WO2023219147A1 - Novel anti-ccr8 antibodies for detecting ccr8 - Google Patents

Novel anti-ccr8 antibodies for detecting ccr8 Download PDF

Info

Publication number
WO2023219147A1
WO2023219147A1 PCT/JP2023/017821 JP2023017821W WO2023219147A1 WO 2023219147 A1 WO2023219147 A1 WO 2023219147A1 JP 2023017821 W JP2023017821 W JP 2023017821W WO 2023219147 A1 WO2023219147 A1 WO 2023219147A1
Authority
WO
WIPO (PCT)
Prior art keywords
antibody
ccr8
seq
amino acid
acid sequence
Prior art date
Application number
PCT/JP2023/017821
Other languages
French (fr)
Japanese (ja)
Inventor
哲也 吉田
舞 吉川
盛男 柳樂
道也 平田
里美 東雲
あずみ 上山
礼美 松本
秀和 田中
永也 大倉
志文 坂口
尚 和田
Original Assignee
塩野義製薬株式会社
国立大学法人大阪大学
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 塩野義製薬株式会社, 国立大学法人大阪大学 filed Critical 塩野義製薬株式会社
Publication of WO2023219147A1 publication Critical patent/WO2023219147A1/en

Links

Images

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/395Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/28Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/63Introduction of foreign genetic material using vectors; Vectors; Use of hosts therefor; Regulation of expression
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/53Immunoassay; Biospecific binding assay; Materials therefor
    • G01N33/574Immunoassay; Biospecific binding assay; Materials therefor for cancer

Definitions

  • the present invention relates to a novel anti-CCR8 antibody useful for detecting CCR8.
  • Non-Patent Document 1 Powerful negative regulatory mechanisms, including immunosuppression mediated by regulatory T cells (Treg cells) within the tumor microenvironment, are a major obstacle to tumor treatment.
  • CD4-positive Treg cells infiltrating tumors can strongly inhibit anti-tumor immune responses and can be a major obstacle to effective cancer treatment.
  • Tumor immunosuppression mediated by CD4-positive FoxP3-positive Treg cells has been well documented in animal tumor models, and systemic Treg cell removal, including within the tumor, results in an antitumor effect; It has been reported that no effect is observed in removing intratumoral infiltrating Treg cells (Non-Patent Document 2).
  • Non-patent Documents 3 to 8 In humans, an increased ratio of CD4+CD25+ Treg cells (cell population containing Treg cells) in the total CD4+ T cell population has been detected in tumors of various cancer patients, including lung, breast, and ovarian tumors. It has been reported that there is a negative correlation between the abundance ratio and patient survival rate (Non-patent Documents 3 to 8).
  • CCR8 also called CY6, CKR-L1, or TER1 is a G protein-coupled seven-transmembrane CC chemokine receptor protein expressed in the thymus and spleen, and its gene is located on 3p21 in human chromosomes. exist.
  • Human CCR8 consists of 355 amino acids (Non-Patent Document 9).
  • CCL1 is known as an endogenous ligand for CCR8 (Non-Patent Document 10).
  • Human CCR8 cDNA is Genbank ACC No.
  • the mouse CCR8 cDNA is composed of the base sequence shown in NM_005201, and the mouse CCR8 cDNA is shown in Genbank ACC No. It is composed of the base sequence shown in NM_007720.
  • CCR8 is also specifically expressed in tumor-infiltrating Treg cells, and when breast cancer cells were transplanted into CCR8-deficient mice and wild-type mice, the growth and metastasis of breast cancer in CCR8-deficient mice was significantly higher than in wild-type mice. It has been shown that this was suppressed (Patent Document 1 and Non-Patent Document 11).
  • Non-Patent Documents 12 and 13 also describe that CCR8 is involved in the pathology of cancer. Furthermore, it has been disclosed that administration of anti-CCR8 antibodies to cancer model animals showed antitumor effects (Patent Documents 2 to 12).
  • Patent Documents 1, 13, and 14 describe that CCR8 is useful as a cancer diagnostic marker, but do not specifically describe anti-CCR8 antibodies useful for CCR8 detection. Further, Patent Document 15 discloses anti-CCR8 antibodies useful for CCR8 detection, but all antibodies have neutralizing activity, and anti-CCR8 antibodies that do not have neutralizing activity useful for CCR8 detection Antibodies are not specifically listed. Furthermore, Patent Document 15 does not describe that anti-CCR8 antibodies are useful for cancer diagnosis.
  • An object of the present invention is to provide an anti-CCR8 antibody useful for detecting CCR8. Another object of the present invention is to provide a diagnostic agent containing the anti-CCR8 antibody.
  • the present inventors discovered a novel anti-CCR8 antibody that specifically binds to CCR8 and does not have neutralizing activity useful for CCR8 detection. Furthermore, it has been found that the novel CCR8 antibody of the present invention does not compete with anti-CCR8 antibodies having neutralizing activity in binding to CCR8. Furthermore, it has been found that the novel CCR8 antibody of the present invention detects CCR8 expressed in tumor-infiltrating Treg cells and is useful for cancer diagnosis and companion diagnosis.
  • the present invention relates to the following.
  • CDR1 consisting of the amino acid sequence of SEQ ID NO: 2
  • a light chain variable region comprising CDR2 consisting of the amino acid sequence of SEQ ID NO: 3 and CDR3 consisting of the amino acid sequence of SEQ ID NO: 4
  • CDR1 consisting of the amino acid sequence of SEQ ID NO: 5
  • the antibody or antibody fragment thereof according to (1) which comprises a light chain variable region having the amino acid sequence of SEQ ID NO: 8 and a heavy chain variable region having the amino acid sequence of SEQ ID NO: 9.
  • the antibody or antibody fragment thereof according to (1) further having a light chain constant region having the amino acid sequence of SEQ ID NO: 10 and a heavy chain constant region having the amino acid sequence of SEQ ID NO: 11.
  • the method described in (4) for cancer diagnosis. (5-2) The method described in (4) for assisting cancer diagnosis.
  • (5-3) The method described in (4) for detecting intratumoral infiltrating Treg cells.
  • the method described in (4) for detecting CCR8-expressing tumor cells The method described in (4) for detecting CCR8-expressing tumor cells. (6) The method according to (4), wherein CCR8 contained in a sample collected from a subject to whom a medicine containing an anti-CCR8 antibody has been administered is detected. (7) The method according to (6), wherein the anti-CCR8 antibody contained in the anti-CCR8 antibody-containing medicine has neutralizing activity. (8) The anti-CCR8 antibody contained in the medicine containing the anti-CCR8 antibody contains an antibody comprising a light chain variable region having the amino acid sequence of SEQ ID NO: 8 and a heavy chain variable region having the amino acid sequence of SEQ ID NO: 9. The method according to (6), wherein there is no conflict in binding to.
  • the anti-CCR8 antibody contained in the medicine containing an anti-CCR8 antibody is a monoclonal antibody comprising a light chain variable region having the amino acid sequence of SEQ ID NO: 12 and a heavy chain variable region having the amino acid sequence of SEQ ID NO: 13, or The method according to (6), which is a fragment thereof.
  • the method according to (6), wherein the subject is a cancer patient.
  • a CCR8 detection kit comprising a composition containing the antibody or antibody fragment thereof according to any one of (1) to (3).
  • (12-2) The kit according to (11) for assisting cancer diagnosis.
  • (12-3) The kit described in (11) for detecting intratumoral infiltrating Treg cells.
  • the kit described in (11) for detecting CCR8-expressing tumor cells (13) The kit according to (11), which detects CCR8 contained in a sample collected from a subject who has been administered a medicine containing an anti-CCR8 antibody. (14) The kit according to (13), wherein the anti-CCR8 antibody contained in the anti-CCR8 antibody-containing medicine has neutralizing activity. (15) The anti-CCR8 antibody contained in the medicine containing the anti-CCR8 antibody contains an antibody comprising a light chain variable region having the amino acid sequence of SEQ ID NO: 8 and a heavy chain variable region having the amino acid sequence of SEQ ID NO: 9. The kit according to (13), which does not compete in binding to.
  • the anti-CCR8 antibody contained in the anti-CCR8 antibody-containing medicine is a monoclonal antibody comprising a light chain variable region having the amino acid sequence of SEQ ID NO: 12 and a heavy chain variable region having the amino acid sequence of SEQ ID NO: 13;
  • (18-3) Use of the antibody or antibody fragment thereof according to any one of (1) to (3) for producing a cancer diagnostic agent.
  • (19-3) Use of the antibody or antibody fragment thereof according to any one of (1) to (3) for producing a diagnostic agent for determining the suitability of administration of a medicine containing an anti-CCR8 antibody.
  • the anti-CCR8 antibody contained in the anti-CCR8 antibody-containing medicine is a monoclonal antibody comprising a light chain variable region having the amino acid sequence of SEQ ID NO: 12 and a heavy chain variable region having the amino acid sequence of SEQ ID NO: 13, or The diagnostic agent according to (19), which is a fragment thereof.
  • the anti-CCR8 antibody contained in the anti-CCR8 antibody-containing medicine is a monoclonal antibody comprising a light chain variable region having the amino acid sequence of SEQ ID NO: 12 and a heavy chain variable region having the amino acid sequence of SEQ ID NO: 13; The method according to (22), which is a fragment thereof.
  • the anti-CCR8 antibody contained in the anti-CCR8 antibody-containing medicine is used to infiltrate into tumor-infiltrating Treg cells. How to measure the amount of binding.
  • An expression vector comprising the polynucleotide described in (26).
  • novel anti-CCR8 antibody of the present invention specifically binds to CCR8, it can be used to detect CCR8 in biological samples. Furthermore, the novel anti-CCR8 antibody of the present invention detects CCR8 expressed in tumor-infiltrating Treg cells, and is therefore very useful for cancer diagnosis and companion diagnosis.
  • the competitive inhibitory activity of rat hybridoma supernatant against L263G8 antibody was evaluated by flow cytometry. If the antibody has competitive inhibitory activity against the L263G8 antibody, a peak shift will be observed compared to the negative control (Cont).
  • the X axis shows the detected fluorescence intensity, and the Y axis shows the number of detected cells.
  • the neutralizing activity of the rat hybridoma supernatant against the binding between CCL1 and CCR8 was evaluated by measuring intracellular calcium influx activity. When the antibody exhibits neutralizing activity, intracellular calcium influx activity is suppressed, and therefore the increase in fluorescence level over time is also suppressed.
  • the binding of the 3-3F antibody, L263G8 antibody, and 433H antibody to human CCR8-expressing Ramos cells was evaluated by flow cytometry.
  • the X axis shows the detected fluorescence intensity, and the Y axis shows the number of detected cells.
  • the X axis shows the detected fluorescence intensity, and the Y axis shows the number of detected cells.
  • the binding of the 3-3F antibody to cells expressing various chemokine receptors was evaluated by flow cytometry.
  • the X axis shows the detected fluorescence intensity
  • the Y axis shows the number of detected cells.
  • “3-3F” shows the peak of the 3-3F antibody
  • “Cont” shows the peak of the negative control.
  • the binding of the 3-3F antibody to intratumoral infiltrating Treg cells was evaluated by flow cytometry.
  • the X axis shows the detected fluorescence intensity
  • the Y axis shows the number of detected cells.
  • the binding of 3-3F antibody, fluorescently labeled 3-3F antibody, and L263G8 antibody to intratumoral infiltrating Treg cells was evaluated by flow cytometry.
  • the X axis shows the fluorescence intensity detected by FoxP3
  • the Y axis shows the fluorescence intensity detected by CCR8 by various anti-CCR8 antibodies.
  • the correlation between 3-3F antibody signal (Y axis) and CCR8 mRNA signal (X axis) is shown by flow cytometry analysis of intratumoral infiltrating Treg cells. Showing the correlation between the binding signal (X axis) to Treg cells of a neutralizing antibody (OP-1 antibody) against human CCR8 (OP-1 antibody) administered to human CCR8 knock-in mice and the 3-3F antibody signal (Y axis) in intratumoral infiltrating Treg cells. .
  • r indicates a correlation coefficient, and a straight line indicates linear regression.
  • antibody production techniques known in the art can be used. Examples include the method described in Immunochemistry in Practice (Blackwell Scientific Publications). Additionally, genetic engineering techniques known in the art are available. Examples include the methods described in Molecular Cloning, A Laboratory Manual, Fourth Edition, Cold Spring Harbor Laboratory Press (2012), Current Protocols Essential Laboratory Techniques, Current Protocols (2012).
  • the amino acid sequence of human CCR8 is shown in UniProtKB/Swiss-Prot:P51685 (SEQ ID NO: 1).
  • the extramembrane domain of human CCR8 corresponds to the N-terminal region consisting of amino acids 1 to 35, the loop 1 region consisting of amino acids 94 to 107, the loop 2 region consisting of amino acids 172 to 202, and the loop 3 region consisting of amino acids 264 to 280. do.
  • Position 17 in the amino acid sequence of human CCR8 is tyrosine, and position 17 is deleted in the amino acid sequence of mouse CCR8.
  • a hybridoma producing the anti-CCR8 antibody of the present invention can be produced using human CCR8 protein, a gene encoding the full-length human CCR8, human CCR8-expressing cells, etc. as an immunogen.
  • a hybridoma producing an anti-CCR8 antibody can be obtained by fusing mouse myeloma cells with spleen cells of a mouse immunized with DNA using the gene as an antigen. I can do it.
  • Examples of the basic medium for culturing hybridomas include D-MEM medium, RPMI1640 medium, IMDM medium, and ASF104 medium. Alternatively, various inorganic or organic substances can be contained.
  • the monoclonal antibody or antibody fragment thereof of the present invention includes a monoclonal antibody or a fragment of the antibody having the CDR or heavy chain variable region/light chain variable region described herein.
  • the antibody or antibody fragment may be derived from any class (e.g. IgG, IgE, IgM, IgD or IgA, preferably IgG) or subclass of immunoglobulin molecules, e.g. mouse, rat, shark, rabbit, It may be obtained from any species including pigs, hamsters, camels, llamas, goats or humans.
  • the antibody or antibody fragment is preferably a humanized monoclonal antibody or an antibody fragment of a humanized monoclonal antibody.
  • epitope refers to the region of an antigen that is bound by an antibody targeting that antigen, and when the antigen is a protein, includes specific amino acids that are in direct contact with the antibody.
  • the present invention includes not only monoclonal antibodies or antibody fragments thereof that recognize the completely same epitope as the monoclonal antibody or antibody fragment thereof of the present invention, but also monoclonal antibodies or antibody fragments thereof that recognize partially the same epitope. .
  • the CDR sequence of the monoclonal antibody of the present invention preferably has the following sequence.
  • Light chain CDR1 SEQ ID NO: 2.
  • Light chain CDR2 SEQ ID NO: 3.
  • Light chain CDR3 SEQ ID NO: 4.
  • Heavy chain CDR1 SEQ ID NO: 5.
  • Heavy chain CDR2 SEQ ID NO: 6.
  • Heavy chain CDR3 SEQ ID NO: 7.
  • antibody fragment of a monoclonal antibody refers to a fragment that is part of the monoclonal antibody of the present invention and that specifically binds to human CCR8 and selectively inhibits human CCR8 like the monoclonal antibody. means.
  • the "anti-CCR8 antibody” of the present invention includes “antibody fragment of a monoclonal antibody.”
