH1 region of the heavy chain and the CL region of the light chain are a lysine residue at position 133 in a human IgG heavy chain and a lysine residue at position 169 in a human IgG light chain.
  • the modified antibody of [33] a modified antibody having a functional substance on the side chain of a lysine residue in the constant region, A modified antibody comprising a functional substance and a C 4 -C 10 alkyl-carbonyl having a modifying moiety containing a bioorthogonal functional group reacted therewith only in the side chain of a lysine residue unique to the antibody.
  • a modified antibody having a functional substance on a side chain of a lysine residue in the constant region is represented by the following formula (III): F-R'-C n H 2n -CO-NH- (III) (In the formula, F is a functional substance, R ′ is a divalent group including a moiety generated by a reaction between a functional substance and a bioorthogonal functional group, n is an integer of 4 to 10, NH- is a group present on the side chain of a lysine residue in the constant region.
  • the modified antibody according to [35] which has a portion represented by [1] only in the side chain of a lysine residue specific to the antibody.
  • the method of the present invention relates to a method for preparing a constant region (eg, a CH1 region such as a lysine residue at position 133 in a human IgG heavy chain, a lysine residue at position 169 in a human IgG light chain, etc.) In the CL region).
  • a constant region eg, a CH1 region such as a lysine residue at position 133 in a human IgG heavy chain, a lysine residue at position 169 in a human IgG light chain, etc.
  • the modified antibodies of the present invention are useful, for example, as pharmaceuticals and reagents (eg, diagnostics, research reagents), and intermediates for their preparation.
  • FIG. 1-1 shows the amino acid sequence of the heavy chain of trastuzumab (SEQ ID NO: 14) in which the sugar chain has been cleaved with PNGase. Boxes and double underlined letters indicate peptide fragments (SEQ ID NOs: 16-22) by trypsin digestion. The shaded letters and the numbers subscripted indicate the modified lysine residue in the modified trastuzumab and its amino acid number based on EU numbering.
  • FIG. 1-2 shows the amino acid sequence of the light chain of trastuzumab (SEQ ID NO: 15). Boxes, single underscores, and double underscores indicate peptide fragments (SEQ ID NOs: 23-27) by trypsin digestion.
  • FIG. 2 shows the peptide fragment of LSCAASGFNIKDTYIHWVR (SEQ ID NO: 16) containing a site for modification of lysine residues (octanoic acid-introduced (+126.105 Da)) by trypsin digestion of octanoic acid-modified trastuzumab obtained in Example 2. It is a figure which shows an MS spectrum (m / z 1182.61437, bivalence).
  • FIG. 3 shows a peptide fragment of LSCAASGFNIKDTYIHWVR (SEQ ID NO: 16) containing a site for modification of lysine residues (octanoic acid-introduced (+126.105 Da)) by trypsin digestion of octanoic acid-modified trastuzumab obtained in Example 2. It is a figure which shows a CID spectrum.
  • FIG. 4 shows a peptide fragment of FTISADTSKNTAYLQMNSLR (SEQ ID NO: 17) containing a site for modification to lysine residues (octanoic acid-introduced (+126.105 Da)) by trypsin digestion of octanoic acid-modified trastuzumab obtained in Example 2.
  • FIG. 4 is a view showing an MS spectrum (m / z.796.5363, trivalent).
  • FIG. 5 shows a peptide fragment of FTISADTSKNTAYLQMNSLR (SEQ ID NO: 17) containing a site for modification to lysine residues (octanoic acid-introduced (+126.105 Da)) by trypsin digestion of octanoic acid-modified trastuzumab obtained in Example 2. It is a figure which shows a CID spectrum.
  • FIG. 17 shows a peptide fragment of FTISADTSKNTAYLQMNSLR (SEQ ID NO: 17) containing a site for modification to lysine residues (octanoic acid-introduced (+126.105 Da)) by trypsin digestion of octanoic acid-modified trastuzumab obtained in Example 2. It is a figure which shows a CID spectrum.
  • FIG. 6 shows a peptide fragment of GPSVFPLAPSSSKSTSGTAGLGCLVK (SEQ ID NO: 18) containing a site for modification of a lysine residue (octanoic acid-introduced substance (+126.105 Da)) by trypsin digestion of octanoic acid-modified trastuzumab obtained in Example 2. It is a figure which shows MS spectrum (m / z 654.60949, 4 valence).
  • FIG. 7 shows a peptide fragment of GPSVFPLAPSSSKSTSGTAGLGCLVK (SEQ ID NO: 18) containing a site for modification of lysine residues by trypsin digestion of octanoic acid-modified trastuzumab obtained in Example 2 (octanoic acid-introduced substance (+126.105 Da)). It is a figure which shows a CID spectrum.
  • FIG. 18 shows a peptide fragment of GPSVFPLAPSSSKSTSGTAGLGCLVK
  • FIG. 8 shows a peptide fragment of SCDKTHTCPPCPAPELLGGGPSVFFLPPKPK (SEQ ID NO: 19) containing a site for modification to lysine residues (octanoic acid-introduced (+126.105 Da)) by trypsin digestion of octanoic acid-modified trastuzumab obtained in Example 2.
  • FIG. 4 is a view showing an MS spectrum (m / z 1154.25572, trivalent).
  • FIG. 9 shows a peptide fragment of SCDKTHTCPPCPAPELLGGGPSVFFLPPKPK (SEQ ID NO: 19) containing a site for modification to lysine residues by trypsin digestion of octanoic acid-modified trastuzumab obtained in Example 2 (octanoic acid-introduced substance (+126.105 Da)). It is a figure which shows a CID spectrum.
  • SCDKTHTCPPCPAPELLGGGPSVFFLPPKPKPK SEQ ID NO: 19
  • FIG. 10 shows a peptide fragment of THTCPPCPAPELLGGPSVFLFPPKPK (SEQ ID NO: 20) containing a site for modification of lysine residues (octanoic acid-introduced (+126.105 Da)) by trypsin digestion of octanoic acid-modified trastuzumab obtained in Example 2. It is a figure which shows MS spectrum (m / z 1485.78552, bivalence).
  • FIG. 11 shows a peptide fragment of THTCPPCPAPELLGGPSVFLFPPPKPK (SEQ ID NO: 20) containing a site for modification of lysine residues by lysine residue by trypsin digestion of octanoic acid-modified trastuzumab obtained in Example 2 (octaneic acid-introduced substance (+126.105 Da)). It is a figure which shows a CID spectrum.
  • FIG. 12 shows the peptide fragment of VSNKALPAPIEK (SEQ ID NO: 21) containing a site for modification to lysine residues (octanoic acid-introduced (+126.105 Da)) by trypsin digestion of octanoic acid-modified trastuzumab obtained in Example 2.
  • FIG. 4 is a view showing an MS spectrum (m / z.696.8122, divalent).
  • FIG. 13 shows the peptide fragment of VSNKALPAPIEK (SEQ ID NO: 21) containing a site for modification to lysine residues (octanoic acid-introduced (+126.105 Da)) by trypsin digestion of octanoic acid-modified trastuzumab obtained in Example 2. It is a figure which shows a CID spectrum.
  • FIG. 21 shows the peptide fragment of VSNKALPAPIEK (SEQ ID NO: 21) containing a site for modification to lysine residues (octanoic acid-introduced (+126.105 Da)) by trypsin digestion of octanoic acid-modified trastuzumab obtained in Example 2. It is a figure which shows a CID spectrum.
  • FIG. 14 shows a peptide fragment of EPQVYTLPSPSREEMTKNQVSLTCLVK (SEQ ID NO: 22) containing a site for modification of a lysine residue by lysine digestion of octanoic acid-modified trastuzumab obtained in Example 2 (octanoic acid-introduced substance (+126.105 Da)). It is a figure which shows MS spectrum (m / z 794.17197, tetravalent).
  • FIG. 15 shows a peptide fragment of EPQVYTLPSPSREEMTKNQVSLTCLVK (SEQ ID NO: 22) containing a site for modification to lysine residues (octanoic acid-introduced (+126.105 Da)) by trypsin digestion of octanoic acid-modified trastuzumab obtained in Example 2. It is a figure which shows a CID spectrum.
  • SEQ ID NO: 22 a peptide fragment of EPQVYTLPSPSREEMTKNQVSLTCLVK
  • FIG. 16 shows the peptide fragment of ASQDVNTAVAWYQQKPGKAPK (SEQ ID NO: 23) containing a site for modification of lysine residues (octanoic acid-introduced (+126.105 Da)) by trypsin digestion of octanoic acid-modified trastuzumab obtained in Example 2. It is a figure which shows MS spectrum (m / z 1207.14994, bivalence).
  • FIG. 17 shows a peptide fragment of ASQDVNTAVAWYQQKPGKAPK (SEQ ID NO: 23) containing a site for modification of lysine residues (octanoic acid-introduced (+126.105 Da)) by trypsin digestion of octanoic acid-modified trastuzumab obtained in Example 2. It is a figure which shows a CID spectrum.
  • SEQ ID NO: 23 SEQ ID NO: 23
  • FIG. 18 shows a peptide fragment of VDNALQSGNSQESVTEQDSKDSTYSLSLSTLTLSK (SEQ ID NO: 24) containing a site for modification of lysine residues (octanoic acid-introduced (+126.105 Da)) by trypsin digestion of octanoic acid-modified trastuzumab obtained in Example 2. It is a figure which shows MS spectrum (m / z 1249.27813, trivalence).
  • FIG. 19 shows a peptide fragment of VDNALQSGNSQESVTEQDSKDSTYSLSSSTTLSK (SEQ ID NO: 24) containing a site for modification to lysine residues by lysine residue obtained by trypsin digestion of octanoic acid-modified trastuzumab obtained in Example 2 (SEQ ID NO: 24). It is a figure which shows a CID spectrum.
  • FIG. 20 shows a peptide fragment of ADYEKHK (SEQ ID NO: 25) containing a site for modification to lysine residues (octanoic acid-introduced (+126.105 Da)) by trypsin digestion of octanoic acid-modified trastuzumab obtained in Example 2.
  • FIG. 3 is a view showing an MS spectrum (m / z 508.77751, divalent).
  • FIG. 21 shows the peptide fragment of ADYEKHK (SEQ ID NO: 25) containing a site for modification to lysine residues (octanoic acid-introduced (+126.105 Da)) by trypsin digestion of octanoic acid-modified trastuzumab obtained in Example 2. It is a figure which shows a CID spectrum.
  • FIG. 21 shows the peptide fragment of ADYEKHK (SEQ ID NO: 25) containing a site for modification to lysine residues (octanoic acid-introduced (+126.105 Da)) by trypsin digestion of octanoic acid-modified trastuzumab obtained in Example 2. It is a figure which shows a CID spectrum.
  • FIG. 21 shows the peptide fragment of ADYEKHK (SEQ ID NO: 25) containing a
  • FIG. 22 shows a peptide fragment of HKVYACEVTHQGLSSPVTK (SEQ ID NO: 26) containing a site for modification to lysine residues (octanoic acid-introduced (+126.105 Da)) by trypsin digestion of octanoic acid-modified trastuzumab obtained in Example 2. It is a figure which shows MS spectrum (m / z 1134.08444, bivalence).
  • FIG. 23 shows a peptide fragment of HKVYACEVTHQGLSSPVTK (SEQ ID NO: 26) containing a site for modification to lysine residues (octanoic acid-introduced (+126.105 Da)) by trypsin digestion of octanoic acid-modified trastuzumab obtained in Example 2. It is a figure which shows a CID spectrum.
  • FIG. 24 shows a peptide fragment of HKVYACEVTHQGLSSPVTKSFNR (SEQ ID NO: 27) containing a site for modification of lysine residues (octanoic acid-introduced (+126.105 Da)) by trypsin digestion of octanoic acid-modified trastuzumab obtained in Example 2. It is a figure which shows MS spectrum (m / z 924.48353, trivalence).
  • FIG. 25 shows a peptide fragment of HKVYACEVTHQGLSSPVTKSFNR (SEQ ID NO: 27) containing a site for modification to lysine residues (octanoic acid-introduced (+126.105 Da)) by trypsin digestion of octanoic acid-modified trastuzumab obtained in Example 2. It is a figure which shows a CID spectrum.
  • FIG. 26 shows a CLUSTAL O (1.2.4) multiple sequence alignment of modified variants of LplA. Amino acid substitutions are underlined in bold.
  • FIG. 27 shows (A) SDS-PAGE of the reaction mixture and (B) a fluorogram of the gel.
  • the present invention provides a method for producing a modified antibody having a modifying moiety.
  • the method of the invention comprises reacting an antibody with a lipoic acid analog having a modifying moiety in the presence of lipoic acid protein ligase to produce a modified antibody having a modifying moiety on the side chain of a lysine residue in the constant region. Including.
  • the antibody used in the method of the present invention is a polyclonal antibody or a monoclonal antibody.
  • the antibody used in the method of the present invention may be an antibody (eg, glycoprotein) modified with a biomolecule (eg, sugar) or an antibody not modified with a biomolecule.
  • a biomolecule eg, sugar
  • an antibody against a biological-derived component or a virus-derived component is preferable.
  • biological components include components (eg, proteins) derived from animals such as mammals and birds (eg, chickens), insects, microorganisms, plants, fungi, and fish.
  • the biological component is a component derived from a mammal.
  • mammals include primates (eg, humans, monkeys, chimpanzees), rodents (eg, mice, rats, guinea pigs, hamsters, rabbits), pets (eg, dogs, cats), livestock (eg, Cattle, pigs, goats) and working animals (eg, horses, sheep).
  • the biological component is more preferably a primate or rodent-derived component (eg, a protein), and even more preferably a human-derived component (eg, a protein) from the viewpoint of clinical application of the present invention. is there.
  • virus-derived components include components (eg, proteins) derived from influenza virus (eg, avian influenza virus, swine influenza virus), AIDS virus, Ebola virus, and phage virus.
  • Antibody is an antibody against any antigen.
  • an antigen may be a component found in an organism or virus as described above.
  • antigens also include, for example, proteins [including oligopeptides and polypeptides. It may be a protein modified with a biomolecule such as sugar (eg, glycoprotein)], a sugar chain, a nucleic acid, and a low molecular compound.
  • the antibody may be an antibody having a protein as an antigen.
  • proteins include cell membrane receptors, cell membrane proteins other than cell membrane receptors (eg, extracellular matrix proteins), ligands, and soluble receptors.
  • the protein that is the antigen of the antibody may be a disease target protein.
  • diseases target protein include the following.
  • Ocular diseases Factor D IGF-1R, PGDFR, Ang2, VEGF-A, CD-105 (Endoglin), IGF-1R, ⁇ -amyloid
  • the antibody has a native polypeptide chain structure.
  • the “natural polypeptide chain structure” of an antibody refers to the first polypeptide chain consisting of the variable region (VH) and the constant regions (CH1, CH2, CH3) in the naturally occurring heavy chain of the antibody, and the naturally occurring antibody. It refers to the structure consisting of the second polypeptide chain consisting of the variable region (VL) and the constant region (CL) in the light chain. Therefore, antibodies (ie, VH, CH1, CH2, CH3, VL, and CL) which are fusion proteins having a structure in which a predetermined amino acid sequence (eg, a tag sequence, a spacer sequence) has been added or inserted into a predetermined region of the antibody. Antibodies having other portions) are excluded from antibodies having a natural polypeptide chain structure.
  • the antibodies used in the method of the invention are monoclonal antibodies.
  • the monoclonal antibody for example, a chimeric antibody, a humanized antibody, a human antibody, an antibody to which a predetermined sugar chain is added (for example, modified to have a sugar chain binding consensus sequence such as an N-type sugar chain binding consensus sequence) Antibodies), bispecific antibodies, scFv antibodies, Fab antibodies, F (ab ') 2 antibodies, VHH antibodies, Fc region proteins, and Fc fusion proteins.
  • Isotypes of antibodies such as monoclonal antibodies include, for example, IgG, IgM, IgA, IgD, IgE, and IgY.
  • the antibody may also be a bivalent antibody (eg, IgG, IgD, IgE) or a tetravalent or higher-valent antibody (eg, IgA antibody, IgM antibody).
  • the antibody used in the method of the present invention may be a full-length antibody, or an antibody fragment (eg, Fab, F (ab ') 2 , Fab', Fv, single chain antibody).
  • the antibody fragment preferably has either the CH1 region of the heavy chain or the CL region of the light chain, and more preferably has both the CH1 region of the heavy chain and the CL region of the light chain.
  • the antibody used in the method of the present invention is a human antibody or a humanized antibody.
  • a high degree of modification has been confirmed in a specific constant region of a humanized antibody (trastuzumab). Therefore, according to the method of the present invention, not only a humanized antibody but also a human antibody having the same constant region as a humanized antibody can highly modify a specific constant region.
  • adalimumab panitumumab, golimumab, ustekinumab, canakinumab, ofatumumab, denosumab (IgG2), ipilimumab, belimumab, raxivacumab, ramcilumab, nivolumabumalb (IgG4), Secukinumabumabu, g IgG2) and oraratumab (if no IgG subtype is mentioned, it indicates that it is an IgG1).
  • humanized antibodies examples include daclizumab, palivizumab, trastuzumab, alentuzumab, omalizumab, efalizumab, bevacizumab, natalizumab (IgG4), tocilizumab, eclidumab (IgG2), mogamulizumab, pertuzumab, pertuzumab, pertuzumab, pertuzumab, pertuzumab, pertuzumab, pertuzumab, penuzunumab , Dartatumumab, ikesekidumab (IgG4), reslizumab (IgG4), and atezolizumab (if no IgG subtype is mentioned, it indicates that it is IgG1).
  • the antibody used in the method of the present invention is an IgG.
  • IgG examples include IgG1, IgG2, IgG3, and IgG4.
  • IgG human IgG is preferable.
  • the antibody used in the method of the present invention is Fab, or F (ab ') 2 .
  • Fab or F (ab ') 2 .
  • a high degree of modification has been confirmed in specific constant regions (CH1, CL) of Fab. Therefore, according to the method of the present invention, not only Fabs but also F (ab ') 2 having the same constant regions as Fabs can be highly modified in specific constant regions.
  • Lipoic acid protein ligase (Lpl) used in the method of the present invention is an enzyme that catalyzes a reaction in which lipoic acid is bound to a lysine residue in a protein and acylated.
  • Lpl is widely found in various organisms such as microorganisms, and is known to be an enzyme capable of utilizing lipoic acid analogs having various structures as substrates (Fernandez-Suarez M et al. Nat Biotechnol). 2007 25: 1483-1487; U.S. Patent No. 8,137,925; U.S. Patent No. 9,284,541; WO 2017/095806).
  • the Lpl used in the method of the present invention is preferably Lpl derived from a microorganism from the viewpoint of easy availability.
  • Lpl used in the method of the present invention include bacteria belonging to the genus Escherichia (eg, Escherichia coli), bacteria belonging to the genus Bacillus (eg, Bacillus subtilis), and coli.
  • Bacteria belonging to the genus Corynebacterium eg, Corynebacterium glutamicum
  • bacteria belonging to the genus Staphylococcus eg, Staphylococcus epidermis from Staphylococcus epidermis
  • Staphylococcus epidermis Staphylococcus epidermis
  • Lpl used in the method of the present invention includes, for example, a protein selected from the group consisting of the following (A) to (C): (A) a protein comprising an amino acid sequence selected from the group consisting of SEQ ID NOs: 2, 4, 6, and 8; (B) an amino acid sequence selected from the group consisting of SEQ ID NOs: 2, 4, 6, and 8, including an amino acid sequence containing substitution, deletion, insertion, or addition of one or several amino acids, and lipoic acid A protein having protein ligase activity; and (C) a lipoic acid protein comprising an amino acid sequence having 90% or more identity to an amino acid sequence selected from the group consisting of SEQ ID NOs: 2, 4, 6, and 8. A protein having ligase activity.
  • one or several amino acid residues are changed by one, two, three or four kinds of mutations selected from the group consisting of deletion, substitution, addition and insertion of amino acid residues.
  • the amino acid residue mutation may be introduced into one region in the amino acid sequence, or may be introduced into a plurality of different regions.
  • the term "one or several” refers to numbers that do not significantly impair the activity of the protein.
  • the number indicated by the term “one or several” is, for example, 1 to 100, preferably 1 to 80, more preferably 1 to 50, 1 to 30, 1 to 20, 1 to 10 or 1 to 5 (eg, 1, 2, 3, 4, or 5).
  • the above may be 97% or more, 98% or more, or 99% or more.
  • NCBI National Center for Biotechnology Information
  • Lpl used in the method of the present invention may be a protein selected from the group consisting of (A ′) to (C ′): (A ′) In the amino acid sequence of SEQ ID NO: 2, an amino acid sequence having one or more (eg, 1 to 6, preferably 1 to 5) mutations selected from the group consisting of the following (i) to (vii): A protein comprising and having lipoic acid protein ligase activity: (I) substitution with an arginine residue at position 121, an alanine residue or a threonine residue; (Ii) substitution of a serine residue at position 136 with a leucine residue; (Iii) substitution with a tyrosine residue at position 140, an alanine residue or a valine residue; (Iv) substitution at position 142 of a glutamic acid serine residue, threonine residue, or valine residue; (V) deletion of histidine residue at position 149; (Vi) substitution with
  • the protein selected from the group consisting of (A ') to (C') preferably has a higher lipoic acid protein ligase activity than the protein consisting of the amino acid sequence of SEQ ID NO: 2.
  • Lipoic acid protein ligase activity can be measured by an LplA activity (labeling of IgG with octanoic azide) assay using IgG (IgG H chain or L chain or a combination thereof) as a substrate (see Example 7).
  • Lipoic acid protein ligase activity refers to the activity of binding lipoic acid to a lysine residue in a protein for acylation.
  • Lpl may have a mutation introduced at a specific site as long as it retains lipoic acid protein ligase activity.
  • the positions of amino acid residues at which mutations can be introduced, which can retain desired properties, will be apparent to those skilled in the art. Specifically, one of skill in the art would: 1) compare the amino acid sequences of multiple proteins with similar properties, and 2) identify relatively conserved and relatively non-conserved regions, 3) From the relatively conserved region and the relatively unconserved region, a region that can play a significant role in the function and a region that cannot play a significant role in the function can be predicted, respectively. We can recognize the correlation between structure and function. Therefore, those skilled in the art can specify the position of an amino acid residue at which a mutation may be introduced in the amino acid sequence of Lpl.
  • substitution of the amino acid residue may be a conservative substitution.
  • conservative substitution refers to the replacement of a given amino acid residue with an amino acid residue having a similar side chain. Families of amino acid residues having similar side chains are well known in the art.
  • such families include amino acids having a basic side chain (eg, lysine, arginine, histidine), amino acids having an acidic side chain (eg, aspartic acid, glutamic acid), and amino acids having an uncharged polar side chain (Eg, asparagine, glutamine, serine, threonine, tyrosine, cysteine), amino acids having a nonpolar side chain (eg, glycine, alanine, valine, leucine, isoleucine, proline, phenylalanine, methionine, tryptophan), ⁇ -branched side chain (Eg, threonine, valine, isoleucine), an amino acid having an aromatic side chain (eg, tyrosine, phenylalanine, tryptophan, histidine), an amino acid having a hydroxyl group (eg, alcoholic, phenolic) -containing side chain ( Example, serine, thread Nin, tyrosine), and
  • conservative substitutions of amino acids include substitution between aspartic acid and glutamic acid, substitution between arginine and lysine and histidine, substitution between tryptophan and phenylalanine, and substitution between phenylalanine and valine.
  • Lpl may also be a fusion protein linked via a peptide bond to a heterologous moiety.
  • a heterologous portion include a peptide component that facilitates purification of a target protein (eg, a histidine tag, a tag portion such as Strep-tag II; glutathione-S-transferase, maltose binding protein, and mutants thereof).
  • a peptide component that improves the solubility of the target protein eg, Nus-tag
  • a peptide component that functions as a chaperone eg, a trigger factor
  • a peptide component having other functions eg, For example, a full-length protein or a portion thereof
  • the lipoic acid analog having a modifying moiety used in the method of the present invention is not particularly limited as long as it acts as a substrate for Lpl.
  • the lipoic acid analog in the lipoic acid analog having a modified portion is a carboxylic acid having a linear or branched hydrocarbon group and optionally having a cyclic portion such as a cyclic hydrocarbon group. Lipoic acid analogs with such modified moieties are known to act as substrates for Lpl (see references above, especially US Pat. No. 8,137,925).
  • the lipoic acid analog having a modified moiety is a lipoic acid having a modified moiety.
  • the lipoic acid in the lipoic acid having a modifying moiety is a carboxylic acid having a linear or branched hydrocarbon group.
  • the lipoic acid analog having a modified moiety has a structure in which a modified moiety is added to an alkyl-carboxylic acid having the following structure (in other words, a structure in which a hydrogen atom in lipoic acid is substituted by a modified moiety).