  • Fab fragment of antigen binding
  • Fab' fragment of antigen binding
  • F(ab') 2 single chain antibody
  • scFv single chain Fv
  • Examples include disulfide stabilized antibodies (disulfide stabilized Fv; hereinafter referred to as dsFv), dimerized V region fragments (hereinafter referred to as diabody), peptides containing CDRs, etc. Pytic Patents, Vol. 6, No. 5, pp. 441-456, 1996).
  • Fab is obtained by decomposing the peptide part above the two disulfide bonds (SS bonds) that bridge the two H chains in the hinge region of IgG with the enzyme papain. It is an antibody fragment with a molecular weight of approximately 50,000 and antigen-binding activity, consisting of one half and the entire L chain.
  • Fab used in the present invention can be obtained by treating the monoclonal antibody of the present invention with papain.
  • Fab can also be produced by inserting DNA encoding the Fab of the monoclonal antibody of the present invention into a cellular expression vector, and expressing the vector by introducing the vector into cells.
  • Fab' is an antibody fragment with a molecular weight of approximately 50,000 and having antigen-binding activity obtained by cleaving the SS bond between the hinges of F(ab') 2 .
  • Fab' used in the present invention can be obtained by treating F(ab') 2 of the monoclonal antibody of the present invention with a reducing agent dithiothreitol.
  • Fab' can also be produced by inserting the DNA encoding Fab' of the monoclonal antibody of the present invention into a cellular expression vector, and expressing the vector by introducing it into E. coli, yeast, or animal cells. .
  • F(ab') 2 is obtained by decomposing the lower part of the two SS bonds in the hinge region of IgG with the enzyme pepsin, and is composed of two Fab' regions linked at the hinge region, and has a molecular weight of approximately It is an antibody fragment with an antigen binding activity of 100,000.
  • F(ab') 2 used in the present invention can be obtained by treating the monoclonal antibody of the present invention with pepsin.
  • F(ab') 2 of the monoclonal antibody of the present invention can also be expressed by inserting the DNA encoding it into an expression vector for cells and introducing the vector into E. coli, yeast, or animal cells. ) 2 can be manufactured.
  • scFv is a VH-P-VL or VL-P-VH polypeptide in which one VH and one VL are linked using an appropriate peptide linker (hereinafter referred to as P), and is capable of antigen binding. It is an active antibody fragment.
  • the VH and VL contained in the scFv used in the present invention may be those of the monoclonal antibody of the present invention.
  • the scFv used in the present invention is produced by constructing an scFv expression vector using cDNA encoding the VH and VL of the monoclonal antibody of the present invention, and expressing it by introducing it into E. coli, yeast, or animal cells. be able to.
  • dsFv refers to a polypeptide in which one amino acid residue in each of VH and VL is replaced with a cysteine residue, which are linked via an SS bond.
  • Amino acid residues to be substituted for cysteine residues can be selected based on the prediction of the three-dimensional structure of the antibody according to the method described by Reiter et al. (Protein Engineering, 7, 697, (1994)).
  • the VH or VL contained in the dsFv used in the present invention may be one of the monoclonal antibody of the present invention.
  • the dsFv used in the present invention is constructed by inserting the cDNA encoding the VH and VL of the monoclonal antibody of the present invention into an appropriate expression vector, and then transducing the expression vector into Escherichia coli, yeast, or an animal. It can be produced by introducing it into cells and expressing it.
  • Diabody is an antibody fragment in which scFv with the same or different antigen-binding specificity forms a dimer, and has bivalent antigen-binding activity for the same antigen or two types of specific antigen-binding activity for different antigens. It is.
  • a bivalent diabody that specifically reacts with the monoclonal antibody of the present invention encodes an scFv having a peptide linker of 3 to 10 residues using cDNA encoding the VH and VL of the monoclonal antibody of the present invention. It can be produced by constructing DNA, inserting the DNA into a cell expression vector, and introducing the expression vector into E. coli, yeast, or animal cells to express the diabody.
  • a peptide containing a CDR is composed of at least one region of a VH or VL CDR. Multiple CDRs can be linked directly or via a suitable peptide linker.
  • the CDR-containing peptide used in the present invention is obtained by constructing a CDR-encoding DNA using cDNA encoding the VH and VL of the monoclonal antibody of the present invention, and inserting the DNA into an expression vector for animal cells. It can be produced by introducing the vector into E. coli, yeast, or animal cells and expressing it.
  • Peptides containing CDRs can also be produced by chemical synthesis methods such as the Fmoc method (fluorenylmethyloxycarbonyl method) and the tBoc method (t-butyloxycarbonyl method).
  • the monoclonal antibody of the present invention or its antibody fragment is characterized by binding to human CCR8. In particular, those that bind specifically are preferred.
  • Specific binding can be characterized by an equilibrium dissociation constant of at least about 1 ⁇ 10 ⁇ 6 M or less (eg, a lower Kd indicates tighter binding).
  • the Kd value is preferably 1 ⁇ 10 ⁇ 7 M or less, more preferably 1 ⁇ 10 ⁇ 8 M or less, and still more preferably 1 ⁇ 10 ⁇ 9 M or less.
  • the invention includes monoclonal antibodies or antibody fragments thereof that compete with the monoclonal antibodies or antibody fragments thereof described herein for binding to human CCR8.
  • “Competing monoclonal antibody or antibody fragment thereof” refers to the monoclonal antibody of the present invention or antibody fragment thereof (preferably a light chain variable region having the amino acid sequence of SEQ ID NO: 12 and a heavy chain having the amino acid sequence of SEQ ID NO: 13). It refers to an antibody or an antibody fragment thereof that inhibits the specific binding of a monoclonal antibody containing a variable region (or a fragment thereof) to human CCR8. It is determined whether a monoclonal antibody or antibody fragment thereof competes with a monoclonal antibody or antibody fragment thereof of the invention.
  • the monoclonal antibody of the present invention Among the antibodies that were confirmed to bind to the antigen (human CCR8) in the presence of the isotype control antibody, those antibodies whose binding signal was significantly reduced in the presence of the monoclonal antibody of the present invention or its antibody fragment were selected as the monoclonal antibody of the present invention. It can be identified as an antibody that competes with the antibody or antibody fragment thereof. The reduction in binding signal is preferably 50%, more preferably 70%. Further, the Ki value of an antibody that competes with the monoclonal antibody of the present invention or its antibody fragment for binding of the monoclonal antibody of the present invention or its antibody fragment to an antigen is preferably 1 ⁇ 10 ⁇ 7 M or less, more preferably It is 1 ⁇ 10 ⁇ 8 M or less, more preferably 1 ⁇ 10 ⁇ 9 M or less.
  • the monoclonal antibody of the present invention or its antibody fragment is characterized in that it hardly inhibits the binding between CCR8 and CCL1, that is, it has almost no neutralizing activity.
  • the ability to inhibit the binding between CCR8 and CCL1 for example, in the case of human CCL1, using human CCR8-expressing 293 cells, Ca 2+ influx upon addition of human CCL1 was measured, and the signal when human CCL1 was not added was determined to be 100% inhibition rate, human The IC50 value can be determined by calculating the IC50 value, assuming that the signal obtained when CCL1 is added and when the antibody is not added is regarded as an inhibition rate of 0%. For example, it can be measured by the method described in Example 3 below. Human CCL1 was obtained from UniProtKB/Swiss-Prot No. It has an amino acid sequence such as P22362.
  • the monoclonal antibody of the present invention can be produced by a conventional method in the art using the CDRs or heavy chain variable region/light chain variable region described herein.
  • Monoclonal antibodies of the invention also include chimeric, humanized, fully human, antibody small molecule conjugates (ADCs) and bispecific antibodies. Since humanized monoclonal antibodies have reduced antigenicity in the human body, they are useful when administered to humans for therapeutic purposes.
  • a humanized monoclonal antibody is one in which the complementarity determining regions (CDRs) of a non-human mammal, such as a mouse antibody, are grafted onto the framework regions (FRs) of a human antibody. Therefore, the FRs of the humanized monoclonal antibody are of human origin.
  • a suitable FR is Kabat E. A. You can make a selection by referring to the literature by et al.
  • FRs are selected that allow CDRs to form a good antigen-binding site.
  • amino acids in the FRs of the antibody variable region may be substituted so that the CDRs of the reconstituted humanized monoclonal antibody form a suitable antigen-binding site (Sato. et al, Cancer Res. (1993) ), 53, 851).
  • the proportion of FR amino acids to be replaced is 0-15%, preferably 0-5% of the total FR region.
  • the humanized monoclonal antibody of the present invention preferably comprises: CDR1 consisting of the amino acid sequence of SEQ ID NO: 2, A light chain variable region comprising CDR2 consisting of the amino acid sequence of SEQ ID NO: 3 and CDR3 consisting of the amino acid sequence of SEQ ID NO: 4, and CDR1 consisting of the amino acid sequence of SEQ ID NO: 5, It contains a heavy chain variable region including CDR2 consisting of the amino acid sequence of SEQ ID NO: 6 and CDR3 consisting of the amino acid sequence of SEQ ID NO: 7. More preferably, It includes a light chain variable region having the amino acid sequence of SEQ ID NO: 8 and a heavy chain variable region having the amino acid sequence of SEQ ID NO: 9.
  • sequence of the constant region of the monoclonal antibody of the present invention is not particularly limited, but examples include a light chain constant region having the amino acid sequence of SEQ ID NO: 10 and a heavy chain constant region having the amino acid sequence of SEQ ID NO: 11.
  • the constant region of a human antibody is used.
  • Preferred constant regions of human antibodies include C ⁇ as a heavy chain, such as C ⁇ 1, C ⁇ 2, C ⁇ 3, and C ⁇ 4, and C ⁇ and C ⁇ as light chains. Additionally, the C region of human antibodies may be modified to improve the stability of the antibody or its production.
  • the human antibody used in humanization may be of any isotype such as IgG, IgM, IgA, IgE, IgD, etc., but it is preferable to use IgG in the present invention.
  • the monoclonal antibody of the present invention can be produced by a general production method (for example, see WO95/14041, WO96/02576, etc.).
  • the antibody gene is inserted into an expression vector so that it is expressed under the control of an expression control region, such as an enhancer/promoter.
  • a host cell can then be transformed with this expression vector to express the antibody.
  • host cells for the above transformants include vertebrate cells such as COS cells and CHO cells, prokaryotic cells, and yeast.
  • the transformant can be cultured according to methods well known to those skilled in the art, and the monoclonal antibody of the present invention is produced within or outside the transformant cells.
  • the culture medium used for the culture can be appropriately selected from various commonly used media depending on the host cell employed. For example, in the case of COS cells, RPMI-1640 medium, Dulbecco's modified Eagle's minimum essential medium (DMEM), etc.
  • DMEM Dulbecco's modified Eagle's minimum essential medium
  • a culture medium to which a serum component such as fetal bovine serum (FBS) is added, if necessary, can be used.
  • FBS fetal bovine serum
  • the culture temperature for culturing the transformant may be any temperature as long as it does not significantly reduce the intracellular protein synthesis ability, but it is preferably cultured at 32 to 42°C, most preferably at 37°C. It is preferable. Furthermore, if necessary, the culture can be carried out in air containing 1 to 10% (v/v) carbon dioxide gas.
  • the fraction containing the monoclonal antibody of the present invention produced intracellularly or extracellularly in the transformant as described above can be separated and separated by various known separation methods that utilize the physical and chemical properties of the protein.
  • Can be purified include, for example, treatment with a normal protein precipitant, ultrafiltration, molecular sieve chromatography (gel filtration), adsorption chromatography, ion exchange chromatography, affinity chromatography, high performance liquid chromatography (HPLC). ), various chromatography methods, dialysis methods, and combinations thereof can be employed.
  • a normal protein precipitant ultrafiltration, molecular sieve chromatography (gel filtration), adsorption chromatography, ion exchange chromatography, affinity chromatography, high performance liquid chromatography (HPLC).
  • HPLC high performance liquid chromatography
  • the monoclonal antibody of the present invention or its antibody fragment may be further modified with various molecules such as polyethylene glycol (PEG), radioactive substances, and toxins. Methods known in this field can be used to modify antibodies.
  • PEG polyethylene glycol
  • the monoclonal antibody of the present invention may also be fused with other proteins to its N-terminus or C-terminus (Clinical Cancer Research, 2004, 10, 1274-1281). Proteins to be fused can be appropriately selected by those skilled in the art.
  • the monoclonal antibodies of the present invention include antibodies in which N-glycoside-linked sugar chains are bound to the Fc region of the antibody. Note that fucose does not need to be bound to N-acetylglucosamine at the reducing end of the N-glycoside-linked sugar chain.
  • An example of an antibody in which an N-glycoside-linked sugar chain is bound to the Fc region of the antibody and fucose is not bound to N-acetylglucosamine at the reducing end of the N-glycoside-linked sugar chain is, for example, an ⁇ 1,6-fucosyltransferase gene. Examples include antibodies produced using CHO cells deficient in CHO cells (WO 2005/035586, WO 02/31140).
  • the antibody of the present invention in which an N-glycoside-linked sugar chain is bound to the Fc region of the antibody and fucose is not bound to N-acetylglucosamine at the reducing end of the N-glycoside-linked sugar chain, has high ADCC activity.
  • the monoclonal antibody or antibody fragment thereof of the present invention has ADCC (Antibody-dependent cell mediated cytotoxicity) activity against cells expressing CCR8 in order to remove Treg cells or macrophage cells. You may do so.
  • ADCC activity means that in vivo, an antibody that binds to a cell surface antigen such as a target cell activates the effector cell through the binding between the Fc region of the antibody and the Fc receptor present on the surface of the effector cell. Refers to activity that damages cells, etc. Effector cells include natural killer cells, activated macrophages, and the like.
  • an antibody bound to a cell surface antigen (CCR8) such as a Treg cell or a macrophage cell is activated through the binding between the Fc region of the antibody and an Fc receptor present on the surface of an effector cell. It also includes cases where effector cells are activated, Treg cells or macrophage cells are injured, and as a result, tumor cells and the like are injured.
  • CCR8 cell surface antigen
  • the monoclonal antibody of the present invention or its antibody fragment does not compete with CCR8-neutralizing antibodies for binding to CCR8.
  • a CCR8 neutralizing antibody means an antibody having neutralizing activity against CCR8. Whether or not it has neutralizing activity against CCR8 can be determined, for example, by measuring whether the physiological effect of any CCR8 ligand (eg, CCL1) on CCR8 is suppressed. Examples include, but are not limited to, measuring the binding of CCL1 to CCR8, the migration of CCR8-expressing cells by CCL1, an increase in intracellular Ca 2+ , or changes in the expression of genes sensitive to CCL1 stimulation. This can be mentioned. For example, it can be measured by the method described in Example 3 below.
  • the neutralizing activity of the "CCR8 neutralizing antibody” against the binding between CCR8 and CCL1 can be measured by adding a diluted antibody solution diluted with a medium to human CCR8-expressing 293 cells that have been pre-incorporated with a Ca 2+ indicator. can.
  • the affinity between CCR8 and CCR8 ligand is strongest for CCL1, and antibodies having high inhibitory activity against CCL1 are useful.
  • the "CCR8 neutralizing antibody” preferably has an IC50 value of neutralizing activity against the binding of CCR8 and CCL1 of 10 nM or less.
  • the neutralizing activity preferably has an IC50 value of 5 nM or less, further preferably 2 nM or less, particularly preferably 1 nM or less, and most preferably 0.5 nM or less.
  • the "CCR8 neutralizing antibody” used as the anti-CCR8 antibody contained in the anti-CCR8 antibody-containing medicine preferably has ADCC activity.