  • the lipoic acid analog having a modified moiety is preferably a C 4 -C 10 alkyl-carboxylic acid having a modified moiety, more preferably a C 5 -C 9 alkyl-carboxylic acid having a modified moiety, and more preferably a C 4 -C 9 alkyl-carboxylic acid having a modified moiety.
  • 6 ⁇ C 8 alkyl - are more preferred carboxylic acids
  • C 7 alkyl having a modifying moiety - carboxylic acid is particularly preferred.
  • the lipoic acid analog having a modified moiety is a compound represented by the following formula (I) having a modified moiety at the alkyl terminal of “alkyl-carboxylic acid” from the viewpoint of improving reaction efficiency and the like. is there.
  • R—C n H 2n —COOH (I) (In the formula, R is a modifying moiety; n is an integer of 4 to 10. ]
  • a hyphen (-) has two moieties on both sides thereof (eg, two moieties of R and C n H 2n and two moieties of C n H 2n and COOH) covalently bonded to each other. (The same applies to other expressions).
  • n is preferably an integer of 5 to 9, more preferably an integer of 6 to 8, and particularly preferably 7.
  • an antibody having a modified moiety on the side chain of a lysine residue in the constant region is represented by the following formula (II)
  • Modified antibodies having a moiety on the side chain of a lysine residue in the constant region are generated.
  • R—C n H 2n —CO—NH— (II) (In the formula, R and n are the same as in formula (I); NH- is a group present on the side chain of a lysine residue in the constant region. ]
  • the modified moiety is not particularly limited as long as the moiety imparts an arbitrary function to such an antibody.
  • a bioorthogonal functional group or a functional substance may be used.
  • one or more (eg, two, three, four) modified moieties may be added to lipoic acid, but preferably, one modified moiety is added to lipoic acid. It may be.
  • a bio-orthogonal functional group does not react with biological constituents (eg, amino acids, nucleic acids, lipids, sugars, and phosphates) or has a slow reaction rate with biological constituents, but does not react with components other than biological constituents. Refers to a group that reacts selectively.
  • Bioorthogonal functional groups are well known in the art (eg, Sharpless ⁇ KB et al., Angew. Chem. Int. Ed. 40, 2004 (2015); Bertozzi ⁇ CR et al., Science 291, 257 (2001); Bertozzi CR et al., Nature Chemical Biology 1, 13 (2005)).
  • a bioorthogonal functional group is retained in an antibody after the reaction according to the method of the present invention, it can impart reactivity to a functional substance to such an antibody.
  • the bio-orthogonal functional group is a bio-orthogonal functional group for an antibody (protein).
  • the bioorthogonal functional group for an antibody is a group that reacts with a predetermined functional group without reacting with the side chains of the 20 natural amino acid residues constituting the antibody.
  • A alanine
  • N asparagine
  • C cysteine
  • C glutamine
  • Q glycine
  • G isoleucine
  • I isoleucine
  • L isoleucine
  • M isoleucine
  • M isoleucine
  • M isoleucine
  • M isoleucine
  • P proline
  • S serine
  • T threonine
  • W tryptophan
  • Y valine
  • V valine
  • D glutamic acid
  • E Arginine
  • H histidine
  • L lysine
  • glycine without side chains ie, being a hydrogen atom
  • glycine whose side chains are hydrocarbon groups (ie, from the group consisting of sulfur, nitrogen, and oxygen atoms)
  • Alanine, isoleucine, leucine, phenylalanine, and valine are inert to normal reactions. Therefore, the bioorthogonal functional group for the antibody (protein) has asparagine, glutamine, methionine, proline, serine, threonine, as well as the side chains of those amino acids having side chains that are inactive to normal reactions.
  • bioorthogonal functional groups that cannot react with proteins include azide residues, aldehyde residues, thiol residues, and alkene residues (in other words, the minimum unit having a carbon-carbon double bond is It is only necessary to have a certain vinylene (ethenylene) moiety. The same applies to the following.)
  • An alkyne residue in other words, it suffices to have an ethinylene moiety which is a minimum unit having a triple bond between carbon atoms. The same applies to the following).
  • Halogen residue tetrazine residue, nitrone residue, hydroxylamine residue, nitrile residue, hydrazine residue, ketone residue, boronic acid residue, cyanobenzothiazole residue, allyl residue, phosphine residue, maleimide Residue, disulfide residue, thioester residue, ⁇ -halocarbonyl residue (eg, a fluorine atom, a chlorine atom, a bromine atom or an iodine atom at the ⁇ -position Carbonyl residue, the same applies hereinafter), an isonitrile residue, a sydnone residue, and a selenium residue.
  • ⁇ -halocarbonyl residue eg, a fluorine atom, a chlorine atom, a bromine atom or an iodine atom at the ⁇ -position Carbonyl residue, the same applies hereinafter
  • an isonitrile residue a sydnone residue,
  • Antibodies can be proteins that cannot contain free thiols. In proteins that cannot contain free thiols, thiols function as bioorthogonal functional groups. Therefore, in the case of the present invention, the bioorthogonal functional group includes a thiol.
  • the bio-orthogonal functional group is an azide residue, a thiol residue, an alkyne residue, a maleimide residue, and a disulfide residue among the above-described bio-orthogonal functional groups from the viewpoint of improving reaction efficiency and the like.
  • the functional substance is not particularly limited as long as it is a substance that imparts an arbitrary function to such an antibody when retained in the antibody after the reaction according to the method of the present invention. Details of the functional substance will be described later.
  • reaction of Antibody with Lipoic Acid Analog Having Modified Portion in the Presence of Lipoic Acid Protein Ligase The reaction can be appropriately performed under conditions (mild conditions) that cannot cause denaturation / decomposition (eg, cleavage of amide bond) of the antibody.
  • a suitable reaction system for example, a buffer (eg, a phosphate buffer) at ordinary temperature (for example, about 15 to 35 ° C., preferably about 20 to 30 ° C.).
  • the pH of the buffer is, for example, 5 to 9, preferably 5.5 to 8.5, and more preferably 6.0 to 8.0.
  • the buffer may contain a suitable cofactor such as ATP.
  • the reaction time is, for example, 10 minutes to 150 hours, preferably 20 minutes to 120 hours, more preferably 30 minutes to 100 hours, and even more preferably 60 minutes to 80 hours.
  • concentration of Lpl can be appropriately adjusted.
  • the molar ratio (Y / X) of the lipoic acid analog having the modified moiety to the antibody (X) is determined by the type of the antibody and the lipoic acid analog having the modified moiety, and the lipoic acid analog having the modified moiety. It is not particularly limited since it varies depending on the number of sites in the antibody to be modified by (e.g., DAR) and the like, but is, for example, 0.1 to 100, preferably 0.5 to 80, and more preferably Is from 1 to 70, still more preferably from 2 to 50, and particularly preferably from 3 to 30.
  • Confirmation of generation of a modified antibody having a modified portion on the side chain of a lysine residue in the constant region depends on the type of the specific raw material (antibody and lipoic acid analog having the modified portion) and the molecular weight of the product
  • Confirmation of the position of the modified lysine residue in the antibody can be performed, for example, by peptide mapping.
  • Peptide mapping can be performed, for example, by protease (eg, trypsin, chymotrypsin) treatment and mass spectrometry.
  • protease eg, trypsin, chymotrypsin
  • an endoprotease is preferable.
  • Such endoproteases include, for example, trypsin, chymotrypsin, Glu-C, Lys-N, Lys-C, Asp-N.
  • Confirmation of the number of modified moieties possessed by the modified antibody having a modified moiety on the side chain of a lysine residue in the constant region is performed, for example, by electrophoresis, chromatography, or mass spectrometry, preferably by mass spectrometry. be able to.
  • the modified antibody having a modified portion on the side chain of a lysine residue in the constant region can be appropriately purified by any method such as chromatography (eg, the above-described chromatography and affinity
  • the modifying moiety is a modifying moiety that includes a bioorthogonal functional group.
  • the method of the present invention may be performed by the following method, which includes an additional step for producing a modified antibody having a functional substance. (1) An antibody is reacted with a lipoic acid analog having a modified portion containing a bioorthogonal functional group in the presence of lipoic acid protein ligase A, so that the side chain of a lysine residue in the constant region has a bioorthogonal function.
  • the above step (1) comprises the step of reacting an antibody with a lipoic acid analog having a modifying portion in the presence of a lipoic acid protein ligase to form a modified portion on the side chain of a lysine residue in the constant region. Generating a modified antibody).
  • the above step (2) is an antibody for reacting a bioorthogonal functional group in the modified antibody with a functional substance.
  • the functional substance is not particularly limited as long as it is a substance that imparts an arbitrary function to such an antibody when retained in the antibody after the reaction according to the method of the present invention, and includes, for example, a drug, a labeling substance, and a stabilizing agent. And preferably a drug or a labeling substance.
  • the functional substance may also be a single functional substance or a substance in which two or more functional substances are linked.
  • the drug may be a drug for any disease.
  • diseases include, for example, cancer (eg, lung cancer, gastric cancer, colon cancer, pancreatic cancer, kidney cancer, liver cancer, thyroid cancer, prostate cancer, bladder cancer, ovarian cancer, uterine cancer, bone cancer, skin cancer, Brain tumor, melanoma), autoimmune disease / inflammatory disease (eg, allergic disease, rheumatoid arthritis, systemic lupus erythematosus), cranial nerve disease (eg, cerebral infarction, Alzheimer's disease, Parkinson's disease, amyotrophic lateral sclerosis), Infections (eg, bacterial infections, viral infections), hereditary or rare diseases (eg, hereditary spherocytosis, non-dystrophic myotonia), eye diseases (eg, age-related macular degeneration, diabetic retinopathy, Retinitis pigmentosa), bone and orthopedic diseases (eg, osteoarthritis), blood diseases (eg, leukemia
  • the drug is an anti-cancer agent.
  • the anticancer agent include a chemotherapeutic agent, a toxin, a radioisotope, and a substance containing the same.
  • Chemotherapeutic agents include, for example, DNA damaging agents, antimetabolites, enzyme inhibitors, DNA intercalating agents, DNA cleaving agents, topoisomerase inhibitors, DNA binding inhibitors, tubulin binding inhibitors, cytotoxic nucleosides, Platinum compounds may be mentioned.
  • toxins include bacterial toxins (eg, diphtheria toxin) and plant toxins (eg, ricin).
  • radioactive isotope examples include a radioactive isotope of a hydrogen atom (eg, 3 H), a radioactive isotope of a carbon atom (eg, 14 C), a radioactive isotope of a phosphorus atom (eg, 32 P), and a radioactive isotope of a sulfur atom.
  • a radioactive isotope of a hydrogen atom eg, 3 H
  • a radioactive isotope of a carbon atom eg, 14 C
  • a radioactive isotope of a phosphorus atom eg, 32 P
  • a radioactive isotope of a sulfur atom examples include a radioactive isotope of a sulfur atom.
  • Radioactive isotope (eg, 35 S ), radioactive isotope of yttrium (eg, 90 Y), radioactive isotope of technetium (eg, 99m Tc), radioactive isotope of indium (eg, 111 In), radioactivity of iodine atom Isotopes (eg, 123 I, 125 I, 129 I, 131 I), radioisotopes of samarium (eg, 153 Sm), radioisotopes of rhenium (eg, 186 Re), radioisotopes of astatine (eg, 211 At) and a radioisotope of bismuth (eg, 212 Bi).
  • iodine atom Isotopes eg, 123 I, 125 I, 129 I, 131 I
  • radioisotopes of samarium eg, 153 Sm
  • radioisotopes of rhenium e