  • Examples of "neutralizing antibodies for CCR8” include antibodies described in Examples of Patent Documents 4 to 9 and 15, anti-CCR8 antibodies with product number L263G8 from BioLegend, and anti-CCR8 antibodies with product number 191704 from R&D.
  • an anti-CCR8 antibody that does not compete in binding to CCR8 with an antibody comprising a light chain variable region having the amino acid sequence of SEQ ID NO: 8 and a heavy chain variable region having the amino acid sequence of SEQ ID NO: 9 is preferable; Most preferred is an anti-CCR8 antibody comprising a light chain variable region having the sequence and a heavy chain variable region having the amino acid sequence of SEQ ID NO: 13.
  • the monoclonal antibody or antibody fragment thereof of the present invention is preferably an antibody or antibody fragment thereof that strongly recognizes human CCR8.
  • an antibody or an antibody fragment thereof that strongly recognizes human CCR8 it is possible to select an antibody or an antibody fragment thereof based on the strength of binding activity and binding selectivity for CCR8 as an index, thereby creating an antibody that more strongly recognizes human CCR8. or an antibody fragment thereof can be selected.
  • the monoclonal antibody of the present invention or its antibody fragment does not compete with the neutralizing antibody for CCR8 in binding to CCR8, detection of CCR8 is possible even in a sample to which a neutralizing antibody for CCR8 has already bound. Furthermore, depending on the CCR8-neutralizing antibody bound to the sample, the CCR8-binding activity of the monoclonal antibody of the present invention or its antibody fragment may be increased.
  • the monoclonal antibody or antibody fragment thereof of the present invention can detect CCR8 contained in a sample collected from a subject, such as a cancer patient, who has been administered a medicine containing an anti-CCR8 antibody, and It is also possible to measure the amount of anti-CCR8 antibody contained in a medicament containing a CCR8 antibody that binds to intratumoral infiltrating Treg cells.
  • the anti-CCR8 antibody contained in the anti-CCR8 antibody-containing medicine the above-mentioned "CCR8 neutralizing antibody" is preferable, the anti-CCR8 antibodies described in the Examples of Patent Documents 4 to 9 are more preferable, and the anti-CCR8 antibodies described in the Examples of Patent Documents 4 to 9 are more preferable.
  • anti-CCR8 antibodies described in the Examples, which do not compete in binding to CCR8 with antibodies comprising a light chain variable region having the amino acid sequence of SEQ ID NO: 8 and a heavy chain variable region having the amino acid sequence of SEQ ID NO: 9.
  • Anti-CCR8 antibodies are particularly preferred, and anti-CCR8 antibodies comprising a light chain variable region having the amino acid sequence of SEQ ID NO: 12 and a heavy chain variable region having the amino acid sequence of SEQ ID NO: 13 are most preferred.
  • the monoclonal antibody of the present invention or its antibody fragment specifically binds to CCR8, it can be used to detect CCR8 in samples collected from humans.
  • samples collected from humans include, but are not limited to, blood, plasma, serum, urine, organs, tissues, bone marrow, lymph nodes, and the like.
  • a labeled detection antibody When the monoclonal antibody of the present invention or an antibody fragment thereof is used as an antibody for detecting CCR8, a labeled detection antibody may be used when detecting CCR8, or a labeled secondary antibody for the detection antibody may be used. Good too.
  • substances used for labeling include fluorescent substances, microparticles, enzymes, biotin, radiolabels, magnetic particles, and the like, with fluorescent substances being preferred.
  • the fluorescent substance is not particularly limited as long as it does not impair the antigen binding property of the antibody or its antibody fragment, and includes, for example, rhodamine, coumarin, oxazine, carbopyronine, cyanine, pyromecene, naphthalene, biphenyl, Examples include anthracene, phenanthrene, pyrene, carbazole, Cy, EvoBlue, fluorescein, and derivatives thereof.
  • microparticles are not particularly limited as long as they do not impair the antigen binding properties of the antibody or antibody fragment thereof, and include, for example, metal colloid particles such as colloidal gold, colored particles such as colored latex particles, and the like.
  • the enzyme is not particularly limited as long as it does not impair the antigen binding property of the antibody or its antibody fragment, and examples include horseradish peroxidase (HRP), alkaline phosphatase, peroxidase, ⁇ -D-galactosidase, microperoxidase, and the like.
  • HRP horseradish peroxidase
  • alkaline phosphatase alkaline phosphatase
  • peroxidase peroxidase
  • ⁇ -D-galactosidase ⁇ -D-galactosidase
  • microperoxidase and the like.
  • radioactive labels examples include, but are not limited to, 3 H, 14 C, 15 N, 35 S, 90 Y, 99 Tc, 111 In, 125 I, 131 I, and the like.
  • a tag can be added to the antibody of the present invention or an antibody fragment thereof.
  • tags include FLAG, HA (hemagglutinin), GST (glutathione-S-transferase), Myc, polyhistidine (6 ⁇ His tag, etc.), and the like. These tags can be used for immobilization, purification, detection, etc. of antibodies.
  • a tagged antibody or antibody fragment is produced by adding a polynucleotide encoding a tag to a polynucleotide encoding the antibody of the present invention or an antibody fragment thereof, and then in vivo in animal cells or the like or in a cell-free manner. It can be produced by introducing it into an in vitro protein expression system such as a translation system.
  • the detection method of CCR8 of the present invention includes fluorescence immunoassay (FIA), enzyme immunoassay (ELISA or EIA), radioimmunoassay (RIA), chemiluminescence immunoassay, surface plasmon resonance, immunochromatography, etc.
  • FAA fluorescence immunoassay
  • ELISA enzyme immunoassay
  • RIA radioimmunoassay
  • chemiluminescence immunoassay chemiluminescence immunoassay
  • surface plasmon resonance immunochromatography
  • Flow cytometry, immunohistological staining, or ELISA is preferred, with flow cytometry being particularly preferred.
  • the method for detecting CCR8 of the present invention is a flow cytometry method, for example, it is a method of simultaneously measuring the expression of CCR8 and a marker specific to Treg cells, which is expressed in Treg cells contained in a sample such as intratumoral infiltrating cells. CCR8 can be detected. Furthermore, Treg cells expressing CCR8 can also be fractionated from a sample using a similar method. Examples of markers specific to Treg cells include FoxP3, CD25, and TIGIT, with FoxP3 being preferred.
  • the X axis is the detected fluorescence intensity of the Treg cell-specific marker
  • the Y axis is the detected fluorescence intensity of CCR8.
  • a plot showing Treg cells expressing Treg cells appears in the upper right area. Note that when plotting, the X-axis and Y-axis may be set in the opposite manner to the above.
  • the cell surface antigen to be gated before measuring the expression of markers specific to CCR8 and Treg cells can be selected as appropriate depending on the purpose of measurement. , for example, CD3, CD4, CD45, etc. may be selected.
  • the method for detecting CCR8 of the present invention is an immunohistological staining method
  • it can be carried out by known methods, including steps such as preparation of tissue sections such as paraffin sections, immunoreaction, washing, and color development.
  • the CCR8 detection method of the present invention is an ELISA method, it is carried out, for example, as follows. First, one antibody (capture antibody) is immobilized on the well surface of an ELISA plate. Next, after blocking is performed to prevent nonspecific adsorption to the well surface, a sample is added, and CCR8 in the sample is brought into contact with the antibody to form a complex. After removing proteins that did not bind to the antibody by washing, the other labeled antibody (detection antibody) is added to the well, brought into contact with CCR8 to form a complex, and detection and quantification are performed using the label. Quantification can be performed by a known method, such as quantifying a signal detected based on the label used using a calibration curve prepared using a standard sample of CCR8.
  • the monoclonal antibody of the present invention or its antibody fragment can be used as a diagnostic agent for cancer.
  • cancer for example, breast cancer, endometrial cancer, cervical cancer, ovarian cancer, prostate cancer, lung cancer, stomach cancer, pancreatic cancer, head and neck cancer, esophageal cancer, bladder cancer, melanoma, colorectal cancer, kidney cancer, non-Hodgkin lymphoma, urothelial cancer.
  • It can be used as a diagnostic agent for cancer, sarcoma, blood cell cancer (leukemia, lymphoma, etc.), cholangiocarcinoma, gallbladder cancer, thyroid cancer, testicular cancer, thymus cancer, liver cancer, etc., preferably breast cancer, ovarian cancer, lung cancer, etc.
  • gastric cancer pancreatic cancer, head and neck cancer, esophageal cancer, melanoma, colon cancer, renal cancer, and urothelial cancer.
  • cancer refers not only to epithelial malignant tumors such as ovarian cancer and gastric cancer, but also to non-epithelial malignant tumors including hematopoietic cancers such as chronic lymphocytic leukemia and Hodgkin's lymphoma.
  • cancer refers not only to epithelial malignant tumors such as ovarian cancer and gastric cancer, but also to non-epithelial malignant tumors including hematopoietic cancers such as chronic lymphocytic leukemia and Hodgkin's lymphoma.
  • cancer refers not only to epithelial malignant tumors such as ovarian cancer and gastric cancer, but also to non-epithelial malignant tumors including hematopoietic cancers such as chronic lymphocytic leukemia and Hodgkin's lymphoma.
  • cancer refers not only to epithelial malignant tumors such as ovarian cancer and gastric cancer, but also to non-epithelial mal
  • the monoclonal antibody of the present invention or its antibody fragment can also be used as a diagnostic agent to determine the suitability of administering a drug containing an anti-CCR8 antibody.
  • the anti-CCR8 antibody of the "medicine containing an anti-CCR8 antibody” the above-mentioned "CCR8 neutralizing antibody” is preferable, the anti-CCR8 antibodies described in the Examples of Patent Documents 4 to 9 are more preferable, and the anti-CCR8 antibodies described in the Examples of Patent Documents 4 to 9 are more preferable.
  • anti-CCR8 antibodies described in the Examples, which do not compete in binding to CCR8 with antibodies comprising a light chain variable region having the amino acid sequence of SEQ ID NO: 8 and a heavy chain variable region having the amino acid sequence of SEQ ID NO: 9.
  • Anti-CCR8 antibodies are particularly preferred, and anti-CCR8 antibodies comprising a light chain variable region having the amino acid sequence of SEQ ID NO: 12 and a heavy chain variable region having the amino acid sequence of SEQ ID NO: 13 are most preferred.
  • “Pharmaceuticals containing anti-CCR8 antibodies” are useful as anticancer agents, such as breast cancer, endometrial cancer, cervical cancer, ovarian cancer, prostate cancer, lung cancer, stomach cancer, pancreatic cancer, head and neck cancer, esophageal cancer, and bladder cancer. , melanoma, colorectal cancer, kidney cancer, non-Hodgkin's lymphoma, urothelial cancer, sarcoma, blood cell cancer (leukemia, lymphoma, etc.), bile duct cancer, gallbladder cancer, thyroid cancer, testicular cancer, thymus cancer, liver cancer, etc.
  • it is very useful as a medicine for treating and/or preventing breast cancer, ovarian cancer, lung cancer, gastric cancer, pancreatic cancer, head and neck cancer, esophageal cancer, melanoma, colon cancer, kidney cancer, and urothelial cancer.
  • a kit containing the monoclonal antibody of the present invention can be used as a CCR8 detection kit.
  • the kit contains the monoclonal antibody of the present invention or an antibody fragment thereof, and further includes a gating antibody, a labeled secondary antibody, constituent reagents (diluting solution, washing solution, reaction stop solution, etc.), a fluorescence detection reagent, a CCR8 standard product, and usage. It may also include instructions, etc.
  • the pH buffer component contained in the constituent reagents is not particularly limited, but includes, for example, acids such as phosphoric acid, boric acid, acetic acid, citric acid, formic acid, and cacodylic acid, or salts thereof; amino acids such as glycine; trishydroxyaminomethane; (Tris), HEPES, MES, and other good buffers.
  • the surfactant contained in the constituent reagents is not particularly limited, but nonionic surfactants are preferred, and any of the ester ether type, ester type, and ether type can be used. More specifically, Tween20, Tween40, Tween80, Triton-X100, polyoxyethylene (60) sorbitan monostearate, polyoxyethylene (65) sorbitan tristearate, polyoxyethylene (80) sorbitan monooleate, polyoxy Examples include ethylene alkylphenyl ether, Nonidet P-40, CHAPS, and the like. These surfactants may be used alone or in combination of two or more.
  • the present invention includes polynucleotides encoding the light chain variable region or heavy chain variable region of the monoclonal antibody of the present invention.
  • the invention further includes expression vectors containing the polynucleotides.
  • the polynucleotide is not particularly limited as long as it encodes the light chain variable region or heavy chain variable region of the monoclonal antibody of the present invention, and may be a polymer consisting of multiple nucleotides such as deoxyribonucleic acid (DNA) or ribonucleic acid (RNA). It is. It may contain non-natural bases.
  • the polynucleotide of the present invention can be used to produce antibodies by genetic engineering techniques. It can also be used as a probe to screen for antibodies having functions equivalent to the monoclonal antibodies of the present invention.
  • a polynucleotide encoding the monoclonal antibody of the present invention, or a portion thereof as a probe hybridize with the polynucleotide under stringent conditions by a technique such as hybridization or gene amplification technique (e.g. PCR), Moreover, DNA encoding an antibody having an activity equivalent to that of the monoclonal antibody of the present invention can be obtained. Such DNA is also included in the polynucleotide of the present invention.
  • Hybridization techniques (Sambrook, J et al., Molecular Cloning 2nd ed., 9.47-9.58, Cold Spring Harbor Lab. press, 1989) are well known to those skilled in the art.
  • Examples of hybridization conditions include low stringency conditions. Low stringency conditions include, for example, 42°C, 0.1x SSC, 0.1% SDS in washing after hybridization, preferably 50°C, 0.1x SSC, 0.1%. This is a condition of SDS. More preferable hybridization conditions include highly stringent conditions. High stringency conditions are, for example, 65° C., 5 ⁇ SSC, and 0.1% SDS. Under these conditions, it can be expected that polynucleotides with higher homology can be obtained more efficiently as the temperature is raised. However, multiple factors such as temperature and salt concentration can be considered as factors that affect the stringency of hybridization, and those skilled in the art will be able to achieve similar stringency by appropriately selecting these factors. .
  • Antibodies that are functionally equivalent to the monoclonal antibodies of the present invention and encoded by polynucleotides obtained by these hybridization techniques or gene amplification techniques usually have high homology with these antibodies in their amino acid sequences.
  • the monoclonal antibodies of the present invention also include antibodies that are functionally equivalent to the monoclonal antibodies of the present invention and have high homology with the amino acid sequence of the antibodies.
  • High homology generally refers to at least 75% identity, preferably 85% or more identity, and more preferably 95% or more identity at the amino acid level.
  • the monoclonal antibody of the present invention or an antibody fragment thereof may have an effect of removing intratumoral infiltrating Treg cells or a tumor cell. Whether or not the monoclonal antibody of the present invention has an effect of removing tumor-infiltrating Treg cells can be determined, for example, by the method described in Examples of Patent Document 2.
  • the monoclonal antibody or antibody fragment thereof of the present invention is also useful in pharmaceutical compositions.
  • Pharmaceutical compositions containing the monoclonal antibodies of the present invention or antibody fragments thereof can be administered orally or parenterally, systemically or locally.
  • parenteral administration for example, intravenous injection such as drip, intramuscular injection, intraperitoneal injection, subcutaneous injection, intranasal administration, inhalation, etc. can be selected.
  • a pharmaceutical composition containing the monoclonal antibody or antibody fragment thereof of the present invention is very useful as a medicament for the treatment and/or prevention of CCR8-related diseases.
  • it is very useful as a medicament for treating and/or preventing cancer in which CCR8-expressing Treg cells infiltrate into tumors.
  • cancer for example, breast cancer, endometrial cancer, cervical cancer, ovarian cancer, prostate cancer, lung cancer, stomach cancer, pancreatic cancer, head and neck cancer, esophageal cancer, bladder cancer, melanoma, colorectal cancer, kidney cancer, non-Hodgkin lymphoma, urothelial cancer.
  • Cancer sarcoma, blood cell cancer (leukemia, lymphoma, etc.), bile duct cancer, gallbladder cancer, thyroid cancer, testicular cancer, thymus cancer, liver cancer, etc., preferably breast cancer, ovarian cancer, lung cancer, stomach cancer, pancreatic cancer, head and neck cancer. It is very useful as a medicine for treating and/or preventing cancer, esophageal cancer, melanoma, colon cancer, renal cancer, and urothelial cancer.
  • Example 1 Preparation of rat hybridoma producing anti-human CCR8 antibody (1) Preparation of hybridoma Human CCR8-expressing Rat-1 cells, which are the immunogen, were prepared using an expression vector in which the human CCR8 gene was cloned into pQCXIP (Clontech). The cells were transfected, followed by drug selection with puromycin (1 ⁇ g/ml) for one month. Rats were immunized once with human CCR8-expressing Rat-1 cells, and about two weeks later, lymph nodes were collected and hybridomas were produced by a conventional method.
  • DAPI 4,6-diamidino-2-phenylindole
  • the results are shown in Table 1. No. 30, No. 3.No. 12, No. The 37 hybridoma supernatants showed the strongest binding to human CCR8. In addition, binding to parent cells was No. 3 has the lowest coupling, and the S/N ratio (signal to noise ratio) is No. 3 was the highest. Next, competitive inhibition against anti-human CCR8 antibody (L263G8, BioLegend) was evaluated. The above four hybridoma supernatants or the hybridoma supernatant (No.
  • DAPI was added at a concentration of 100 ng/ml, and the fluorescence value (average value) bound to the cells was quantified by flow cytometry.
  • the inhibition rate (%) was calculated from the average fluorescence value of each test antibody, with the average fluorescence value when GIT medium was added as a negative control being 100% and the average fluorescence value of the positive control being 0%.
  • the results are shown in Table 1 and FIG. No.
  • the hybridoma supernatant of No. 2 exhibited competitive inhibitory activity against the L263G8 antibody, whereas the four hybridoma supernatants obtained in (1) above showed almost no competitive inhibitory activity against the L263G8 antibody.
  • GIT medium was used as a negative control.
  • hybridoma producing an anti-CCR8 antibody with neutralizing activity the supernatant of a hybridoma (No. 2) producing the 2-7B antibody described in the Examples of WO2020/138489 was used.
  • human CCL1 BioLegend
  • HITACHI F-7000 spectrofluorometer
  • No. 1 is the best hybridoma that produces anti-CCR8 antibodies without neutralizing activity. Three clones were selected.
  • Example 2 Determination of antibody sequence No. Regarding the 3-3F antibody produced by the three clones, the amino acid sequences of the light chain variable region and heavy chain variable region of the antibody were determined from hybridoma cells according to a conventional method (Table 3).
  • Example 3 Evaluation of neutralizing activity of purified antibody
  • the established hybridoma was cultured in a serum-free medium, and the culture supernatant was purified by Protein G affinity and gel filtration to obtain a purified antibody.
  • the neutralizing activity of this purified 3-3F antibody was measured by the method described below.
  • L263G8 antibody which is a commercially available anti-human CCR8 antibody, and 2C7 antibody, 10A11 antibody, and 2-7B antibody described in the Examples of WO2020/138489 were used. Using.
  • a diluted antibody solution diluted with a medium was added to human CCR8-expressing 293 cells into which a Ca 2+ indicator had been incorporated in advance, and Ca 2+ influx due to the addition of 200 nM human CCL1 (manufactured by Biolegend) was measured by FLIPR.
  • the inhibition rate was calculated by setting the signal when human CCL1 was not added as an inhibition rate of 100%, and the signal when human CCL1 was added and the antibody was not added as an inhibition rate of 0%, and the antibody concentration showing a 50% inhibition rate was defined as IC50. Evaluations were performed at least three times and IC50 was expressed as Average ⁇ SD. As a result, no neutralizing activity was observed in the purified 3-3F antibody (Table 4).
  • Example 4 Binding analysis to CCR8 (1) Evaluation of binding activity to CCR8 Binding of 3-3F antibody to human CCR8-expressing Ramos cells and its parent strain (Ramos cells), and OP-1 antibody (neutralizing antibody to human CCR8) In the presence of the humanized 10A11 antibody (light chain variable region: IGKV4-1 N53Q+G29R (SEQ ID NO: 12)/heavy chain variable region: IGHV3-15 T94R (SEQ ID NO: 13)) described in the Examples of WO2020/138489. The binding of the 3-3F antibody was evaluated. In this example, a non-fluorescently labeled 3-3F antibody and a fluorescently labeled Alexa647 antibody were used.
  • APC fluorescently labeled anti-rat IgG antibody (manufactured by Jackson ImmunoResearch Laboratories) was used as a secondary antibody to detect the non-fluorescently labeled substance.
  • APC fluorescently labeled L263G8 antibody and BV421 fluorescently labeled 433H antibody (manufactured by BD Biosciences), which are commercially available anti-human CCR8 antibodies, were used.
  • Rat IgG2a antibody (unlabeled, 100-fold diluted) was used as the isotype antibody. Thereafter, after washing with culture medium (10% FBS), APC fluorescently labeled anti-rat IgG antibody was added and reacted at 4°C for 30 minutes.
  • APC fluorescently labeled L263G8 antibody (100-fold dilution) was added and reacted at 4°C for 30 minutes.
  • APC-mouse IgG2a antibody (100-fold dilution) was used as the isotype antibody.
  • C BV421 fluorescently labeled 433H antibody (100 times diluted) was added and reacted at 4° C. for 30 minutes.
  • BV421-mouse IgG2a antibody (100 times diluted) was used as the isotype antibody.
  • the cells were washed with culture solution (10% FBS), Cellstain-DAPI solution (10,000-fold dilution, manufactured by Dojindo Laboratories) was added, and the cells were measured by flow cytometry.
  • eBioscience Fixable Viability Dye eFluor 780 solution (10,000-fold dilution, manufactured by Thermo Fisher Scientific) was added, reacted at 4°C for 10 minutes, and then washed with culture medium (10% FBS). , resuspended in culture medium (10% FBS) and measured by flow cytometry.
  • Human CCR8 cDNA (SEQ ID NO: 17) was cloned into the EcoRI/BamHI site of the pQCXIP vector to create a pQCXIP-hCCR8 expression vector. 5 ⁇ g of this expression vector was transformed into HEK293T cells (TAKARA) using lipofectamine 3000, and after 2 days drug selection was performed using 2 ⁇ g/ml of puromycin. Human CCR8 expressed on the cell surface of surviving drug-resistant HEK293T cells cultured in the presence of drugs for 3 weeks at 37°C and 5% CO was detected using a flow cytometer using a PE-labeled anti-CCR8 antibody (BioLegend).
  • Example 5 Evaluation of binding of 3-3F antibody to human tumor-infiltrating cells
  • the binding of 3-3F antibody to human tumor-infiltrating cells was evaluated using ovarian cancer patient specimens.
  • a non-fluorescently labeled 3-3F antibody and a fluorescently labeled Alexa647 antibody were used.
  • Alexa647 fluorescently labeled anti-rat IgG antibody (manufactured by Jackson ImmunoResearch Laboratories) was used as a secondary antibody to detect the unlabeled fluorescent substance.
  • an APC fluorescent label of L263G8 antibody which is a commercially available anti-human CCR8 antibody, was used as a positive control.
  • Intratumoral infiltrating cells extracted from ovarian cancer were seeded into a 96-well plate at a rate of 1.5 x 10 5 cells, and Zombie NIR Fixable Viability Kit (manufactured by Biolegend) was added and reacted at room temperature for 15 minutes, followed by Example 4.
  • OP-1 antibody which is a neutralizing antibody against human CCR8 used in , was added at various concentrations and allowed to react at 4°C for 60 minutes.
  • the positive rate of the FoxP3-positive CCR8-positive fraction was 46.6%.
  • the reactivity of each antibody in the presence of OP-1 antibody was also confirmed, no decrease in binding was observed for 3-3F antibody in the presence of OP-1 antibody.
  • the L236G8 antibody used as a comparison control a decrease in binding was observed in the presence of the OP-1 antibody, and the positivity rate of the FoxP3-positive CCR8-positive fraction was reduced to about half.
  • Example 6 Confirmation of CCR8-specific binding of 3-3F antibody to intratumoral infiltrating Treg cells Simultaneous detection of signal by 3-3F antibody and CCR8 mRNA expression on flow cytometry using PrimeFlow RNA assay technology The CCR8 specificity of the 3-3F antibody was confirmed.
  • Tumor tissues derived from ovarian cancer patients were dispersed using Tumor Dissociation Kit, human (Miltenyi Biotec) and cryopreserved. Frozen tumor dispersed cells were thawed and suspended in Stain Buffer (BD Bioscience) supplemented with Human TruStain FcX (BioLegend) and Zombie NIR Fixable Viability Kit (BioLegend). , and left at 4° C.
  • Example 7 Evaluation of binding of 3-3F antibody to mouse tumor-infiltrating cells
  • the binding of 3-3F antibody to mouse tumor-infiltrating cells was evaluated using a mouse kidney cancer lung metastasis model.
  • a BB515 fluorescently labeled 3-3F antibody was used.
  • a portion of this mouse model was prepared using the neutralizing antibody against human CCR8 used in Example 4.
  • OP-1 antibody which is an anti-human IgG Fc antibody
  • the BV421 fluorescent label of M1310G05 antibody which is an anti-human IgG Fc antibody
  • a BV421 fluorescent label of 433H antibody which is a commercially available anti-human CCR8 antibody, was used, and samples separated from the same sample were stained and measured. Renca, a mouse kidney cancer cell cultured in a knock-in mouse (female, 21 weeks old) in which CCR8 of Balb/C mouse described in Example 10 of WO2020/138489 was replaced with human CCR8 gene, was cultured at 4 ⁇ 10.
  • mice Five were injected into the tail vein. Twelve days later, OP-1 antibody was injected through the tail vein. The next day, the lungs to which tumor cells had metastasized were collected from the 0.05 mg/kg administration group (8 mice), the 0.5 mg/kg administration group (6 mice), and the untreated mice (9 mice) under inhalation anesthesia. Single cells were dispersed and purified from lung tissue using a Tumor Dissociation Kit, mouse (Miltenyi Biotec), and BD Pharma Lyse (BD Biosciences), and stained for flow cytometry.
  • Treg cells eFluor 780, CD8 negative, and TCR ⁇ , CD4, CD25, Foxp3 positive fraction for sample 1, eFluor 780, CD8 negative, and CD45, TCR fraction for sample 2) were analyzed using MACSQuantAnalyzer (Miltenyi Biotec).
  • the monoclonal antibody or antibody fragment thereof of the present invention can be used to detect CCR8 in a biological sample. Furthermore, a pharmaceutical composition containing the monoclonal antibody of the present invention or an antibody fragment thereof is very useful as a medicament for treating or preventing CCR8-related diseases.

Abstract

Provided are novel anti-CCR8 antibodies. Said antibodies detect CCR8 expressed in tumor invasive Treg cells or the like and are useful for cancer diagnosis or companion diagnostics.

Description

CCR8検出用新規抗CCR8抗体Novel anti-CCR8 antibody for CCR8 detection
 本発明は、CCR8の検出に有用な新規抗CCR8抗体に関する。 The present invention relates to a novel anti-CCR8 antibody useful for detecting CCR8.
 腫瘍微小環境内の制御性T細胞(Treg細胞)が媒介する免疫抑制をはじめとする強力な負の調節機構は、腫瘍の治療にとって大きな障害である(非特許文献1)。
 例えば、腫瘍に浸潤するCD4陽性Treg細胞は、抗腫瘍免疫応答を強力に阻害している可能性があり、効果的な癌治療の大きな障害となりうる。
 CD4陽性FoxP3陽性Treg細胞が媒介する腫瘍免疫抑制は、動物腫瘍モデルで十分に立証されており、腫瘍内も含めた全身性のTreg細胞除去により抗腫瘍効果が得られるが、一方で50%程度の腫瘍内浸潤Treg細胞除去では効果が認められないことが報告されている(非特許文献2)。
Powerful negative regulatory mechanisms, including immunosuppression mediated by regulatory T cells (Treg cells) within the tumor microenvironment, are a major obstacle to tumor treatment (Non-Patent Document 1).
For example, CD4-positive Treg cells infiltrating tumors can strongly inhibit anti-tumor immune responses and can be a major obstacle to effective cancer treatment.
Tumor immunosuppression mediated by CD4-positive FoxP3-positive Treg cells has been well documented in animal tumor models, and systemic Treg cell removal, including within the tumor, results in an antitumor effect; It has been reported that no effect is observed in removing intratumoral infiltrating Treg cells (Non-Patent Document 2).
 ヒトにおいて全CD4陽性T細胞集団中のCD4陽性CD25陽性Treg細胞比(Treg細胞を含む細胞集団)の増大が、肺、乳房及び卵巣腫瘍をはじめとする、種々の癌患者の腫瘍内で検出され、存在比と患者生存率に負の相関があることが報告されている(非特許文献3~8)。 In humans, an increased ratio of CD4+CD25+ Treg cells (cell population containing Treg cells) in the total CD4+ T cell population has been detected in tumors of various cancer patients, including lung, breast, and ovarian tumors. It has been reported that there is a negative correlation between the abundance ratio and patient survival rate (Non-patent Documents 3 to 8).
 CCR8はCY6、CKR-L1又はTER1とも呼ばれていた胸腺や脾臓等で発現しているG蛋白共役型7回膜貫通型のCCケモカイン受容体タンパク質であり、その遺伝子はヒトクロモゾームでは3p21に存在する。ヒトCCR8は355アミノ酸からなる(非特許文献9)。CCR8に対する内因性リガンドとしてはCCL1が知られている(非特許文献10)。ヒトCCR8 cDNAはGenbank ACC No.NM_005201に示される塩基配列で構成され、マウスCCR8 cDNAはGenbank ACC No.NM_007720に示される塩基配列で構成される。 CCR8, also called CY6, CKR-L1, or TER1, is a G protein-coupled seven-transmembrane CC chemokine receptor protein expressed in the thymus and spleen, and its gene is located on 3p21 in human chromosomes. exist. Human CCR8 consists of 355 amino acids (Non-Patent Document 9). CCL1 is known as an endogenous ligand for CCR8 (Non-Patent Document 10). Human CCR8 cDNA is Genbank ACC No. The mouse CCR8 cDNA is composed of the base sequence shown in NM_005201, and the mouse CCR8 cDNA is shown in Genbank ACC No. It is composed of the base sequence shown in NM_007720.