  • the labeling substance examples include enzymes (eg, peroxidase, alkaline phosphatase, luciferase, ⁇ -galactosidase), affinity substances (eg, streptavidin, biotin, digoxigenin, aptamer), fluorescent substances (eg, fluorescein, fluorescein isothiocyanate, rhodamine) , Green fluorescent protein, red fluorescent protein), luminescent substance (eg, luciferin, aequorin, acridinium ester, tris (2,2′-bipyridyl) ruthenium, luminol), radioisotope (eg, those described above) or And the like.
  • enzymes eg, peroxidase, alkaline phosphatase, luciferase, ⁇ -galactosidase
  • affinity substances eg, streptavidin, biotin, digoxigenin, aptamer
  • fluorescent substances eg,
  • the functional substance is also a high molecular compound, a medium molecular compound, or a low molecular compound, and is preferably a low molecular compound.
  • a low molecular compound is a compound having a molecular weight of 1500 or less.
  • a low molecular compound is a natural compound or a synthetic compound.
  • the molecular weight of the low molecular weight compound may be 1200 or less, 1000 or less, 900 or less, 800 or less, 700 or less, 600 or less, 500 or less, 400 or less, or 300 or less.
  • the molecular weight of the low molecular weight compound may also be 30 or higher, 40 or higher, or 50 or higher.
  • the low molecular weight compound may be a drug or a labeling substance as described above.
  • Examples of the low-molecular compound include amino acids, oligopeptides, vitamins, nucleosides, nucleotides, oligonucleotides, monosaccharides, oligosaccharides, lipids, fatty acids, and salts thereof.
  • the functional substance may be a peptide compound.
  • the peptide compound may be a drug such as an anticancer agent as described above, a labeling substance, or a low molecular compound.
  • the functional substance has various functional groups according to its structure.
  • the functional group of the functional substance and the bio-orthogonal functional group can be appropriately reacted.
  • the functional group that easily reacts with the bio-orthogonal functional group may vary depending on the specific type of the bio-orthogonal functional group. Those skilled in the art can appropriately select an appropriate functional group as a functional group that easily reacts with a bioorthogonal functional group (eg, Boutureira et al., Chem. Rev., 2015, 115, 2174-2195). ).
  • Examples of the functional group that easily reacts with the bioorthogonal functional group include, for example, an azide residue when the bioorthogonal functional group is an alkyne residue, and an aldehyde or ketone residue when the bioorthogonal functional group is an aldehyde residue or a ketone residue.
  • the functional substance and the bioorthogonal functional group may be a divalent group containing a triazole residue (which may or may not be condensed with another ring).
  • the divalent group containing the moiety generated by the reaction between the orthogonal functional group and the orthogonal functional group may be a divalent group containing a hydrazone residue;
  • the biological orthogonal functional group is a thiol residue, and Functional groups that readily react with the orthogonal functional groups Is a disulfide residue (or vice versa),
  • a divalent group containing a moiety formed by a reaction between a functional substance and a bioorthogonal functional group is a divalent group containing a thiosuccinimide residue Or a divalent group containing a disulfide residue (eg, Boutureira et al., Chem.
  • a divalent group containing a triazole residue (which may or may not be condensed with another ring), a divalent group containing a hydrazone residue, a divalent group containing a thiosuccinimide residue, or A divalent group containing a disulfide residue is a preferred example of a divalent group containing a moiety generated by a reaction between a functional substance and a bioorthogonal functional group.
  • a functional substance derivatized so as to have a desired functional group can be used.
  • the functional substance is a soluble protein
  • a substance derivatized so that the soluble protein has a functional group which does not naturally exist can be used.
  • Derivatization is a common technical knowledge in the art (eg, WO 2004/010957, US Patent Application Publication No. 2006/0074008, US Patent Application Publication No. 2005/0238649).
  • derivatization may be performed using a crosslinking agent as described above.
  • derivatization may be performed using a specific linker having a desired functional group.
  • such a linker may be capable of separating a functional substance and an antibody by cleavage of the linker in an appropriate environment (eg, inside or outside a cell).
  • a linker for example, a peptidyl linker (eg, a protease that is present in an intracellular protease (eg, a protease present in a lysosome or an endosome) or an extracellular protease (eg, a secretory protease)) (eg, a protease)
  • a linker that can be cleaved at a local acidic site present in vivo (eg, US Patent No. 5).
  • the linker may be self-immortal (eg, WO 02/083180, WO 04/044933, WO 05/112919).
  • the derivatized functional substance can also be simply referred to as “functional substance”.
  • the modified antibody having a functional substance on the side chain of a lysine residue in the constant region, obtained by the above steps (1) and (2), is produced by a reaction between the functional substance and a bio-orthogonal functional group.
  • divalent groups containing moieties includes, for example, a triazole residue, a hydrazone residue, a thiosuccinimide residue, a disulfide residue, an acetal residue, a ketal residue, an ester residue, a carbamoyl residue, an alkoxyalkyl residue, an imine residue.
  • Residue tertiary alkyloxycarbamate residue, silane residue, hydrazone-containing residue, phosphoramidate residue, aconityl residue, trityl residue, azo residue, vicinal diol residue, selenium residue, electron Aromatic ring-containing residue having a suction group (eg, halogen atom, boronic acid residue, mesyl, tosyl, triflate), coumarin-containing residue, sulfone-containing residue, unsaturated bond-containing chain residue, glycosyl residue Or a divalent group containing a residue selected from the group consisting of: Preferably, such a divalent group is a triazole residue, a hydrazone residue, a thiosuccinimide residue, or a disulfide residue.
  • a suction group eg, halogen atom, boronic acid residue, mesyl, tosyl, triflate
  • coumarin-containing residue s
  • the reaction in step (2) can be appropriately performed under conditions (mild conditions) that do not cause denaturation / decomposition of the antibody (eg, cleavage of an amide bond).
  • a reaction can be carried out in a suitable reaction system, for example, a buffer (eg, a phosphate buffer) at normal temperature (for example, about 15 to 35 ° C., preferably about 20 to 30 ° C.).
  • the pH of the buffer is, for example, 5 to 9, preferably 5.5 to 8.5, and more preferably 6.0 to 8.0.
  • the buffer may contain a suitable cofactor such as ATP.
  • the reaction time is, for example, 10 minutes to 150 hours, preferably 20 minutes to 120 hours, more preferably 30 minutes to 100 hours, and even more preferably 60 minutes to 80 hours.
  • concentration of Lpl can be appropriately adjusted.
  • Reactions of bioorthogonal functional groups are well known (eg, Sharless @ KB et al., Angew. Chem. Int. Ed. 40, 2004 (2015); Bertozzi ⁇ CR et al., Science # 291). , 2357 (2001); Bertozzi CR et al., Nature Chemical Biology 1, 13 (2005)).
  • the molar ratio (Z / Y) of the functional substance (Z) to the modified antibody (Y) having a modified portion containing a bioorthogonal functional group in the side chain of a lysine residue in the constant region is determined by It is not particularly limited since it varies depending on the type of the orthogonal functional group, the functional substance, and the antibody, and the number of sites in the antibody to be modified (eg, DAR). Yes, preferably from 0.5 to 80, more preferably from 1 to 70, even more preferably from 2 to 50, particularly preferably from 3 to 30.
  • Confirmation of the generation of a modified antibody having a functional substance on the side chain of a lysine residue in the constant region depends on the specific raw material and the molecular weight of the product. For example, electrophoresis, chromatography (eg, Gel filtration chromatography, ion exchange chromatography, reverse phase column chromatography, HPLC) or mass spectrometry, preferably mass spectrometry. Confirmation of the position of the modified lysine residue in the antibody can be performed, for example, by peptide mapping. Peptide mapping can be performed, for example, by protease (eg, trypsin, chymotrypsin) treatment and mass spectrometry. As the protease, an endoprotease is preferable.
  • protease eg, trypsin, chymotrypsin
  • Such endoproteases include, for example, trypsin, chymotrypsin, Glu-C, Lys-N, Lys-C, Asp-N.
  • Confirmation of the number of functional substances carried by a modified antibody having a functional substance on the side chain of a lysine residue in the constant region is performed, for example, by electrophoresis, chromatography, or mass spectrometry, preferably by mass spectrometry. Can be performed.
  • the modified antibody having a functional substance on the side chain of a lysine residue in the constant region can be appropriately purified by any method such as chromatography (eg, the above-described chromatography and affinity chromatography).
  • the present invention also provides a modified antibody having a target portion (modified portion or functional substance) on the side chain of a lysine residue in the constant region.
  • the modified antibody of the present invention has a C 4 -C 10 alkyl-carbonyl having a modified portion only on the side chain of a lysine residue unique to the antibody.
  • "An antibody-specific lysine residue” refers to the variable region (VH) and constant region (CH1, CH2, CH3) in the heavy chain of an antibody, and the variable region (VL) and constant region (CL) in the light chain of an antibody. ) refers to one or more lysine residues present in a full length antibody or antibody fragment thereof.
  • an antibody that is a fusion protein having a structure in which a predetermined amino acid sequence (eg, a tag sequence, a spacer sequence) is added or inserted into a predetermined region of the antibody a C 4 -C 10 alkyl-carbonyl having a modified portion is substituted with A modified antibody having a side chain of a lysine residue corresponding to a predetermined amino acid sequence added or inserted is excluded from the modified antibody of the present invention.
  • a predetermined amino acid sequence eg, a tag sequence, a spacer sequence
  • the modified antibody of the present invention is produced as an antibody having a different structure depending on the type of the lipoic acid analog having a modified moiety used in the above-described method of the present invention.
  • the lipoic acid analog having a modified moiety is a C 4 -C 10 alkyl-carboxylic acid having a modified moiety
  • the antibody having the modified moiety on the side chain of a lysine residue in the constant region has the modified moiety.
  • Antibodies are generated that have a C 4 -C 10 alkyl-carbonyl in the side chain of a lysine residue in the constant region.
  • the lipoic acid analog having a modified moiety is a C 6 -C 8 alkyl-carboxylic acid having a modified moiety
  • an antibody having a modified moiety on the side chain of a lysine residue in the constant region may be a C 6 having a modified moiety.
  • Antibodies are generated that have ⁇ C 8 alkyl-carbonyl in the side chain of a lysine residue in the constant region.
  • the lipoic acid analog having a modified moiety is a C 7 alkyl-carboxylic acid having a modified moiety
  • an antibody having a modified moiety on the side chain of a lysine residue in the constant region may be used as a C 7 alkyl-carbonyl having a modified moiety.
  • a modified antibody having a modified portion in the side chain of a lysine residue in a specific constant region (CH1 region of a heavy chain, CL region of a light chain) can be produced.
  • CH1 region of a heavy chain, CL region of a light chain CH1 region of a heavy chain, CL region of a light chain
  • the modified antibody of the present invention may also be a modified antibody having a modified portion on the side chain of a lysine residue present in both the CH1 region of the heavy chain and the CL region of the light chain.
  • the modified antibody of the present invention is a modified antibody having a modified portion in the side chain of both the lysine residue at position 133 in the human IgG heavy chain and the lysine residue at position 169 in the human IgG light chain.