 CCR8は、腫瘍内浸潤Treg細胞にも特異的に発現しており、CCR8欠損マウスと野生型マウスに乳癌細胞を移植したところ、CCR8欠損マウスにおける乳癌の増殖と転移が野生型マウスと比較して抑制されていたことが示されている(特許文献1及び非特許文献11)。非特許文献12~13にも、CCR8が癌の病態に関与していることが記載されている。さらに、抗CCR8抗体を癌モデル動物に投与することにより抗腫瘍効果を示したことが開示されている(特許文献2~12)。 CCR8 is also specifically expressed in tumor-infiltrating Treg cells, and when breast cancer cells were transplanted into CCR8-deficient mice and wild-type mice, the growth and metastasis of breast cancer in CCR8-deficient mice was significantly higher than in wild-type mice. It has been shown that this was suppressed (Patent Document 1 and Non-Patent Document 11). Non-Patent Documents 12 and 13 also describe that CCR8 is involved in the pathology of cancer. Furthermore, it has been disclosed that administration of anti-CCR8 antibodies to cancer model animals showed antitumor effects (Patent Documents 2 to 12).
 特許文献1、13及び14には、CCR8が癌診断マーカーとして有用であることが記載されているが、CCR8検出に有用な抗CCR8抗体は具体的に記載されていない。また、特許文献15には、CCR8検出に有用な抗CCR8抗体が開示されているが、いずれの抗体も中和活性を有しており、CCR8検出に有用な中和活性を有さない抗CCR8抗体は具体的に記載されていない。さらに、特許文献15には、抗CCR8抗体が癌診断に有用であることも記載されていない。 Patent Documents 1, 13, and 14 describe that CCR8 is useful as a cancer diagnostic marker, but do not specifically describe anti-CCR8 antibodies useful for CCR8 detection. Further, Patent Document 15 discloses anti-CCR8 antibodies useful for CCR8 detection, but all antibodies have neutralizing activity, and anti-CCR8 antibodies that do not have neutralizing activity useful for CCR8 detection Antibodies are not specifically listed. Furthermore, Patent Document 15 does not describe that anti-CCR8 antibodies are useful for cancer diagnosis.
米国特許第10087259号明細書US Patent No. 10087259 国際公開第2018/181425号International Publication No. 2018/181425 国際公開第2018/112032号International Publication No. 2018/112032 国際公開第2020/138489号International Publication No. 2020/138489 国際公開第2021/142002号International Publication No. 2021/142002 国際公開第2021/152186号International Publication No. 2021/152186 国際公開第2021/163064号International Publication No. 2021/163064 国際公開第2021/178749号International Publication No. 2021/178749 国際公開第2021/194942号International Publication No. 2021/194942 国際公開第2021/260208号International Publication No. 2021/260208 国際公開第2021/260209号International Publication No. 2021/260209 国際公開第2021/260210号International Publication No. 2021/260210 国際公開第03/096020号International Publication No. 03/096020 国際公開第2017/198631号International Publication No. 2017/198631 国際公開第2007/044756号International Publication No. 2007/044756
 本発明の目的は、CCR8の検出に有用な抗CCR8抗体を提供することである。さらには、該抗CCR8抗体を含有する診断薬を提供することである。 An object of the present invention is to provide an anti-CCR8 antibody useful for detecting CCR8. Another object of the present invention is to provide a diagnostic agent containing the anti-CCR8 antibody.
 本発明者らは、鋭意研究の結果、CCR8に特異的に結合し、CCR8検出に有用な中和活性を有さない新規抗CCR8抗体を見出した。また、本発明の新規CCR8抗体は、CCR8に対する結合が中和活性を有する抗CCR8抗体とは競合しないことを見出した。さらに、本発明の新規CCR8抗体は、腫瘍内浸潤Treg細胞等に発現するCCR8を検出し、癌診断やコンパニオン診断に有用であることを見出した。 As a result of extensive research, the present inventors discovered a novel anti-CCR8 antibody that specifically binds to CCR8 and does not have neutralizing activity useful for CCR8 detection. Furthermore, it has been found that the novel CCR8 antibody of the present invention does not compete with anti-CCR8 antibodies having neutralizing activity in binding to CCR8. Furthermore, it has been found that the novel CCR8 antibody of the present invention detects CCR8 expressed in tumor-infiltrating Treg cells and is useful for cancer diagnosis and companion diagnosis.
 本発明は、以下に関する。
(1)配列番号2のアミノ酸配列からなるCDR1、
配列番号3のアミノ酸配列からなるCDR2及び
配列番号4のアミノ酸配列からなるCDR3を含む軽鎖可変領域並びに
配列番号5のアミノ酸配列からなるCDR1、
配列番号6のアミノ酸配列からなるCDR2及び
配列番号7のアミノ酸配列からなるCDR3を含む重鎖可変領域
を含む、CCR8に結合するモノクローナル抗体又はその抗体断片。
(2)配列番号8のアミノ酸配列を有する軽鎖可変領域、及び、配列番号9のアミノ酸配列を有する重鎖可変領域を含む、(1)記載の抗体又はその抗体断片。
(3)さらに、配列番号10のアミノ酸配列を有する軽鎖定常領域、及び、配列番号11のアミノ酸配列を有する重鎖定常領域を有する、(1)記載の抗体又はその抗体断片。
(4)(1)~(3)のいずれかに記載の抗体又はその抗体断片を含有する組成物を使用する、CCR8の検出方法。
(5)癌診断のための、(4)記載の方法。
(5-2)癌診断を補助するための、(4)記載の方法。
(5-3)腫瘍内浸潤Treg細胞を検出するための、(4)記載の方法。
(5-4)CCR8発現腫瘍細胞を検出するための、(4)記載の方法。
(6)抗CCR8抗体を含有する医薬を投与した被験者から採取した試料に含まれるCCR8を検出する、(4)記載の方法。
(7)抗CCR8抗体を含有する医薬に含まれる抗CCR8抗体が中和活性を有する、(6)記載の方法。
(8)抗CCR8抗体を含有する医薬に含まれる抗CCR8抗体が、配列番号8のアミノ酸配列を有する軽鎖可変領域、及び、配列番号9のアミノ酸配列を有する重鎖可変領域を含む抗体とCCR8に対する結合が競合しない、(6)記載の方法。
(9)抗CCR8抗体を含有する医薬に含まれる抗CCR8抗体が、配列番号12のアミノ酸配列を有する軽鎖可変領域、及び、配列番号13のアミノ酸配列を有する重鎖可変領域を含むモノクローナル抗体又はその断片である、(6)記載の方法。
(10)被験者が癌の患者である、(6)記載の方法。
(11)(1)~(3)のいずれかに記載の抗体又はその抗体断片を含有する組成物を含む、CCR8の検出キット。
(12)癌診断のための、(11)記載のキット。
(12-2)癌診断を補助するための、(11)記載のキット。
(12-3)腫瘍内浸潤Treg細胞を検出するための、(11)記載のキット。
(12-4)CCR8発現腫瘍細胞を検出するための、(11)記載のキット。
(13)抗CCR8抗体を含有する医薬を投与した被験者から採取した試料に含まれるCCR8を検出する、(11)記載のキット。
(14)抗CCR8抗体を含有する医薬に含まれる抗CCR8抗体が中和活性を有する、(13)記載のキット。
(15)抗CCR8抗体を含有する医薬に含まれる抗CCR8抗体が、配列番号8のアミノ酸配列を有する軽鎖可変領域、及び、配列番号9のアミノ酸配列を有する重鎖可変領域を含む抗体とCCR8に対する結合が競合しない、(13)記載のキット。
(16)抗CCR8抗体を含有する医薬に含まれる抗CCR8抗体が、配列番号12のアミノ酸配列を有する軽鎖可変領域、及び、配列番号13のアミノ酸配列を有する重鎖可変領域を含むモノクローナル抗体又はその断片である、(13)記載のキット。
(17)被験者が癌の患者である、(13)記載のキット。
(18)(1)~(3)のいずれかに記載の抗体又はその抗体断片を含有する、癌の診断薬。
(18-2)癌診断のための、(1)~(3)のいずれかに記載の抗体又はその抗体断片。
(18-3)癌の診断薬を製造するための、(1)~(3)のいずれかに記載の抗体又はその抗体断片の使用。
(19)(1)~(3)のいずれかに記載の抗体又はその抗体断片を含有する、抗CCR8抗体を含有する医薬の投与の適否を判断するための診断薬。
(19-2)抗CCR8抗体を含有する医薬の投与の適否を判断するための、(1)~(3)のいずれかに記載の抗体又はその抗体断片。
(19-3)、抗CCR8抗体を含有する医薬の投与の適否を判断する診断薬を製造するための、(1)~(3)のいずれかに記載の抗体又はその抗体断片の使用。
(20)抗CCR8抗体を含有する医薬が抗癌剤である、(19)記載の診断薬。
(21)抗CCR8抗体を含有する医薬に含まれる抗CCR8抗体が、配列番号12のアミノ酸配列を有する軽鎖可変領域、及び、配列番号13のアミノ酸配列を有する重鎖可変領域を含むモノクローナル抗体又はその断片である、(19)記載の診断薬。
(22)(1)~(3)のいずれかに記載の抗体又はその抗体断片を含有する組成物を使用する、抗CCR8抗体を含有する医薬の投与の適否を判断する方法。
(23)抗CCR8抗体を含有する医薬が抗癌剤である、(22)記載の方法。
(24)抗CCR8抗体を含有する医薬に含まれる抗CCR8抗体が、配列番号12のアミノ酸配列を有する軽鎖可変領域、及び、配列番号13のアミノ酸配列を有する重鎖可変領域を含むモノクローナル抗体又はその断片である、(22)記載の方法。
(25)(1)~(3)のいずれかに記載の抗体又はその抗体断片を含有する組成物を使用する、抗CCR8抗体を含有する医薬に含まれる抗CCR8抗体の腫瘍内浸潤Treg細胞への結合量を測定する方法。
(26)(1)又は(2)記載の抗体の軽鎖可変領域又は重鎖可変領域をコードするポリヌクレオチド。
(27)(26)記載のポリヌクレオチドを含む発現ベクター。
The present invention relates to the following.
(1) CDR1 consisting of the amino acid sequence of SEQ ID NO: 2,
A light chain variable region comprising CDR2 consisting of the amino acid sequence of SEQ ID NO: 3 and CDR3 consisting of the amino acid sequence of SEQ ID NO: 4, and CDR1 consisting of the amino acid sequence of SEQ ID NO: 5,
A monoclonal antibody or antibody fragment thereof that binds to CCR8 and includes a heavy chain variable region including CDR2 consisting of the amino acid sequence of SEQ ID NO: 6 and CDR3 consisting of the amino acid sequence of SEQ ID NO: 7.
(2) The antibody or antibody fragment thereof according to (1), which comprises a light chain variable region having the amino acid sequence of SEQ ID NO: 8 and a heavy chain variable region having the amino acid sequence of SEQ ID NO: 9.
(3) The antibody or antibody fragment thereof according to (1), further having a light chain constant region having the amino acid sequence of SEQ ID NO: 10 and a heavy chain constant region having the amino acid sequence of SEQ ID NO: 11.
(4) A method for detecting CCR8 using a composition containing the antibody or antibody fragment thereof according to any one of (1) to (3).
(5) The method described in (4) for cancer diagnosis.
(5-2) The method described in (4) for assisting cancer diagnosis.
(5-3) The method described in (4) for detecting intratumoral infiltrating Treg cells.
(5-4) The method described in (4) for detecting CCR8-expressing tumor cells.
(6) The method according to (4), wherein CCR8 contained in a sample collected from a subject to whom a medicine containing an anti-CCR8 antibody has been administered is detected.
(7) The method according to (6), wherein the anti-CCR8 antibody contained in the anti-CCR8 antibody-containing medicine has neutralizing activity.
(8) The anti-CCR8 antibody contained in the medicine containing the anti-CCR8 antibody contains an antibody comprising a light chain variable region having the amino acid sequence of SEQ ID NO: 8 and a heavy chain variable region having the amino acid sequence of SEQ ID NO: 9. The method according to (6), wherein there is no conflict in binding to.
(9) The anti-CCR8 antibody contained in the medicine containing an anti-CCR8 antibody is a monoclonal antibody comprising a light chain variable region having the amino acid sequence of SEQ ID NO: 12 and a heavy chain variable region having the amino acid sequence of SEQ ID NO: 13, or The method according to (6), which is a fragment thereof.
(10) The method according to (6), wherein the subject is a cancer patient.
(11) A CCR8 detection kit comprising a composition containing the antibody or antibody fragment thereof according to any one of (1) to (3).
(12) The kit according to (11) for cancer diagnosis.
(12-2) The kit according to (11) for assisting cancer diagnosis.
(12-3) The kit described in (11) for detecting intratumoral infiltrating Treg cells.
(12-4) The kit described in (11) for detecting CCR8-expressing tumor cells.
(13) The kit according to (11), which detects CCR8 contained in a sample collected from a subject who has been administered a medicine containing an anti-CCR8 antibody.
(14) The kit according to (13), wherein the anti-CCR8 antibody contained in the anti-CCR8 antibody-containing medicine has neutralizing activity.
(15) The anti-CCR8 antibody contained in the medicine containing the anti-CCR8 antibody contains an antibody comprising a light chain variable region having the amino acid sequence of SEQ ID NO: 8 and a heavy chain variable region having the amino acid sequence of SEQ ID NO: 9. The kit according to (13), which does not compete in binding to.
(16) The anti-CCR8 antibody contained in the anti-CCR8 antibody-containing medicine is a monoclonal antibody comprising a light chain variable region having the amino acid sequence of SEQ ID NO: 12 and a heavy chain variable region having the amino acid sequence of SEQ ID NO: 13; The kit according to (13), which is a fragment thereof.
(17) The kit according to (13), wherein the subject is a cancer patient.
(18) A cancer diagnostic agent containing the antibody or antibody fragment thereof according to any one of (1) to (3).
(18-2) The antibody or antibody fragment thereof according to any one of (1) to (3) for cancer diagnosis.
(18-3) Use of the antibody or antibody fragment thereof according to any one of (1) to (3) for producing a cancer diagnostic agent.
(19) A diagnostic agent containing the antibody or antibody fragment thereof according to any one of (1) to (3), for determining the appropriateness of administration of a medicine containing an anti-CCR8 antibody.
(19-2) The antibody or antibody fragment thereof according to any one of (1) to (3) for determining suitability of administration of a medicine containing an anti-CCR8 antibody.
(19-3) Use of the antibody or antibody fragment thereof according to any one of (1) to (3) for producing a diagnostic agent for determining the suitability of administration of a medicine containing an anti-CCR8 antibody.
(20) The diagnostic agent according to (19), wherein the medicine containing the anti-CCR8 antibody is an anticancer agent.
(21) The anti-CCR8 antibody contained in the anti-CCR8 antibody-containing medicine is a monoclonal antibody comprising a light chain variable region having the amino acid sequence of SEQ ID NO: 12 and a heavy chain variable region having the amino acid sequence of SEQ ID NO: 13, or The diagnostic agent according to (19), which is a fragment thereof.
(22) A method for determining the suitability of administration of a medicament containing an anti-CCR8 antibody, using a composition containing the antibody or antibody fragment thereof according to any one of (1) to (3).
(23) The method according to (22), wherein the medicine containing the anti-CCR8 antibody is an anticancer agent.
(24) The anti-CCR8 antibody contained in the anti-CCR8 antibody-containing medicine is a monoclonal antibody comprising a light chain variable region having the amino acid sequence of SEQ ID NO: 12 and a heavy chain variable region having the amino acid sequence of SEQ ID NO: 13; The method according to (22), which is a fragment thereof.
(25) Using a composition containing the antibody or antibody fragment thereof according to any one of (1) to (3), the anti-CCR8 antibody contained in the anti-CCR8 antibody-containing medicine is used to infiltrate into tumor-infiltrating Treg cells. How to measure the amount of binding.
(26) A polynucleotide encoding the light chain variable region or heavy chain variable region of the antibody according to (1) or (2).
(27) An expression vector comprising the polynucleotide described in (26).
 本発明の新規抗CCR8抗体は、CCR8に特異的に結合するので、生体試料においてCCR8を検出するために使用することができる。さらに、本発明の新規抗CCR8抗体は、腫瘍内浸潤Treg細胞に発現するCCR8を検出するので、癌診断やコンパニオン診断に非常に有用である。 Since the novel anti-CCR8 antibody of the present invention specifically binds to CCR8, it can be used to detect CCR8 in biological samples. Furthermore, the novel anti-CCR8 antibody of the present invention detects CCR8 expressed in tumor-infiltrating Treg cells, and is therefore very useful for cancer diagnosis and companion diagnosis.
ラットハイブリドーマ上清のL263G8抗体に対する競合阻害活性をフローサイトメトリー法により評価した。抗体にL263G8抗体に対する競合阻害活性がある場合は、陰性コントロール(Cont)と比較してピークのシフトが認められる。X軸は検出蛍光強度、Y軸は検出細胞数を示す。The competitive inhibitory activity of rat hybridoma supernatant against L263G8 antibody was evaluated by flow cytometry. If the antibody has competitive inhibitory activity against the L263G8 antibody, a peak shift will be observed compared to the negative control (Cont). The X axis shows the detected fluorescence intensity, and the Y axis shows the number of detected cells. ラットハイブリドーマ上清による、CCL1とCCR8の結合に対する中和活性を細胞内カルシウム流入活性測定により評価した。抗体が中和活性を示す場合は、細胞内カルシウム流入活性が抑制されるため、蛍光量の経時的上昇も抑制される。The neutralizing activity of the rat hybridoma supernatant against the binding between CCL1 and CCR8 was evaluated by measuring intracellular calcium influx activity. When the antibody exhibits neutralizing activity, intracellular calcium influx activity is suppressed, and therefore the increase in fluorescence level over time is also suppressed. ヒトCCR8発現Ramos細胞における3-3F抗体、L263G8抗体及び433H抗体の結合をフローサイトメトリー法により評価した。X軸は検出蛍光強度、Y軸は検出細胞数を示す。The binding of the 3-3F antibody, L263G8 antibody, and 433H antibody to human CCR8-expressing Ramos cells was evaluated by flow cytometry. The X axis shows the detected fluorescence intensity, and the Y axis shows the number of detected cells. ヒトCCR8発現Ramos細胞における、ヒトCCR8に対する中和抗体であるOP-1抗体存在下での、3-3F抗体、L263G8抗体及び433H抗体の結合をフローサイトメトリー法により評価した。X軸は検出蛍光強度、Y軸は検出細胞数を示す。The binding of 3-3F antibody, L263G8 antibody, and 433H antibody to human CCR8-expressing Ramos cells in the presence of OP-1 antibody, which is a neutralizing antibody against human CCR8, was evaluated by flow cytometry. The X axis shows the detected fluorescence intensity, and the Y axis shows the number of detected cells. 各種ケモカイン受容体発現細胞に対する3-3F抗体の結合をフローサイトメトリー法により評価した。X軸は検出蛍光強度、Y軸は検出細胞数を示し、各グラフで「3-3F」は3-3F抗体のピーク、「Cont」は陰性コントロールのピークを示す。The binding of the 3-3F antibody to cells expressing various chemokine receptors was evaluated by flow cytometry. The X axis shows the detected fluorescence intensity, and the Y axis shows the number of detected cells. In each graph, "3-3F" shows the peak of the 3-3F antibody, and "Cont" shows the peak of the negative control. 腫瘍内浸潤Treg細胞に対する3-3F抗体の結合をフローサイトメトリー法により評価した。X軸は検出蛍光強度、Y軸は検出細胞数を示す。The binding of the 3-3F antibody to intratumoral infiltrating Treg cells was evaluated by flow cytometry. The X axis shows the detected fluorescence intensity, and the Y axis shows the number of detected cells. 腫瘍内浸潤Treg細胞に対する3-3F抗体、蛍光標識3-3F抗体及びL263G8抗体の結合をフローサイトメトリー法により評価した。X軸はFoxP3検出蛍光強度、Y軸は各種抗CCR8抗体によるCCR8検出蛍光強度を示す。The binding of 3-3F antibody, fluorescently labeled 3-3F antibody, and L263G8 antibody to intratumoral infiltrating Treg cells was evaluated by flow cytometry. The X axis shows the fluorescence intensity detected by FoxP3, and the Y axis shows the fluorescence intensity detected by CCR8 by various anti-CCR8 antibodies. 腫瘍内浸潤Treg細胞のフローサイトメトリー解析による、3-3F抗体シグナル(Y軸)とCCR8 mRNAシグナル(X軸)の相関を示す。The correlation between 3-3F antibody signal (Y axis) and CCR8 mRNA signal (X axis) is shown by flow cytometry analysis of intratumoral infiltrating Treg cells. 腫瘍内浸潤Treg細胞における、ヒトCCR8ノックインマウスに投与したヒトCCR8に対する中和抗体(OP-1抗体)のTreg細胞に対する結合シグナル(X軸)と3-3F抗体シグナル(Y軸)の相関を示す。図中のrは相関係数、直線は線形回帰を示す。Showing the correlation between the binding signal (X axis) to Treg cells of a neutralizing antibody (OP-1 antibody) against human CCR8 (OP-1 antibody) administered to human CCR8 knock-in mice and the 3-3F antibody signal (Y axis) in intratumoral infiltrating Treg cells. . In the figure, r indicates a correlation coefficient, and a straight line indicates linear regression. 腫瘍内浸潤Treg細胞における、ヒトCCR8ノックインマウスに投与したヒトCCR8に対する中和抗体(OP-1抗体)のTreg細胞に対する結合シグナル(X軸)と433H抗体シグナル(Y軸)の相関を示す。図中のrは相関係数、直線は線形回帰を示す。The correlation between the binding signal (X axis) to Treg cells of a neutralizing antibody (OP-1 antibody) against human CCR8 (OP-1 antibody) administered to human CCR8 knockin mice and the 433H antibody signal (Y axis) in tumor-infiltrating Treg cells is shown. In the figure, r indicates a correlation coefficient, and a straight line indicates linear regression.
 本明細書において使用される用語は、特に言及する場合を除いて、当該分野で通常用いられる意味で用いられる。 The terms used herein have the meanings commonly used in the field, unless otherwise specified.
 本発明においては、当該分野で公知の抗体作製手法が利用可能である。例えば、Immunochemistry in Practice (Blackwell Scientific Publications)に記載された方法等が挙げられる。
 また、当該分野で公知の遺伝子操作的手法が利用可能である。例えば、Molecular Cloning, A Laboratory Manual, Fourth Edition, Cold Spring Harbor Laboratory Press (2012), Current Protocols Essential Laboratory Techniques, Current Protocols (2012)に記載された方法等が挙げられる。
In the present invention, antibody production techniques known in the art can be used. Examples include the method described in Immunochemistry in Practice (Blackwell Scientific Publications).
Additionally, genetic engineering techniques known in the art are available. Examples include the methods described in Molecular Cloning, A Laboratory Manual, Fourth Edition, Cold Spring Harbor Laboratory Press (2012), Current Protocols Essential Laboratory Techniques, Current Protocols (2012).
 ヒトCCR8のアミノ酸配列は、UniProtKB/Swiss-Prot:P51685に示されている(配列番号1)。ヒトCCR8の膜外ドメインは、1~35位アミノ酸からなるN末領域、94~107位アミノ酸からなるloop1領域、172~202位アミノ酸からなるloop2領域、264~280位アミノ酸からなるloop3領域が該当する。ヒトCCR8のアミノ酸配列における17位はチロシンであり、マウスCCR8のアミノ酸配列においては、17位は欠損している。 The amino acid sequence of human CCR8 is shown in UniProtKB/Swiss-Prot:P51685 (SEQ ID NO: 1). The extramembrane domain of human CCR8 corresponds to the N-terminal region consisting of amino acids 1 to 35, the loop 1 region consisting of amino acids 94 to 107, the loop 2 region consisting of amino acids 172 to 202, and the loop 3 region consisting of amino acids 264 to 280. do. Position 17 in the amino acid sequence of human CCR8 is tyrosine, and position 17 is deleted in the amino acid sequence of mouse CCR8.
 本発明の抗CCR8抗体を産生するハイブリドーマは、ヒトCCR8タンパク質、ヒトCCR8の全長をコードする遺伝子、ヒトCCR8発現細胞等を免疫原として作製することができる。ヒトCCR8の全長をコードする遺伝子を免疫原とする場合は、例えば、該遺伝子を抗原としてDNA免疫したマウスの脾臓細胞とマウスミエローマ細胞を融合させることにより、抗CCR8抗体を産生するハイブリドーマを得ることができる。 A hybridoma producing the anti-CCR8 antibody of the present invention can be produced using human CCR8 protein, a gene encoding the full-length human CCR8, human CCR8-expressing cells, etc. as an immunogen. When using a gene encoding the full length of human CCR8 as an immunogen, for example, a hybridoma producing an anti-CCR8 antibody can be obtained by fusing mouse myeloma cells with spleen cells of a mouse immunized with DNA using the gene as an antigen. I can do it.
 ハイブリドーマを培養するための基本培地としては、例えば、D-MEM培地、RPMI1640培地、IMDM培地、ASF104培地等が挙げられ、該基本培地には、目的に応じて、例えば血清、ホルモン、サイトカイン及び/または種々無機あるいは有機物質等を含有させることができる。 Examples of the basic medium for culturing hybridomas include D-MEM medium, RPMI1640 medium, IMDM medium, and ASF104 medium. Alternatively, various inorganic or organic substances can be contained.
 本発明のモノクローナル抗体又はその抗体断片の一つの態様としては、本明細書記載のCDR又は重鎖可変領域/軽鎖可変領域を有するモノクローナル抗体又は該抗体の断片が挙げられる。該抗体又は抗体断片は、免疫グロブリン分子の任意のクラス(例えば、IgG、IgE、IgM、IgD若しくはIgA。好ましくは、IgG。)又はサブクラス由来であり得、例えば、マウス、ラット、サメ、ウサギ、ブタ、ハムスター、ラクダ、ラマ、ヤギ又はヒトを含む任意の種から取得されてもよい。該抗体又は抗体断片として、好ましくは、ヒト化モノクローナル抗体又はヒト化モノクローナル抗体の抗体断片である。 One embodiment of the monoclonal antibody or antibody fragment thereof of the present invention includes a monoclonal antibody or a fragment of the antibody having the CDR or heavy chain variable region/light chain variable region described herein. The antibody or antibody fragment may be derived from any class (e.g. IgG, IgE, IgM, IgD or IgA, preferably IgG) or subclass of immunoglobulin molecules, e.g. mouse, rat, shark, rabbit, It may be obtained from any species including pigs, hamsters, camels, llamas, goats or humans. The antibody or antibody fragment is preferably a humanized monoclonal antibody or an antibody fragment of a humanized monoclonal antibody.
 本明細書中、エピトープは、その抗原を標的とする抗体によって結合される抗原の領域を意味し、抗原がタンパク質である場合、抗体に直接接触する特定のアミノ酸を含む。本発明には、本発明のモノクローナル抗体又はその抗体断片と完全に同じエピトープを認識するモノクローナル抗体又はその抗体断片のみならず、部分的に同じエピトープを認識するモノクローナル抗体又はその抗体断片も包含される。 As used herein, epitope refers to the region of an antigen that is bound by an antibody targeting that antigen, and when the antigen is a protein, includes specific amino acids that are in direct contact with the antibody. The present invention includes not only monoclonal antibodies or antibody fragments thereof that recognize the completely same epitope as the monoclonal antibody or antibody fragment thereof of the present invention, but also monoclonal antibodies or antibody fragments thereof that recognize partially the same epitope. .
 本発明のモノクローナル抗体のCDR配列は、好ましくは以下の配列を有している。
(1)軽鎖CDR1:配列番号2。
(2)軽鎖CDR2:配列番号3。
(3)軽鎖CDR3:配列番号4。
(4)重鎖CDR1:配列番号5。
(5)重鎖CDR2:配列番号6。
(6)重鎖CDR3:配列番号7。
The CDR sequence of the monoclonal antibody of the present invention preferably has the following sequence.
(1) Light chain CDR1: SEQ ID NO: 2.
(2) Light chain CDR2: SEQ ID NO: 3.
(3) Light chain CDR3: SEQ ID NO: 4.
(4) Heavy chain CDR1: SEQ ID NO: 5.
(5) Heavy chain CDR2: SEQ ID NO: 6.
(6) Heavy chain CDR3: SEQ ID NO: 7.
 本発明において「モノクローナル抗体の抗体断片」とは、本発明のモノクローナル抗体の一部であって、当該モノクローナル抗体と同様にヒトCCR8に特異的に結合し、ヒトCCR8を選択的に阻害する断片を意味する。
 本発明の「抗CCR8抗体」には、「モノクローナル抗体の抗体断片」を含む。
In the present invention, the term "antibody fragment of a monoclonal antibody" refers to a fragment that is part of the monoclonal antibody of the present invention and that specifically binds to human CCR8 and selectively inhibits human CCR8 like the monoclonal antibody. means.
The "anti-CCR8 antibody" of the present invention includes "antibody fragment of a monoclonal antibody."
 具体的には、ヒトCCR8に対して特異的に結合するFab(fragment of antigen binding)、Fab’、F(ab’)、一本鎖抗体(single chain Fv;以下、scFvと表記する)、ジスルフィド安定化抗体(disulfide stabilized Fv; 以下、dsFvと表記する)、2量化体V領域断片(以下、Diabodyと表記する)、CDRを含むペプチド等を挙げることができる(エキスパート・オピニオン・オン・テラピューティック・パテンツ、第6巻、第5号、第441~456頁、1996年)。 Specifically, Fab (fragment of antigen binding), Fab', F(ab') 2 , single chain antibody (single chain Fv; hereinafter referred to as scFv) that specifically binds to human CCR8, Examples include disulfide stabilized antibodies (disulfide stabilized Fv; hereinafter referred to as dsFv), dimerized V region fragments (hereinafter referred to as diabody), peptides containing CDRs, etc. Pytic Patents, Vol. 6, No. 5, pp. 441-456, 1996).
 Fabは、IgGのヒンジ領域で2本のH鎖を架橋している2つのジスルフィド結合(S-S結合)の上部のペプチド部分を酵素パパインで分解して得られる、H鎖のN末端側約半分とL鎖全体で構成される、分子量約5万の抗原結合活性を有する抗体断片である。本発明で使用されるFabは、本発明のモノクローナル抗体をパパイン処理して得ることができる。また、本発明のモノクローナル抗体のFabをコードするDNAを細胞用発現ベクターに挿入し、該ベクターを細胞へ導入することにより発現させることによってもFabを製造することができる。 Fab is obtained by decomposing the peptide part above the two disulfide bonds (SS bonds) that bridge the two H chains in the hinge region of IgG with the enzyme papain. It is an antibody fragment with a molecular weight of approximately 50,000 and antigen-binding activity, consisting of one half and the entire L chain. Fab used in the present invention can be obtained by treating the monoclonal antibody of the present invention with papain. Furthermore, Fab can also be produced by inserting DNA encoding the Fab of the monoclonal antibody of the present invention into a cellular expression vector, and expressing the vector by introducing the vector into cells.