  • the position of the amino acid residue in the light chain is indicated by the position in amino acid sequence number 15, and the position of the amino acid residue before the 120th position in the amino acid sequence number 14 in the heavy chain is defined by the position in amino acid sequence number 14.
  • amino acid residues from position 121 onwards in the amino acid sequence number 14 of the heavy chain are shown at the position of EU numbering (therefore, the lysine residue at position 133 in the human IgG heavy chain and 169 in the human IgG light chain).
  • the position of the lysine residue at position follows Eu @ numbering in human IgG, as in the present specification.).
  • the CH1 region of the heavy chain and the CL region of the light chain are amongst the constant regions near the antigen binding site, and the desired modifications present in these regions also depend on the type of modification and the target containing the antigen. However, it may be able to interact strongly with targets, including antigens.
  • modified antibodies of the present invention having desired modifications in the CH1 region and CL region may be useful as medicines or reagents.
  • the modified antibody of the present invention can have a modified portion in the side chain of two lysine residues (eg, two lysine residues at position 133) in the CH1 region of two heavy chains.
  • the modified antibodies of the present invention can also have a modified moiety in the side chain of two lysine residues (eg, two lysine residues at position 169) in the CL regions of the two light chains.
  • the modified antibody of the present invention may be one in which such a lysine residue is highly modified.
  • the modification ratio of the lysine residue at position 133 in the total modification of human IgG CH1 is 30% or more, preferably 40% or more, more preferably 50% or more, and even more preferably 60% or more. %, Particularly preferably 70% or more.
  • the modification ratio of lysine residues at positions other than position 133 in human IgG @ CH1 in the modification of human IgG @ CH1 is low (eg, less than 30%, less than 20%, less than 10%, or 5%). Less than).
  • the modification ratio of the lysine residue at position 169 in the modification of the whole human IgG @ CL may be 30% or more.
  • the modification ratio of lysine residues at positions other than position 169 in human IgG @ CL in the total modification of human IgG @ CL is low (eg, less than 30%, less than 20%, less than 10%, or 5%). Less than).
  • the modified antibodies of the present invention also include lysine residues at other positions in the heavy chain of the antibody (eg, lysine residues in the constant region (eg, lysine residue at position 222) or lysine residues in the variable region). May be modified.
  • the modification ratio of the lysine residue at position 222 in the modification of human IgGICH2 may be 30% or more.
  • the modification ratio of lysine residues at positions other than position 222 in human IgG @ CH2 in the modification of human IgG @ CH2 is low (eg, less than 30%, less than 20%, (Less than 10%, or less than 5%).
  • the modified antibody of the present invention is also a polyclonal antibody or a monoclonal antibody.
  • the modified antibody of the present invention may be an antibody (eg, glycoprotein) modified with a biomolecule (eg, sugar) or an antibody not modified with a biomolecule.
  • the modified antibody of the present invention may be any antibody against any component such as a biological component, a virus-derived component, and a component found in the environment, but is preferably an antibody against a biological-derived component or a virus-derived component.
  • the biological component include components (eg, proteins) derived from animals such as mammals and birds (eg, chickens), insects, microorganisms, plants, fungi, and fish.
  • the biological component is a component derived from a mammal.
  • mammals examples include primates (eg, humans, monkeys, chimpanzees), rodents (eg, mice, rats, guinea pigs, hamsters, rabbits), companion animals (eg, dogs, cats), livestock (eg, Cattle, pigs, goats) and working animals (eg, horses, sheep).
  • the biological component is more preferably a primate or rodent-derived component (eg, a protein), and even more preferably, from the viewpoint of clinical application of the present invention, a human-derived component (eg, a protein).
  • virus-derived components include components (eg, proteins) derived from influenza virus (eg, avian influenza virus, swine influenza virus), AIDS virus, Ebola virus, and phage virus.
  • the modified antibody of the present invention is also an antibody against any antigen.
  • an antigen may be a component found in an organism or virus as described above.
  • antigens also include, for example, proteins [including oligopeptides and polypeptides. It may be a protein modified with a biomolecule such as sugar (eg, glycoprotein)], a sugar chain, a nucleic acid, and a low molecular compound.
  • the modified antibody of the present invention may be an antibody having a protein as an antigen.
  • proteins include cell membrane receptors, cell membrane proteins other than cell membrane receptors (eg, extracellular matrix proteins), ligands, and soluble receptors.
  • the protein that is the antigen of the modified antibody of the present invention may be a disease target protein.
  • diseases target proteins include (1) cancer regions, (2) autoimmune diseases / inflammatory diseases, (3) cranial nerve diseases, (4) infectious diseases, (5) hereditary / rare diseases, and (6) Examples include the above-mentioned proteins in eye diseases, (7) bone and orthopedic fields, (8) blood diseases, and (9) other diseases.
  • the modified antibodies of the present invention have a native polypeptide chain structure, as described above.
  • the modified antibodies of the present invention are monoclonal antibodies.
  • the monoclonal antibody for example, a chimeric antibody, a humanized antibody, a human antibody, an antibody to which a predetermined sugar chain is added (for example, modified to have a sugar chain binding consensus sequence such as an N-type sugar chain binding consensus sequence) Antibodies), bispecific antibodies, scFv antibodies, Fab antibodies, F (ab ') 2 antibodies, VHH antibodies, Fc region proteins, and Fc fusion proteins.
  • Isotypes of antibodies such as monoclonal antibodies include, for example, IgG, IgM, IgA, IgD, IgE, and IgY.
  • the antibody may also be a bivalent antibody (eg, IgG, IgD, IgE) or a tetravalent or higher-valent antibody (eg, IgA antibody, IgM antibody).
  • the modified antibodies of the present invention may be full length antibodies, or antibody fragments (eg, Fab, F (ab ') 2 , Fab', Fv, single chain antibodies).
  • the antibody fragment preferably has either the CH1 region of the heavy chain or the CL region of the light chain, and more preferably has both the CH1 region of the heavy chain and the CL region of the light chain.
  • the modified antibodies of the present invention are human or humanized antibodies.
  • specific examples of the human antibody and the humanized antibody are the same as those described above.
  • the modified antibodies of the invention are IgG.
  • IgG examples include IgG1, IgG2, IgG3, and IgG4.
  • IgG human IgG is preferable.
  • the modified antibodies of the invention are Fab, or F (ab ') 2 .
  • the modified antibody of the present invention is a modified antibody having a modification portion on the side chain of a lysine residue in the constant region.
  • the modified antibody of the present invention having a modified portion on the side chain of a lysine residue in the constant region has a portion represented by the following formula (II) only on the side chain of the antibody-specific lysine residue: R—C n H 2n —CO—NH— (II) (In the formula, R is a modifying moiety; n is an integer of 4 to 10, NH- is a group present on the side chain of a lysine residue in the constant region. ]
  • n is preferably an integer of 5 to 9, more preferably an integer of 6 to 8, and particularly preferably 7.
  • the modifying moiety in the modified antibodies of the present invention comprises a bioorthogonal functional group or a functional substance.
  • a modified antibody of the present invention can be prepared by reacting the antibody with a lipoic acid analog having a modified portion containing a bioorthogonal functional group in the presence of lipoic acid protein ligase A as described above. Producing a modified antibody having a modified moiety containing a bioorthogonal functional group on the side chain of a lysine residue in the region.
  • the modified antibody of the present invention (containing a bioorthogonal functional group as a modifying moiety) thus obtained is useful as an intermediate for the synthesis of an antibody having a functional substance.
  • the thus obtained modified antibody of the present invention (containing a functional substance as a modifying moiety) is useful as a medicine or a reagent (eg, a diagnostic agent, a research reagent).
  • the modifying moiety in the modified antibodies of the present invention comprises a bioorthogonal functional group.
  • the modified antibody of the present invention (containing a bioorthogonal functional group as a modified moiety) thus obtained is useful as an intermediate for the synthesis of antibodies having various functional substances (in particular, the modified moiety).
  • Lipoic acid analogs having a functional substance cannot function as an Lpl substrate due to the size of the functional substance, or can function as an Lpl substrate but have a low reaction rate as an enzyme).
  • the modified antibody of the present invention is a modified antibody having a functional substance on the side chain of a lysine residue in the constant region.
  • the modified antibody of the present invention having a functional substance on the side chain of a lysine residue in the constant region has a portion represented by the following formula (III) only in the side chain of the antibody-specific lysine residue: F-R'-C n H 2n -CO-NH- (III) (In the formula, F is a functional substance, R ′ is a divalent group including a moiety generated by a reaction between a functional substance and a bioorthogonal functional group, n is an integer of 4 to 10, NH- is a group present on the side chain of a lysine residue in the constant region. ]
  • the functional substance, the bio-orthogonal functional group, and the divalent group containing the moiety generated by the reaction between the functional substance and the bio-orthogonal functional group are the same as those described above. is there.
  • n is preferably an integer of 5 to 9, more preferably an integer of 6 to 8, and particularly preferably 7.
  • the modified antibody of the present invention having a functional substance on the side chain of a lysine residue in the constant region can be obtained by the method of the present invention including the above-described steps (1) and (2).
  • the modified antibody of the present invention thus obtained is useful as a medicine or a reagent (eg, a diagnostic agent, a research reagent).
  • Example 1 Search for Modification Activity of IgG Antibody (1-1) Expression and purification of E. coli LplA As lipoic acid protein ligase (Lpl), a fusion protein of LplA (EcLplA) derived from Escherichia coli and glutathione-S-transferase (GST) was used.
  • Lpl lipoic acid protein ligase
  • EcLplA fusion protein of LplA derived from Escherichia coli and glutathione-S-transferase
  • EcLplA expression plasmid In order to obtain a plasmid that expresses a fusion protein of EcLplA and GST (hereinafter simply referred to as “EcLplA expression plasmid”), the base sequence encoding the full-length LplA sequence is cloned into a pCold GST DNA vector (TaKaRa), and pCold -LplA (Ec) was constructed. A histidine tag derived from a vector is added to pCold-IplA (Ec).
  • PCold-LplA (Ec) was constructed by the following method.
  • E. FIG. A DNA fragment containing a base sequence encoding EcLplA by PCR using genomic DNA of E. coli strain MG1655 as a template chain and lplA (Ec) fw (SEQ ID NO: 9) and lplA (Ec) rv (SEQ ID NO: 10) as primers I got PCR was performed using PrimeStar polymerase (TaKaRa) with the reaction composition described in the attached protocol. The PCR cycle was performed at 94 ° C for 5 minutes, followed by 30 cycles of 98 ° C for 5 seconds, 55 ° C for 10 seconds, and 72 ° C for 2 minutes, and finally, at 4 ° C.
  • TaKaRa PrimeStar polymerase