 Fab’は、F(ab’)のヒンジ間のS-S結合を切断した分子量約5万の抗原結合活性を有する抗体断片である。本発明で使用されるFab’は、本発明のモノクローナル抗体のF(ab’)を還元剤ジチオスレイトール処理して得ることができる。また、本発明のモノクローナル抗体のFab’をコードするDNAを細胞用発現ベクターに挿入し、該ベクターを大腸菌、酵母又は動物細胞へ導入することにより発現させることによってもFab’を製造することができる。 Fab' is an antibody fragment with a molecular weight of approximately 50,000 and having antigen-binding activity obtained by cleaving the SS bond between the hinges of F(ab') 2 . Fab' used in the present invention can be obtained by treating F(ab') 2 of the monoclonal antibody of the present invention with a reducing agent dithiothreitol. Fab' can also be produced by inserting the DNA encoding Fab' of the monoclonal antibody of the present invention into a cellular expression vector, and expressing the vector by introducing it into E. coli, yeast, or animal cells. .
 F(ab’)は、IgGのヒンジ領域の2個のS-S結合の下部を酵素ペプシンで分解して得られる、2つのFab’領域がヒンジ部分で結合して構成される、分子量約10万の抗原結合活性を有する抗体断片である。本発明で使用されるF(ab’)は、本発明のモノクローナル抗体をペプシン処理して得ることができる。また、本発明のモノクローナル抗体のF(ab’)をコードするDNAを細胞用発現ベクターに挿入し、該ベクターを大腸菌、酵母又は動物細胞へ導入することにより発現させることによってもF(ab’)を製造することができる。 F(ab') 2 is obtained by decomposing the lower part of the two SS bonds in the hinge region of IgG with the enzyme pepsin, and is composed of two Fab' regions linked at the hinge region, and has a molecular weight of approximately It is an antibody fragment with an antigen binding activity of 100,000. F(ab') 2 used in the present invention can be obtained by treating the monoclonal antibody of the present invention with pepsin. F(ab') 2 of the monoclonal antibody of the present invention can also be expressed by inserting the DNA encoding it into an expression vector for cells and introducing the vector into E. coli, yeast, or animal cells. ) 2 can be manufactured.
 scFvは、一本のVHと一本のVLとを適当なペプチドリンカー(以下、Pと表記する)を用いて連結した、VH-P-VL又はVL-P-VHポリペプチドであり、抗原結合活性を有する抗体断片である。本発明で使用されるscFvに含まれるVH及びVLは、本発明のモノクローナル抗体のものであればよい。本発明で使用されるscFvは、本発明のモノクローナル抗体のVH及びVLをコードするcDNAを用いて、scFv発現ベクターを構築し、大腸菌、酵母又は動物細胞へ導入することにより発現させることによって製造することができる。 scFv is a VH-P-VL or VL-P-VH polypeptide in which one VH and one VL are linked using an appropriate peptide linker (hereinafter referred to as P), and is capable of antigen binding. It is an active antibody fragment. The VH and VL contained in the scFv used in the present invention may be those of the monoclonal antibody of the present invention. The scFv used in the present invention is produced by constructing an scFv expression vector using cDNA encoding the VH and VL of the monoclonal antibody of the present invention, and expressing it by introducing it into E. coli, yeast, or animal cells. be able to.
 dsFvは、VH及びVL中のそれぞれ1アミノ酸残基をシステイン残基に置換したポリペプチドをS-S結合を介して結合させたものをいう。システイン残基に置換するアミノ酸残基はReiterらにより示された方法(Protein Engineering, 7, 697, (1994))に従って、抗体の立体構造予測に基づいて選択することができる。本発明で使用されるdsFvに含まれるVH又はVLは、本発明のモノクローナル抗体のものであればよい。本発明で使用されるdsFvは、本発明のモノクローナル抗体のVH及びVLをコードするcDNAを用いて、適当な発現ベクターに挿入してdsFv発現ベクターを構築し、該発現ベクターを大腸菌、酵母又は動物細胞へ導入し、発現させることによって製造することができる。 dsFv refers to a polypeptide in which one amino acid residue in each of VH and VL is replaced with a cysteine residue, which are linked via an SS bond. Amino acid residues to be substituted for cysteine residues can be selected based on the prediction of the three-dimensional structure of the antibody according to the method described by Reiter et al. (Protein Engineering, 7, 697, (1994)). The VH or VL contained in the dsFv used in the present invention may be one of the monoclonal antibody of the present invention. The dsFv used in the present invention is constructed by inserting the cDNA encoding the VH and VL of the monoclonal antibody of the present invention into an appropriate expression vector, and then transducing the expression vector into Escherichia coli, yeast, or an animal. It can be produced by introducing it into cells and expressing it.
 Diabodyは、抗原結合特異性が同じ又は異なるscFvが2量体を形成した抗体断片であり、同じ抗原に対する2価の抗原結合活性又は異なる抗原に対する2種類の特異的な抗原結合活性を有する抗体断片である。例えば、本発明のモノクローナル抗体に特異的に反応する2価のDiabodyは、本発明のモノクローナル抗体のVH及びVLをコードするcDNAを用いて、3~10残基のペプチドリンカーを有するscFvをコードするDNAを構築し、該DNAを細胞用発現ベクターに挿入し、該発現ベクターを大腸菌、酵母又は動物細胞へ導入することによりDiabodyを発現させることによって製造することができる。 Diabody is an antibody fragment in which scFv with the same or different antigen-binding specificity forms a dimer, and has bivalent antigen-binding activity for the same antigen or two types of specific antigen-binding activity for different antigens. It is. For example, a bivalent diabody that specifically reacts with the monoclonal antibody of the present invention encodes an scFv having a peptide linker of 3 to 10 residues using cDNA encoding the VH and VL of the monoclonal antibody of the present invention. It can be produced by constructing DNA, inserting the DNA into a cell expression vector, and introducing the expression vector into E. coli, yeast, or animal cells to express the diabody.
 CDRを含むペプチドは、VH又はVLのCDRの少なくとも1領域以上を含んで構成される。複数のCDRは、直接又は適当なペプチドリンカーを介して結合させることができる。本発明で使用されるCDRを含むペプチドは、本発明のモノクローナル抗体のVH及びVLをコードするcDNAを用いて、CDRをコードするDNAを構築し、該DNAを動物細胞用発現ベクターに挿入し、該ベクターを大腸菌、酵母又は動物細胞へ導入することにより発現させることにより、製造することができる。また、CDRを含むペプチドは、Fmoc法(フルオレニルメチルオキシカルボニル法)、tBoc法(t-ブチルオキシカルボニル法)等の化学合成法によって製造することもできる。 A peptide containing a CDR is composed of at least one region of a VH or VL CDR. Multiple CDRs can be linked directly or via a suitable peptide linker. The CDR-containing peptide used in the present invention is obtained by constructing a CDR-encoding DNA using cDNA encoding the VH and VL of the monoclonal antibody of the present invention, and inserting the DNA into an expression vector for animal cells. It can be produced by introducing the vector into E. coli, yeast, or animal cells and expressing it. Peptides containing CDRs can also be produced by chemical synthesis methods such as the Fmoc method (fluorenylmethyloxycarbonyl method) and the tBoc method (t-butyloxycarbonyl method).
 本発明のモノクローナル抗体又はその抗体断片は、ヒトCCR8に結合することが特徴である。特に、特異的に結合するものが好ましい。 The monoclonal antibody of the present invention or its antibody fragment is characterized by binding to human CCR8. In particular, those that bind specifically are preferred.
 特異的結合は、少なくとも約1×10-6M又はそれ未満の平衡解離定数(例えば、Kdが小さいほど、より緊密な結合を表す)を特徴とすることができる。Kd値は、好ましくは1×10-7M以下、より好ましくは1×10-8M以下、さらに好ましくは1×10-9M以下である。2つの分子が特異的に結合するかどうかを測定する方法は、当該技術分野で周知であり、競合ELISA法、表面プラズモン共鳴及び同様のものが挙げられる。 Specific binding can be characterized by an equilibrium dissociation constant of at least about 1×10 −6 M or less (eg, a lower Kd indicates tighter binding). The Kd value is preferably 1×10 −7 M or less, more preferably 1×10 −8 M or less, and still more preferably 1×10 −9 M or less. Methods of determining whether two molecules specifically bind are well known in the art and include competitive ELISA methods, surface plasmon resonance, and the like.
 本発明は、ヒトCCR8への結合に関して、本明細書中に記載されているモノクローナル抗体又はその抗体断片と競合するモノクローナル抗体又はその抗体断片を含む。
 「競合するモノクローナル抗体又はその抗体断片」は、本発明のモノクローナル抗体又はその抗体断片(好ましくは、配列番号12のアミノ酸配列を有する軽鎖可変領域、及び、配列番号13のアミノ酸配列を有する重鎖可変領域を含むモノクローナル抗体又はその断片)の、ヒトCCR8への特異的結合を阻害する抗体又はその抗体断片を意味する。あるモノクローナル抗体又はその抗体断片が本発明のモノクローナル抗体又はその抗体断片と競合するかどうかを決定する。Isotype control抗体の共存下、抗原(ヒトCCR8)との結合が確認できた抗体のうち、本発明のモノクローナル抗体又はその抗体断片の共存下では結合シグナルが顕著に低下する抗体を、本発明のモノクローナル抗体又はその抗体断片と競合する抗体として同定することができる。該結合シグナルの低下は、好ましくは50%、より好ましくは70%である。また、本発明のモノクローナル抗体又はその抗体断片と競合する抗体の、該本発明のモノクローナル抗体又はその抗体断片と抗原との結合に対するKi値は、好ましくは1×10-7M以下、より好ましくは1×10-8M以下、さらに好ましくは1×10-9M以下である。
The invention includes monoclonal antibodies or antibody fragments thereof that compete with the monoclonal antibodies or antibody fragments thereof described herein for binding to human CCR8.
"Competing monoclonal antibody or antibody fragment thereof" refers to the monoclonal antibody of the present invention or antibody fragment thereof (preferably a light chain variable region having the amino acid sequence of SEQ ID NO: 12 and a heavy chain having the amino acid sequence of SEQ ID NO: 13). It refers to an antibody or an antibody fragment thereof that inhibits the specific binding of a monoclonal antibody containing a variable region (or a fragment thereof) to human CCR8. It is determined whether a monoclonal antibody or antibody fragment thereof competes with a monoclonal antibody or antibody fragment thereof of the invention. Among the antibodies that were confirmed to bind to the antigen (human CCR8) in the presence of the isotype control antibody, those antibodies whose binding signal was significantly reduced in the presence of the monoclonal antibody of the present invention or its antibody fragment were selected as the monoclonal antibody of the present invention. It can be identified as an antibody that competes with the antibody or antibody fragment thereof. The reduction in binding signal is preferably 50%, more preferably 70%. Further, the Ki value of an antibody that competes with the monoclonal antibody of the present invention or its antibody fragment for binding of the monoclonal antibody of the present invention or its antibody fragment to an antigen is preferably 1×10 −7 M or less, more preferably It is 1×10 −8 M or less, more preferably 1×10 −9 M or less.
 本発明のモノクローナル抗体又はその抗体断片は、CCR8とCCL1との結合をほとんど阻害しない、つまり中和活性をほとんど有さないことが特徴である。
 CCR8とCCL1の結合阻害能については、例えば、ヒトCCL1の場合、ヒトCCR8発現293細胞を用い、ヒトCCL1添加によるCa2+流入を測定し、ヒトCCL1非添加時のシグナルを阻害率100%、ヒトCCL1添加及び抗体非添加時のシグナルを阻害率0%として、IC50値を計算することにより決定することができる。例えば、後述の実施例3記載の方法等にて測定することができる。
 ヒトCCL1は、UniProtKB/Swiss-Prot No.P22362などに示されるアミノ酸配列を有する。
The monoclonal antibody of the present invention or its antibody fragment is characterized in that it hardly inhibits the binding between CCR8 and CCL1, that is, it has almost no neutralizing activity.
Regarding the ability to inhibit the binding between CCR8 and CCL1, for example, in the case of human CCL1, using human CCR8-expressing 293 cells, Ca 2+ influx upon addition of human CCL1 was measured, and the signal when human CCL1 was not added was determined to be 100% inhibition rate, human The IC50 value can be determined by calculating the IC50 value, assuming that the signal obtained when CCL1 is added and when the antibody is not added is regarded as an inhibition rate of 0%. For example, it can be measured by the method described in Example 3 below.
Human CCL1 was obtained from UniProtKB/Swiss-Prot No. It has an amino acid sequence such as P22362.
 本発明のモノクローナル抗体は、本明細書記載のCDR又は重鎖可変領域/軽鎖可変領域を用いて当該技術分野の定法によって作製することができる。 The monoclonal antibody of the present invention can be produced by a conventional method in the art using the CDRs or heavy chain variable region/light chain variable region described herein.
 本発明のモノクローナル抗体は、キメラ、ヒト化、完全ヒト、抗体低分子複合体(ADC)及び二重特異的抗体も含む。
 ヒト化モノクローナル抗体はヒト体内における抗原性が低下しているため、治療目的等でヒトに投与する場合に有用である。ヒト化モノクローナル抗体は、ヒト以外の哺乳動物、例えばマウス抗体の相補性決定領域(CDR; complementarity determining region)をヒト抗体のフレームワーク領域(FR; framework region)へ移植したものである。従って、ヒト化モノクローナル抗体のFRは、ヒト由来のものである。適当なFRは、Kabat E.A.らの文献を参照すれば選択できる。この場合のFRとしては、CDRが良好な抗原結合部位を形成することができるものが選択される。必要に応じ、再構成したヒト化モノクローナル抗体のCDRが適切な抗原結合部位を形成するように、抗体の可変領域のFRのアミノ酸を置換してもよい(Sato. et al, Cancer Res. (1993), 53, 851)。置換されるFRのアミノ酸の割合は、全FR領域の0~15%、好ましくは0~5%である。
Monoclonal antibodies of the invention also include chimeric, humanized, fully human, antibody small molecule conjugates (ADCs) and bispecific antibodies.
Since humanized monoclonal antibodies have reduced antigenicity in the human body, they are useful when administered to humans for therapeutic purposes. A humanized monoclonal antibody is one in which the complementarity determining regions (CDRs) of a non-human mammal, such as a mouse antibody, are grafted onto the framework regions (FRs) of a human antibody. Therefore, the FRs of the humanized monoclonal antibody are of human origin. A suitable FR is Kabat E. A. You can make a selection by referring to the literature by et al. In this case, FRs are selected that allow CDRs to form a good antigen-binding site. If necessary, amino acids in the FRs of the antibody variable region may be substituted so that the CDRs of the reconstituted humanized monoclonal antibody form a suitable antigen-binding site (Sato. et al, Cancer Res. (1993) ), 53, 851). The proportion of FR amino acids to be replaced is 0-15%, preferably 0-5% of the total FR region.