  • a DNA fragment of pCold ⁇ GST ⁇ DNA was obtained by PCR using pCold ⁇ GST ⁇ DNA (SEQ ID NO: 11) as a template DNA and oligonucleotides of SEQ ID NOS: 12 and 13 as primers.
  • the PCR was performed using PrimeStar polymerase with the reaction composition described in the attached protocol.
  • the PCR cycle was 94 ° C for 5 minutes, followed by 30 cycles of 98 ° C for 5 seconds, 55 ° C for 10 seconds and 72 ° C for 5 minutes, and finally 4 cycles.
  • the test was performed under the condition of keeping the temperature at °C.
  • the obtained DNA fragments were ligated using an In-Fusion @ HD cloning kit (Clontech) to construct an EcLplA expression plasmid pCold-lplA (Ec).
  • the nucleotide sequence of lplA (Ec) and the amino acid sequence encoded thereby are shown in SEQ ID NOs: 1 and 2, respectively.
  • the EcLplA expression plasmid pCold-lplA (Ec) was coli JM109 strain (TaKaRa) was introduced by a transformation method using heat shock to obtain a JM109 / pCold-lplA (Ec) strain. Transformation using heat shock was performed according to the protocol described in the attached protocol. This strain was transformed into an LB medium (1.0% (w / v) peptone, 0.50% (w / v) yeast extract, 1.0% (w / v) NaCl) supplemented with 100 ⁇ g / mL ampicillin. The cells were inoculated and cultured with shaking at 37 ° C. overnight.
  • the culture solution was inoculated to a final concentration of 1% in an LB medium in which 100 mL of the medium was inserted into a Sakaguchi flask. After culturing with shaking at 37 ° C. for 3 hours, isopropyl- ⁇ -D-thiogalactopyranoside (IPTG) was added to a final concentration of 1.0 mM, and the culture temperature was further reduced to 15 ° C. Culture was performed for 16 hours.
  • IPTG isopropyl- ⁇ -D-thiogalactopyranoside
  • an equilibration buffer (elution buffer) to which imidazole having a final concentration of 300 mM was added was passed through the column to collect a fraction containing EcLplA, and 1.0 mL of a 0.63 mg / mL fraction was used as purified EcLplA.
  • IgG antibody modifying activities of various enzymes IgG antibody modifying activities of various enzymes were evaluated.
  • the enzymes Lpl, 4'-phosphopantetheinyl transferase (4'-phosphopantheinyl transferase, PPTase), and biotin ligase were used.
  • an IgG antibody an anti-HER2 IgG antibody trastuzumab (Chugai Pharmaceutical) was used.
  • the antibody modification reaction with ⁇ ⁇ ⁇ Lpl was performed as follows. EcLplA prepared in (1-1) was used as Lpl. 8-Azidooctanoic acid was used as a modification.
  • the reaction composition is as follows. 500 ⁇ g of trastuzumab was dissolved in 250 ⁇ L of 1.0 mM sodium phosphate buffer (pH 6.0) and used. A mixed solution having a final concentration of 1.0 mg / mL trastuzumab, ATP 2.5 mM, 8-azidooctanoic acid (Sundia) 0.50 mM, magnesium sulfate 3.2 mM, and sodium phosphate buffer 25 mM (pH 7.0).
  • EcLplA having a final concentration of 0.050 mg / mL was added thereto to prepare a reaction solution having a total volume of 100 ⁇ L, and reacted at 30 ° C. for 15 hours to produce 8-azidooctanoic acid-modified trastuzumab as a modified antibody.
  • 2 ⁇ L of an EDTA-2Na solution was added to the reaction solution (final concentration: 10 mM) to stop the reaction.
  • ⁇ Antibody modification reaction with PPTase was performed as follows. SFP @ synthase (New @ England @ Biolabs) was used as the PPTase. CoA lithium salt (Sigma-Aldrich) was used as a modifier.
  • the reaction composition is as follows. A final concentration of a mixture of trastuzumab 1.0 mg / mL, CoA lithium salt (Sigma-Aldrich) 0.50 mM, magnesium sulfate 2.0 mM, Hepes-KOH buffer 50 mM (pH 7.5) as a reaction solution was added to the final concentration. 1.0 ⁇ M SFP @ synthase was added to make a total volume of 100 ⁇ L, and reacted at 30 ° C. for 15 hours to generate CoA-modified trastuzumab as a modified antibody. 2 ⁇ L of an EDTA-2Na solution was added to the reaction solution (final concentration: 10 mM) to stop the reaction.
  • Antibody modification with biotin ligase was performed as follows. BirA (Abcam) was used as biotin ligase. Biotin was used as a modification.
  • the reaction composition is as follows. A final concentration of 1.050 mg / mL of trastuzumab, 1.0 mM of ATP, 0.50 mM of biotin, 1.0 mM of magnesium sulfate, and 50 mM of Tris-HCl buffer (pH 8.3) was added to a mixed solution having a final concentration of 0.050 mg / mL. mL of BirA was added to make a total volume of 100 ⁇ L, and reacted at 30 ° C. for 15 hours to generate biotin-modified trastuzumab as a modified antibody. 2 ⁇ L of an EDTA-2Na solution was added to the reaction solution (final concentration: 10 mM) to stop the reaction.
  • the modified antibody was purified using a Protein A column (Protein A HP MultiTrap (96 well), GE Healthcare). The Protein A column used was equilibrated with 20 mM Tris (pH 7.6) and 20% (v / v) ethanol. A centrifugal separator was used to pass the solution, and centrifugation was performed at 900 g for 1 minute at each stage.
  • 500 ⁇ L of the enzyme reaction solution containing 500 ⁇ g of trastuzumab was divided into two wells of 250 ⁇ L each, applied, and washed twice with 300 ⁇ L of equilibration buffer. Further, 200 ⁇ L of 100 mM glycine buffer (pH 2.7) was added and eluted. Immediately, 15 ⁇ L of 1.0 M Tris-HCl was added to adjust the pH to around 8.5 to obtain a modified antibody sample.
  • the Lpl reaction product and the PPTase reaction product were evaluated for antibody modification by the following method.
  • the modified antibody sample fluorescently labeled in (1-4) was purified in the same manner as in (1-3).
  • the modification to the antibody was evaluated by quantifying the amount of the fluorescent label in the purified sample after the fluorescent labeling. All analyzes were performed on a 96-well plate, and fluorescence was detected at an excitation wavelength of 520 nm and a fluorescence wavelength of 580-640 nm.
  • the biotin ligase reaction product was evaluated for antibody modification by the following method.
  • the modification to the antibody was evaluated by quantifying the concentration of side chain biotin in the modified antibody sample purified in (1-3) using a Fluorescence Biotin Quantitation Kit (manufactured by Thermo Fisher Scientific).
  • the Fluorescence Biotin Quantitation Kit is capable of quantifying the concentration of side-chain biotin in a solution by fluorescent labeling, and the quantification has been confirmed from 0 to 6.0 ⁇ M.
  • detection was performed at an excitation wavelength of 475 nm and a fluorescence wavelength of 500 to 550 nm. GroMAX (Promega) was used as a plate reader.
  • modification to the antibody was observed only in the Lpl reaction product (Table 2).
  • Example 2 Modification of IgG antibody by LplA and analysis by ESI-MS (2-1) Enzyme reaction and analysis by MS As IgG antibody and LplA, anti-HER2 IgG antibody trastuzumab (Chugai Pharmaceutical) and (1-1) The prepared EcLplA was used.
  • the reaction composition is as follows. 500 ⁇ g of trastuzumab was dissolved in 250 ⁇ L of 1 mM sodium phosphate buffer (pH 6.0) and used.
  • a final concentration of 1.0 mg / mL of trastuzumab, 2.5 mM of ATP, 0.5 mM of octanoic acid, 3.2 mM of magnesium sulfate, and 25 mM of sodium phosphate buffer (pH 7.0) in the final concentration was added to the final mixture.
  • a total of 500 ⁇ L of a reaction solution was prepared by adding 0.05 mg / mL EcLplA, and reacted at 30 ° C. for 64 hours to produce octanoic acid-modified trastuzumab as a modified antibody.
  • the reaction was terminated by adding 10 ⁇ L of an EDTA-2Na solution (final concentration: 10 mM) to the reaction solution.
  • LplA has the ability to modify antibodies, and modifies antibodies with 1 to 10 modifications per antibody.
  • Example 3 Modification of IgG with various LplAs and analysis by ESI-MS (3-1) Expression and purification of various LplAs As other LplAs described in Example 1, LplA derived from Bacillus subtilis (BsLplA) and Corynebacterium glutamicum. LplA (CgLplA) and LplA derived from Staphylococcus epidermides (SeLplA) were used.
  • nucleotide sequences encoding various LplA full-length sequences were cloned into a pCold GST DNA vector, and lplA expression plasmids pCold-lplA (Bs), pCold-lplA (Cg), and pCold-lplA (Se) were constructed.
  • lplA (Bs), lplA (Cg), and lplA (Se) base sequences (6 bases each having a BamHI and HindIII recognition sequence added to the 5′- and 3′-ends), and lplA (Bs), lplA
  • the amino acid sequence of (Cg), lplA (Se) is shown in SEQ ID NOs: 3, 5, and 7, and SEQ ID NOs: 4, 6, and 8.
  • ⁇ pCold-lplA (Bs), pCold-lplA (Cg) and pCold-lplA (Se) were purchased from E. coli. coli JM109 strain.
  • a purified enzyme solution was prepared from a cell extract prepared by culturing the transformant by the method described in (1-1).
  • the modified antibody was purified by a Protein A column in the same manner as in (1-3) to prepare a purified and modified antibody sample.
  • DIBAC-HHHHHHG-OH peptide (SEQ ID NO: 28) DIBAC-HHHHHHG-OH peptide (SEQ ID NO: 28) as a functional substance capable of binding to an azide group by a click reaction was prepared by the following reaction It was synthesized according to the scheme.
  • the peptide synthesizer used was Initiator + Alstra manufactured by Biotage. All reagents used were made by Watanabe Chemical Industry.
  • As the resin H-Gly-Trt (2-Cl) -Resin (0.89 mmol / g) was used.
  • the antibody was purified from the reaction solution after the click reaction using a Protein A column in the same manner as in (1-3).
  • the fraction containing the eluted antibody was solvent-exchanged with a 1,000-fold volume of a 20 mM ammonium acetate solution (pH 6.4) using an ultrafiltration unit (Amicon Ultra 0.5 mL 10 kDa, Merck Millipore), and then concentrated to concentrate IgG. After confirming that the concentration was 0.50 mg / mL or more, analysis by MS was performed.
  • Example 4 Identification of Modification Position by Peptide Mapping (4-1) Identification of Modification Position by Peptide Mapping 10 ⁇ L of the sample solution obtained in Example 2 in 50 mL low-adsorption microtest tube, 50 mM ammonium bicarbonate 10 ⁇ L of a 20 mM dithiothreitol aqueous solution dissolved in a buffer solution and 40% trifluoroethanol was added, and the mixture was heated at 65 ° C. for 1 hour. Reacted. After the reaction, 40 ⁇ L of a 50 mM ammonium bicarbonate buffer was added and stirred, and 10 ⁇ L of a 20 ng / ⁇ L aqueous trypsin solution was added, followed by enzyme digestion at 37 ° C. for 18 hours. After the digestion, 2 ⁇ L of a 20% aqueous trifluoroacetic acid solution was added to stop the reaction, and LC / MS / MS measurement was performed.
  • Sequence ⁇ HT was used as a search engine, and the range of the precursor ion was set to 350 to 5000 Da.
  • trypsin was set as a digestive enzyme, and Maximum ⁇ Missed ⁇ Clearage ⁇ Sites was set to 3.
  • Mass @ Tolerance of the precursor and the fragment ion were set to 5 ppm and 0.5 Da, respectively.
  • Static @ Modification Carbamidomethyl (+57.021 Da) was set as a modification of cysteine residue with iodoacetamide.
  • the divalent y17 also shows the modification of the lysine residue at position 133 in EU numbering of the heavy chain. A corresponding product ion of m / z 875.67 (theoretical value: 875.47) was confirmed (FIG. 7).
  • MS spectrum of peptide fragment of SCDKTHTCPPCPAPELLGGPSVFLFPPPKPK (SEQ ID NO: 19), which is a peptide consisting of 30 amino acids including a site for modification to lysine residue (octanoic acid-introduced substance (+126.105 Da)) by trypsin digestion (actual value: m / Z 1154.252572, theoretical value: 1154.25381; trivalent) (FIG.
  • the monovalent y5 also shows the modification of the lysine residue at position 246 or 248 of the heavy chain.
  • a corresponding product ion of m / z 692.46 (theoretical value: 692.47) was confirmed (FIG. 11).
  • an MS spectrum (actual measurement: m / Z 696.92812, theoretical value: 696.992724, divalent) was observed (FIG. 12), and the CID spectrum also shows modification of the lysine residue at position 326 of the heavy chain, m corresponding to monovalent b4.
  • a product ion of / z 555.38 (theoretical value: 555.35) was confirmed (FIG. 13).
  • MS spectrum of peptide fragment of EPQVYTLPPPSREEMTKNQVSLTCLVK (SEQ ID NO: 22) consisting of a peptide consisting of 26 amino acids including a modification site to lysine residue by lysine digestion (octanoic acid-introduced substance (+126.105 Da)) (actual value: m / Z 794.17197, theoretical value: 794.17052, 4 valences) (FIG. 14), and the CID spectrum shows a modification of the lysine residue at the 360-position of the heavy chain, which corresponds to divalent b18. A product ion of / z 1128.23 (theoretical value: 1128.07) was confirmed (FIG. 15).
  • MS spectrum of a peptide fragment of ASQDVNTAVAWYQQKPGKAPK (SEQ ID NO: 23) comprising a peptide consisting of 21 amino acids including a modification site to lysine residue by lysine digestion (octanoic acid-introduced substance (+126.105 Da)) (actual value: m / Z 1207.14994, theoretical value: 1207.14792, divalent) (FIG. 16), and the CID spectrum shows the modification of the lysine residue at position 42 in amino acid SEQ ID NO: 15 of the light chain.
  • the product ion of m / z 626.41 (theoretical value: 626.42) corresponding to was obtained (FIG. 17).
  • MS spectrum (actual value: m) of a peptide fragment of VDNALQSGNSQESVTEQDSKDSTYSLSLSTLTLSK (SEQ ID NO: 24), which is a peptide consisting of 34 amino acids including a site for modification to a lysine residue (octanoic acid-introduced substance (+126.105 Da)) by trypsin digestion. / Z 1249.227813, theoretical value: 1249.276768, trivalent) (FIG. 18), and the CID spectrum shows a modification of the lysine residue at position 169 of the light chain, which corresponds to divalent b22.
  • Example 5 Using the peptide-spectrum match (Psm) of the tryptic digest of the modified trastuzumab obtained in Example 1 as an index, the ratio of highly modified positions in trastuzumab was calculated to be about 79% in the heavy chain CH1 region. % was a modification to the lysine residue at position 133, and about 33% of the light chain CL region was confirmed to be a modification to the lysine residue at position 169 (Table 3). In the heavy chain CH2 region, modification to the lysine residue at position 222 was about 32% (Table 3).
  • Fab was purified by a Protein G column (HiTrap Protein G HP Columns, 1 mL, GE Healthcare).
  • a Protein G column a column equilibrated with 20 mM Tris (pH 7.6) and 20% (v / v) ethanol was used.
  • 500 ⁇ L of the enzyme reaction solution containing 1 mg of Fab the plate was washed with 4 mL of equilibration buffer. Further, 2 mL of a 100 mM glycine buffer (pH 2.7) was added and eluted. Immediately, 150 ⁇ L of 1 M Tris-HCl was added to adjust the pH to around 8.5 to obtain a purified Fab sample.
  • the purified Fab was subjected to solvent exchange and concentration in the same manner as in (3-4), and the mass was measured by ESI-MS. As a result, a peak was observed at 47638 for the raw Fab, and one octanoic acid azide was introduced into the purified product. Peaks were observed at 47805 introduced and 47971 introduced at two sites.
  • PCR1 primers P4, P5; DNA template pET15-wt-lplA. Protocol: 95 ° C, 3 minutes; 98 ° C, 20 seconds; 60 ° C, 15 seconds; 72 ° C, 15 seconds; 25 cycles. As a result, a DNA fragment F1 was obtained.
  • PCR2 primers P3, P6; DNA template pET15-wt-lplA.
  • Protocol 95 ° C, 3 minutes; 98 ° C, 20 seconds; 60 ° C, 15 seconds; 72 ° C, 15 seconds; 25 cycles.
  • a DNA fragment F2 was obtained.
  • the pET15b vector was digested with XbaI and BamHI, and ligated to a DNA fragment F1 digested with XbaI-SacI and a DNA fragment F2 digested with SacI-BamHI.
  • a pET15b-wt-lplA (SacI) plasmid was constructed.
  • PCR2 primers P9 and P11; DNA template 1 ⁇ L of the obtained PCR1 mixture. Protocol: 95 ° C, 3 minutes; 98 ° C, 20 seconds; 64 ° C, 15 seconds; 72 ° C, 30 seconds; 25 cycles. As a result, a DNA fragment F2 was obtained.
  • PCR3 primers P10, P12; DNA template pET15-lplA (SacI). Protocol: 95 ° C, 3 minutes; 98 ° C, 20 seconds; 64 ° C, 15 seconds; 72 ° C, 30 seconds; 25 cycles. As a result, a DNA fragment F3 was obtained.
  • PCR4 primers P13, P14; equimolar mixture of DNA templates F2 and F3 DNA fragments (0.3 ⁇ M each). Protocol: 95 ° C, 3 minutes; 98 ° C, 20 seconds; 64 ° C, 15 seconds; 72 ° C, 1 minute; 25 cycles.
  • a DNA fragment mixture (library 1) encoding four modified LplA variants (R121A, V, S, T) was obtained.
  • Modified LplA variant encodes a combinatorial library of R121 (A, V, S, T) * Y140 (A, V) * E142 (A, V, S, T) (total of 32) Construction of DNA fragment Primers P15 (SEQ ID NO: 52), P16 (SEQ ID NO: 53), P17 (SEQ ID NO: 54), P18 (SEQ ID NO: 55), P19 (SEQ ID NO: 56), P20 (SEQ ID NO: 57), P21 (SEQ ID NO: 57) No. 58) and P22 (SEQ ID NO: 59) were used.
  • PCR1 primers P15, P16; 1 ⁇ L of DNA template library 1 DNA mixture.
  • Protocol 95 ° C, 3 minutes; 98 ° C, 20 seconds; 65 ° C, 15 seconds; 72 ° C, 1 minute; 20 cycles.
  • PCR2 primers P17 and P19; DNA template 1 ⁇ L of the obtained PCR1 mixture.
  • Protocol 95 ° C, 3 minutes; 98 ° C, 20 seconds; 64 ° C, 15 seconds; 72 ° C, 30 seconds; 25 cycles.
  • PCR3 primers P18, P20; 1 ⁇ L of DNA template library 1 DNA mixture.
  • Protocol 95 ° C, 3 minutes; 98 ° C, 20 seconds; 64 ° C, 15 seconds; 72 ° C, 30 seconds; 25 cycles.
  • DNA fragment 3 was obtained.
  • PCR4 Primers P21 and P22; DNA templates F2 and F3 equimolar mixtures of DNA fragments (0.3 ⁇ M each). Protocol: 95 ° C, 3 minutes; 98 ° C, 20 seconds; 64 ° C, 15 seconds; 72 ° C, 1 minute; 25 cycles.
  • R121 (A, V, S, T) * Y140 (A, V) * E142 (A, V, S, T) was obtained.
  • PCR1 primers P23, P24, P25; DNA template library 2 1 ⁇ L of DNA mixture. Protocol: 95 ° C, 3 minutes; 98 ° C, 20 seconds; 65 ° C, 15 seconds; 72 ° C, 1 minute; 20 cycles.
  • PCR2 primers P26, P28; DNA-templates, 1 ⁇ L of the resulting PCR1 mixture. Protocol: 95 ° C, 3 seconds; 98 ° C, 20 seconds; 64 ° C, 15 seconds; 72 ° C, 30 seconds; 25 cycles. As a result, a DNA fragment F2 was obtained.
  • PCR3 primers P27, P28; 1 ⁇ L of DNA template library 2 DNA mixture.
  • Protocol 95 ° C, 3 minutes; 98 ° C, 20 seconds; 64 ° C, 15 seconds; 72 ° C, 30 seconds; 25 cycles.
  • PCR4 primers P30, P31; an equimolar mixture of DNA templates F2 and F3.
  • Protocol 95 ° C, 3 minutes; 98 ° C, 20 seconds; 64 ° C, 15 seconds; 72 ° C, 1 minute; 25 cycles.
  • the modified LplA variant R121 (A, V, S, T) * Y140 (A, V) * E142 (A, V, S, T) * K176 (A, V, S, T) * 178 (A , V) (mixture of DNA fragments encoding the combinatorial library) (library 3).
  • BL21 (DE3) / pE15b-wt-lplA strain capable of expressing wt-LplA and a BL21 (DE3) / pET15b strain capable of expressing one chromosome copy of the wt-lplA gene were constructed.
  • BL21 (DE3) / pET15b-wtLplA strain was inoculated into 10 wells to measure wtLplA activity expressed in the plasmid on all plates. Then, the microplate was cultured at 300 RPM and 37 ° C. for about 2 hours using a shaker (Lab-Therm LT-X, Kuhner (registered trademark)). When the OD 595 reaches about 1, IPTG (isopropyl ⁇ -D-1-thiogalactopyranoside) is added to each well to a concentration of 1 mM, and cultured for about 2 hours to obtain protein synthesis. Was induced.
  • the cells were centrifuged at 4 ° C., 4000 RPM for 10 minutes using an Eppendorf 5920R centrifuge equipped with an S-4x Universal-L rotor, and the cells were collected and the supernatant was removed.
  • Cells in each well were resuspended in 60 ⁇ L of cell lysis buffer containing: 50 mM Tris-HCl pH 7.5, 5% glycerol (v / v), 1 mM DTT (dithiothreitol), 1 mM PMSF (Phenylmethylsulfonyl fluoride). Thereafter, lysozyme was added to each well to a final concentration of 300 ⁇ g / mL, and a Kuhner shaker was used.
  • the cell suspension was incubated at 100 rpm for 4 hours at 10 ° C. Then, 5 mM MgCl 2 and 1 ⁇ g / mL of DNase I (Sigma) were added and the reaction mixture was incubated at 10 ° C. for 30 minutes at 100 rpm.
  • the obtained crude cell lysate was filtered using a 96-well filter plate (Corning; 0.2 ⁇ m, PDVF hydrophobic membrane), and the flow-through fraction was prepared as a cell-free extract containing the LplA protein.
  • LplA activity labeling of IgG with octanoic acid azide assay of crude cell lysate
  • the reaction was incubated at 30 ° C. for about 16 hours (overnight).
  • DBCO-Cy3 Sigma was added to each reaction solution until the final concentration became 30 ⁇ M, and the mixture was incubated at 30 ° C.
  • each mLplA was calculated by normalizing as follows: [F (i) -F (N)] / F (P), where F (i) is the fluorescence of the i fraction and F (i) (N) is the average fluorescence intensity of 10 reactions using the crude cell-free extract of BL21 (DE3) / pET15b strain, and F (P) is BL21 (DE3) / pET15b-wt-lplA (SacI).
  • Protocol 95 ° C, 3 minutes; 98 ° C, 20 seconds; 64 ° C, 15 seconds; 72 ° C, 1 minute; 25 cycles.
  • the obtained DNA fragment was digested with NcoI and BamHI restriction enzymes, and ligated with the pET15b plasmid digested with the same restriction enzymes.
  • X wt, m231, m746, m766, m876) were constructed.
  • IPTG was added to a final concentration of 1 mM, and the cells were cultured at 37 ° C. for 2 hours.
  • the obtained cells were collected by centrifugation, washed twice with 50 mL of 0.9% NaCl solution, and frozen at ⁇ 20 ° C. until use.
  • the lysed cells were resuspended in 4 mL of buffer A (20 mM Tris-HCl, 20 mM imidazole, 500 mM NaCl, pH 7.4), and the suspension was divided into five 1.5 vials by 0.8 mL.
  • the cells were destroyed by sonication.
  • the insoluble fraction of the lysate was removed by centrifugation at 13000 RPM for about 20 minutes.
  • the resulting cell-free extract was applied to a 1 mL HiTrap Chelating column (GE healthcare) and purified according to standard protocols.
  • the buffer of the eluted fraction containing N-Tag6His-m-LplA was exchanged with a 50 mM NaPi buffer (pH 7) using a 5 mL HiTrap desalting column (GE Healthcare).
  • the resulting solution was concentrated on an Amicon 10K membrane until the final protein concentration was 0.5-1 mg / L.
  • the reaction was incubated at 30 ° C. for 48 hours, and 20 ⁇ L of the reaction mixture was sampled over time. The reaction was stopped by the addition of EDTA (10 mM) and stored frozen ( ⁇ 20 ° C.) until analysis. To this reaction solution, DBCO-Cy3 was added at 100 ⁇ M, and incubated at 30 ° C. for 5 hours. The reaction solution after labeling was applied to an Amicon Ultra 0.5 ml 10 kDa filter, and washed three times with 0.35 mL of PBS to which ethanol (20%) was added.
  • the present invention is useful, for example, for producing regioselectively modified antibodies.
  • SEQ ID NOs: 1 and 2 show the nucleotide sequence of LplA (EcLplA) derived from Escherichia coli and the amino acid sequence encoded thereby, respectively.
  • SEQ ID NOS: 3 and 4 show the amino acid sequences of Bacillus subtilis-derived LplA (BsLplA) (having 6 bases of BamHI and HindIII recognition sequences added at the 5′- and 3′-ends) and BsLplA, respectively. .
  • SEQ ID NOs: 5 and 6 show the amino acid sequences of Corynebacterium glutamicum-derived LplA (CgLplA) (having 6 bases of BamHI and HindIII recognition sequences at the 5′- and 3′-ends) and CgLplA, respectively.
  • SEQ ID NOs: 7 and 8 show the amino acid sequences of Staphylococcus epidermides-derived LplA (SeLplA) (having 6 bases of BamHI and HindIII recognition sequences added at the 5′- and 3′-ends) and BsLplA, respectively.
  • SEQ ID NOS: 9 and 10 show the nucleotide sequences of PCR primers for amplification of EcLplA (lplA (Ec) fw and lplA (Ec) rv), respectively.
  • SEQ ID NO: 11 shows the nucleotide sequence of plasmid vector pCold GST DNA.
  • SEQ ID NOs: 12 and 13 show the nucleotide sequences of PCR primers for pCold GST DNA amplification (pCold GST fw and pCold GST rv), respectively.
  • SEQ ID NOS: 14 and 15 show the amino acid sequence of the heavy chain of trastuzumab and the amino acid sequence of the light chain of trastuzumab, respectively, in which the sugar chain is cleaved with PNGase.
  • SEQ ID NOs: 16 to 22 show the amino acid sequences of peptide fragments of the heavy chain region obtained by trypsin digestion of trastuzumab.
  • SEQ ID NOs: 23 to 27 show the amino acid sequences of peptide fragments of the light chain region obtained by trypsin digestion of trastuzumab.
  • SEQ ID NO: 28 shows the amino acid sequence of a model peptide of a functional substance that modifies trastuzumab in Example 3.
  • SEQ ID NOs: 29 to 37 show known LplA recognition amino acid sequences.
  • SEQ ID NOs: 38 to 68 show the nucleotide sequences of primers P1 to P31, respectively.
  • SEQ ID NOS: 69 and 70 show the base sequence of the modified LplA variant m231 and the amino acid sequence encoded thereby, respectively.
  • SEQ ID NOs: 71 and 72 show the nucleotide sequence of the modified LplA variant m746 and the amino acid sequence encoded thereby, respectively.
  • SEQ ID NOS: 73 and 74 show the nucleotide sequence of the modified LplA variant m766 and the amino acid sequence encoded thereby, respectively.
  • SEQ ID NOs: 75 and 76 show the base sequence of the modified LplA variant m876 and the amino acid sequence encoded thereby, respectively.
  • SEQ ID NOs: 77 and 78 show the nucleotide sequences of primers P32 and P33, respectively.

Landscapes

  • Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Biochemistry (AREA)
  • General Health & Medical Sciences (AREA)
  • Biophysics (AREA)
  • Genetics & Genomics (AREA)
  • Medicinal Chemistry (AREA)
  • Molecular Biology (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Immunology (AREA)
  • Peptides Or Proteins (AREA)
  • Preparation Of Compounds By Using Micro-Organisms (AREA)

Abstract

La présente invention concerne une technique qui permet la modification d'un anticorps. Plus particulièrement, la présente invention concerne les inventions suivantes : (1) un procédé de production d'un anticorps modifié présentant un fragment modifié, le procédé consistant à faire réagir un anticorps avec un analogue d'acide lipoïque présentant un fragment modifié en présence de l'acide lipoïque protéine ligase A pour produire un anticorps modifié présentant un fragment modifié dans une chaîne latérale d'un résidu de lysine dans une région constante ; (2) un anticorps modifié présentant un fragment modifié dans une chaîne latérale d'un résidu de lysine dans une région constante, l'anticorps modifié présentant un C4-C10-alkyl-carbonyle présentant un fragment modifié dans seulement un chaîne latérale d'un résidu lysine spécifique à l'anticorps ; etc.
PCT/JP2019/026521 2018-07-03 2019-07-03 Anticorps modifié et procédé pour sa production WO2020009165A1 (fr)

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
JP2018-127089 2018-07-03
JP2018127089 2018-07-03
RU2019106926 2019-03-12
RU2019106926 2019-03-12

Publications (1)

Publication Number Publication Date
WO2020009165A1 true WO2020009165A1 (fr) 2020-01-09

Family

ID=69060852

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/JP2019/026521 WO2020009165A1 (fr) 2018-07-03 2019-07-03 Anticorps modifié et procédé pour sa production

Country Status (1)

Country Link
WO (1) WO2020009165A1 (fr)

Cited By (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2022154116A1 (fr) 2021-01-18 2022-07-21 味の素株式会社 Composé ou son sel, et anticorps produit à l'aide de celui-ci
WO2022191283A1 (fr) 2021-03-11 2022-09-15 味の素株式会社 Composé ou son sel, et anticorps ainsi obtenu
WO2022196675A1 (fr) 2021-03-16 2022-09-22 味の素株式会社 Complexe ou son sel, et procédé de fabrication associé

Citations (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
JP2003515330A (ja) * 1999-10-29 2003-05-07 ジェネンテック・インコーポレーテッド 抗前立腺幹細胞抗原(psca)抗体組成物及び使用方法
WO2009064366A2 (fr) * 2007-11-09 2009-05-22 Massachusetts Institute Of Technology Procédés et compositions pour un marquage protéique à l'aide d'acide lipoïque ligases
WO2013052581A1 (fr) * 2011-10-04 2013-04-11 Applied Nanotech Holdings, Inc. Dépôt de couches minces de matériaux par libération induite de l'extérieur à partir d'une bande de ruban
WO2017095806A1 (fr) * 2015-11-30 2017-06-08 The Regents Of The University Of California Radiofluoration spécifique de site de peptides avec de l'acide 8-[18f]fluorooctanoïque catalysée par une acide lipoïque ligase
JP2018513146A (ja) * 2015-04-15 2018-05-24 ガニメド ファーマシューティカルズ ゲーエムベーハー クローディン18.2に対する抗体を含む薬物コンジュゲート

Patent Citations (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
JP2003515330A (ja) * 1999-10-29 2003-05-07 ジェネンテック・インコーポレーテッド 抗前立腺幹細胞抗原(psca)抗体組成物及び使用方法
WO2009064366A2 (fr) * 2007-11-09 2009-05-22 Massachusetts Institute Of Technology Procédés et compositions pour un marquage protéique à l'aide d'acide lipoïque ligases
WO2013052581A1 (fr) * 2011-10-04 2013-04-11 Applied Nanotech Holdings, Inc. Dépôt de couches minces de matériaux par libération induite de l'extérieur à partir d'une bande de ruban
JP2018513146A (ja) * 2015-04-15 2018-05-24 ガニメド ファーマシューティカルズ ゲーエムベーハー クローディン18.2に対する抗体を含む薬物コンジュゲート
WO2017095806A1 (fr) * 2015-11-30 2017-06-08 The Regents Of The University Of California Radiofluoration spécifique de site de peptides avec de l'acide 8-[18f]fluorooctanoïque catalysée par une acide lipoïque ligase

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
RASHIDIAN, M. ET AL.: "Enzymatic labeling of proteins: techniques and approaches", BIOCONJUG. CHEM., vol. 24, no. 8, 2013, pages 1277 - 1294, XP055147355, ISSN: 1043-1802, DOI: 10.1021/bc400102w *

Cited By (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2022154116A1 (fr) 2021-01-18 2022-07-21 味の素株式会社 Composé ou son sel, et anticorps produit à l'aide de celui-ci
WO2022191283A1 (fr) 2021-03-11 2022-09-15 味の素株式会社 Composé ou son sel, et anticorps ainsi obtenu
KR20230154853A (ko) 2021-03-11 2023-11-09 아지노모토 가부시키가이샤 화합물 또는 이의 염, 및 이들에 의해 얻어지는 항체
WO2022196675A1 (fr) 2021-03-16 2022-09-22 味の素株式会社 Complexe ou son sel, et procédé de fabrication associé

Similar Documents

Publication Publication Date Title
JP7076152B2 (ja) IgG結合ペプチドによる抗体の特異的修飾
US11028185B2 (en) Using sortases to install click chemistry handles for protein ligation
WO2020090979A1 (fr) Composé comprenant une substance ayant une affinité pour un anticorps, site de clivage et groupe réactif ou sel correspondant
JP2022130634A (ja) 免疫リガンド/ペイロード複合体の生産方法
US11753669B2 (en) Lysine conjugated immunoglobulins
US20220088212A1 (en) C-terminal lysine conjugated immunoglobulins
WO2020009165A1 (fr) Anticorps modifié et procédé pour sa production
JP2014525904A5 (fr)
Pasupuleti et al. Genetic code expansion in E. coli enables production of a functional ‘ready-to-click’T cell receptor-specific scFv
WO2022196675A1 (fr) Complexe ou son sel, et procédé de fabrication associé

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: 19831476

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: 19831476

Country of ref document: EP

Kind code of ref document: A1

NENP Non-entry into the national phase

Ref country code: JP