 本発明のヒト化モノクローナル抗体は、好ましくは、
配列番号2のアミノ酸配列からなるCDR1、
配列番号3のアミノ酸配列からなるCDR2及び
配列番号4のアミノ酸配列からなるCDR3を含む軽鎖可変領域並びに
配列番号5のアミノ酸配列からなるCDR1、
配列番号6のアミノ酸配列からなるCDR2及び
配列番号7のアミノ酸配列からなるCDR3を含む重鎖可変領域
を含む。
 より好ましくは、
配列番号8のアミノ酸配列を有する軽鎖可変領域、及び、配列番号9のアミノ酸配列を有する重鎖可変領域を含む。
The humanized monoclonal antibody of the present invention preferably comprises:
CDR1 consisting of the amino acid sequence of SEQ ID NO: 2,
A light chain variable region comprising CDR2 consisting of the amino acid sequence of SEQ ID NO: 3 and CDR3 consisting of the amino acid sequence of SEQ ID NO: 4, and CDR1 consisting of the amino acid sequence of SEQ ID NO: 5,
It contains a heavy chain variable region including CDR2 consisting of the amino acid sequence of SEQ ID NO: 6 and CDR3 consisting of the amino acid sequence of SEQ ID NO: 7.
More preferably,
It includes a light chain variable region having the amino acid sequence of SEQ ID NO: 8 and a heavy chain variable region having the amino acid sequence of SEQ ID NO: 9.
 本発明のモノクローナル抗体の定常領域の配列は特に限定されないが、配列番号10のアミノ酸配列を有する軽鎖定常領域、及び、配列番号11のアミノ酸配列を有する重鎖定常領域が例示される。 The sequence of the constant region of the monoclonal antibody of the present invention is not particularly limited, but examples include a light chain constant region having the amino acid sequence of SEQ ID NO: 10 and a heavy chain constant region having the amino acid sequence of SEQ ID NO: 11.
 本発明のモノクローナル抗体がヒト化モノクローナル抗体の場合、ヒト抗体の定常領域が使用される。好ましいヒト抗体の定常領域としては、重鎖としてはCγが挙げられ、例えば、Cγ1,Cγ2,Cγ3,Cγ4を、軽鎖としてはCκ、Cλを使用することができる。また、抗体又はその産生の安定性を改善するために、ヒト抗体のC領域を修飾してもよい。ヒト化の際に用いられるヒト抗体は、IgG、IgM、IgA、IgE、IgD等いかなるアイソタイプのヒト抗体でもよいが、本発明においてはIgGを用いることが好ましい。 When the monoclonal antibody of the present invention is a humanized monoclonal antibody, the constant region of a human antibody is used. Preferred constant regions of human antibodies include Cγ as a heavy chain, such as Cγ1, Cγ2, Cγ3, and Cγ4, and Cκ and Cλ as light chains. Additionally, the C region of human antibodies may be modified to improve the stability of the antibody or its production. The human antibody used in humanization may be of any isotype such as IgG, IgM, IgA, IgE, IgD, etc., but it is preferable to use IgG in the present invention.
 本発明のモノクローナル抗体は一般的な製造方法によって作製することができる(例えば、WO95/14041号公報、WO96/02576号公報等参照)。本発明で使用される抗体を製造するには、抗体遺伝子を発現制御領域、例えば、エンハンサー/プロモーターの制御のもとで発現するよう発現ベクターに組み込む。次に、この発現ベクターにより宿主細胞を形質転換し、抗体を発現させることができる。 The monoclonal antibody of the present invention can be produced by a general production method (for example, see WO95/14041, WO96/02576, etc.). To produce the antibody used in the present invention, the antibody gene is inserted into an expression vector so that it is expressed under the control of an expression control region, such as an enhancer/promoter. A host cell can then be transformed with this expression vector to express the antibody.
 上記の形質転換体の宿主細胞としては、例えば、COS細胞、CHO細胞等の脊椎動物細胞、原核細胞、酵母等が挙げられる。形質転換体は、当業者に周知の方法に従って培養することができ、該培養により、形質転換体細胞内又は細胞外に本発明のモノクローナル抗体が産生される。該培養に用いられる培地としては、採用した宿主細胞に応じて慣用される各種のものを適宜選択でき、例えば、COS細胞の場合、RPMI-1640培地やダルベッコ修正イーグル最小必須培地(DMEM)等の培地に、必要に応じウシ胎児血清(FBS)等の血清成分を添加したものを使用できる。該形質転換体の培養の際の培養温度は、細胞内のタンパク質合成能を著しく低下せしめない温度であればいずれでもよいが、好適には32~42℃、最も好適には37℃で培養することが好ましい。また必要に応じて、1~10%(v/v)の炭酸ガスを含む空気中で培養することができる。 Examples of host cells for the above transformants include vertebrate cells such as COS cells and CHO cells, prokaryotic cells, and yeast. The transformant can be cultured according to methods well known to those skilled in the art, and the monoclonal antibody of the present invention is produced within or outside the transformant cells. The culture medium used for the culture can be appropriately selected from various commonly used media depending on the host cell employed. For example, in the case of COS cells, RPMI-1640 medium, Dulbecco's modified Eagle's minimum essential medium (DMEM), etc. A culture medium to which a serum component such as fetal bovine serum (FBS) is added, if necessary, can be used. The culture temperature for culturing the transformant may be any temperature as long as it does not significantly reduce the intracellular protein synthesis ability, but it is preferably cultured at 32 to 42°C, most preferably at 37°C. It is preferable. Furthermore, if necessary, the culture can be carried out in air containing 1 to 10% (v/v) carbon dioxide gas.
 上記により形質転換体の細胞内又は細胞外に生産される本発明のモノクローナル抗体を含む画分は、該タンパク質の物理的性質や化学的性質等を利用した各種公知の分離操作法により、分離・精製することができる。かかる方法としては、具体的には例えば通常のタンパク質沈澱剤による処理、限外濾過、分子ふるいクロマトグラフィー(ゲル濾過)、吸着クロマトグラフィー、イオン交換クロマトグラフィー、アフィニティークロマトグラフィー、高速液体クロマトグラフィー(HPLC)等の各種クロマトグラフィー、透析法、及びこれらの組み合わせ等を採用できる。該方法により、容易に高収率、高純度で本発明のモノクローナル抗体を製造できる。 The fraction containing the monoclonal antibody of the present invention produced intracellularly or extracellularly in the transformant as described above can be separated and separated by various known separation methods that utilize the physical and chemical properties of the protein. Can be purified. Specifically, such methods include, for example, treatment with a normal protein precipitant, ultrafiltration, molecular sieve chromatography (gel filtration), adsorption chromatography, ion exchange chromatography, affinity chromatography, high performance liquid chromatography (HPLC). ), various chromatography methods, dialysis methods, and combinations thereof can be employed. By this method, the monoclonal antibody of the present invention can be easily produced with high yield and high purity.
 本発明のモノクローナル抗体又はその抗体断片は、さらに、ポリエチレングリコール(PEG)、放射性物質、トキシン等の各種分子により修飾されていてもよい。抗体の修飾方法はこの分野で公知の方法を利用することができる。 The monoclonal antibody of the present invention or its antibody fragment may be further modified with various molecules such as polyethylene glycol (PEG), radioactive substances, and toxins. Methods known in this field can be used to modify antibodies.
 また本発明のモノクローナル抗体は、そのN末端あるいはC末端に他のタンパク質を融合してもよい(Clinical Cancer Research, 2004, 10, 1274-1281)。融合するタンパク質は当業者が適宜選択することができる。 The monoclonal antibody of the present invention may also be fused with other proteins to its N-terminus or C-terminus (Clinical Cancer Research, 2004, 10, 1274-1281). Proteins to be fused can be appropriately selected by those skilled in the art.
 本発明のモノクローナル抗体には、抗体のFc領域にN-グリコシド結合糖鎖が結合している抗体が含まれる。なお、該N-グリコシド結合糖鎖の還元末端のN-アセチルグルコサミンにフコースが結合していなくてもよい。抗体のFc領域にN-グリコシド結合糖鎖が結合し、該N-グリコシド結合糖鎖の還元末端のN-アセチルグルコサミンにフコースが結合していない抗体としては、例えばα1,6-フコース転移酵素遺伝子が欠損したCHO細胞(国際公開第2005/035586号、国際公開第02/31140号)を用いて作製される抗体が挙げられる。抗体のFc領域にN-グリコシド結合糖鎖が結合し、該N-グリコシド結合糖鎖の還元末端のN-アセチルグルコサミンにフコースが結合していない本発明の抗体は、高いADCC活性を有する。 The monoclonal antibodies of the present invention include antibodies in which N-glycoside-linked sugar chains are bound to the Fc region of the antibody. Note that fucose does not need to be bound to N-acetylglucosamine at the reducing end of the N-glycoside-linked sugar chain. An example of an antibody in which an N-glycoside-linked sugar chain is bound to the Fc region of the antibody and fucose is not bound to N-acetylglucosamine at the reducing end of the N-glycoside-linked sugar chain is, for example, an α1,6-fucosyltransferase gene. Examples include antibodies produced using CHO cells deficient in CHO cells (WO 2005/035586, WO 02/31140). The antibody of the present invention, in which an N-glycoside-linked sugar chain is bound to the Fc region of the antibody and fucose is not bound to N-acetylglucosamine at the reducing end of the N-glycoside-linked sugar chain, has high ADCC activity.
 本発明のモノクローナル抗体又はその抗体断片は、Treg細胞又はマクロファージ細胞を除去させるため、CCR8を発現する細胞に対してADCC(Antibody-dependent cell mediated cytotoxicity;抗体依存性細胞介在性細胞傷害)活性を有していてもよい。ADCC活性とは、生体内で、標的細胞等の細胞表面抗原などに結合した抗体が、抗体のFc領域とエフェクター細胞表面上に存在するFcレセプターとの結合を介してエフェクター細胞を活性化し、標的細胞等を傷害する活性を意味する。エフェクター細胞としては、ナチュラルキラー細胞、活性化されたマクロファージ等があげられる。なお、本発明においては、生体内で、Treg細胞又はマクロファージ細胞等の細胞表面抗原(CCR8)に結合した抗体が、抗体のFc領域とエフェクター細胞表面上に存在するFcレセプターとの結合を介してエフェクター細胞を活性化し、Treg細胞又はマクロファージ細胞等を傷害し、結果として腫瘍細胞等を傷害する場合も含む。 The monoclonal antibody or antibody fragment thereof of the present invention has ADCC (Antibody-dependent cell mediated cytotoxicity) activity against cells expressing CCR8 in order to remove Treg cells or macrophage cells. You may do so. ADCC activity means that in vivo, an antibody that binds to a cell surface antigen such as a target cell activates the effector cell through the binding between the Fc region of the antibody and the Fc receptor present on the surface of the effector cell. Refers to activity that damages cells, etc. Effector cells include natural killer cells, activated macrophages, and the like. In addition, in the present invention, in vivo, an antibody bound to a cell surface antigen (CCR8) such as a Treg cell or a macrophage cell is activated through the binding between the Fc region of the antibody and an Fc receptor present on the surface of an effector cell. It also includes cases where effector cells are activated, Treg cells or macrophage cells are injured, and as a result, tumor cells and the like are injured.
 本発明のモノクローナル抗体又はその抗体断片は、CCR8に対する結合がCCR8の中和抗体と競合しない。CCR8の中和抗体とは、CCR8に対する中和活性を有する抗体を意味する。CCR8に対する中和活性を有するか否かは、例えば、CCR8リガンドのいずれか(例えば、CCL1)のCCR8に対する生理作用を抑制するか否かを測定することにより判別できる。例としては、これらに限定されるものではないが、CCL1のCCR8への結合、あるいはCCL1によるCCR8発現細胞の遊走又は細胞内Ca2+増加又はCCL1刺激に感受性のある遺伝子の発現変動などを測定することが挙げられる。例えば、後述の実施例3記載の方法等にて測定することができる。
 「CCR8の中和抗体」のCCR8とCCL1の結合に対する中和活性は、あらかじめCa2+指示薬を取り込ませたヒトCCR8発現293細胞に対し、培地で希釈した抗体希釈液を添加して測定することができる。CCR8とCCR8リガンドの親和性は、CCL1が最も強く、CCL1に対して高い阻害活性を有する抗体が有用である。
 「CCR8の中和抗体」は、CCR8とCCL1の結合に対する中和活性のIC50値が10nM以下であることが好ましい。中和活性は、より好ましくは、IC50値が5nM以下、さらに好ましくは2nM以下、特に好ましくは1nM以下、最も好ましくは0.5nM以下である。
 抗CCR8抗体を含有する医薬に含まれる抗CCR8抗体として用いる「CCR8の中和抗体」は、ADCC活性を有するのが好ましい。 
 「CCR8の中和抗体」としては、特許文献4~9及び15の実施例に記載された抗体、BioLegend社の品番L263G8の抗CCR8抗体、R&D社の品番191704の抗CCR8抗体等が例示されるが、配列番号8のアミノ酸配列を有する軽鎖可変領域、及び、配列番号9のアミノ酸配列を有する重鎖可変領域を含む抗体とCCR8に対する結合が競合しない抗CCR8抗体が好ましく、配列番号12のアミノ酸配列を有する軽鎖可変領域、及び、配列番号13のアミノ酸配列を有する重鎖可変領域を含む抗CCR8抗体が最も好ましい。
The monoclonal antibody of the present invention or its antibody fragment does not compete with CCR8-neutralizing antibodies for binding to CCR8. A CCR8 neutralizing antibody means an antibody having neutralizing activity against CCR8. Whether or not it has neutralizing activity against CCR8 can be determined, for example, by measuring whether the physiological effect of any CCR8 ligand (eg, CCL1) on CCR8 is suppressed. Examples include, but are not limited to, measuring the binding of CCL1 to CCR8, the migration of CCR8-expressing cells by CCL1, an increase in intracellular Ca 2+ , or changes in the expression of genes sensitive to CCL1 stimulation. This can be mentioned. For example, it can be measured by the method described in Example 3 below.
The neutralizing activity of the "CCR8 neutralizing antibody" against the binding between CCR8 and CCL1 can be measured by adding a diluted antibody solution diluted with a medium to human CCR8-expressing 293 cells that have been pre-incorporated with a Ca 2+ indicator. can. The affinity between CCR8 and CCR8 ligand is strongest for CCL1, and antibodies having high inhibitory activity against CCL1 are useful.
The "CCR8 neutralizing antibody" preferably has an IC50 value of neutralizing activity against the binding of CCR8 and CCL1 of 10 nM or less. The neutralizing activity preferably has an IC50 value of 5 nM or less, further preferably 2 nM or less, particularly preferably 1 nM or less, and most preferably 0.5 nM or less.
The "CCR8 neutralizing antibody" used as the anti-CCR8 antibody contained in the anti-CCR8 antibody-containing medicine preferably has ADCC activity.
Examples of "neutralizing antibodies for CCR8" include antibodies described in Examples of Patent Documents 4 to 9 and 15, anti-CCR8 antibodies with product number L263G8 from BioLegend, and anti-CCR8 antibodies with product number 191704 from R&D. However, an anti-CCR8 antibody that does not compete in binding to CCR8 with an antibody comprising a light chain variable region having the amino acid sequence of SEQ ID NO: 8 and a heavy chain variable region having the amino acid sequence of SEQ ID NO: 9 is preferable; Most preferred is an anti-CCR8 antibody comprising a light chain variable region having the sequence and a heavy chain variable region having the amino acid sequence of SEQ ID NO: 13.
 本発明のモノクローナル抗体又はその抗体断片は、ヒトCCR8を強く認識する抗体又はその抗体断片が好ましい。ヒトCCR8を強く認識する抗体又はその抗体断片を選択する際に、CCR8に対する結合活性の強度や結合選択性を指標にして抗体又はその抗体断片を選択することで、より強くヒトCCR8を認識する抗体又はその抗体断片を選択することができる。 The monoclonal antibody or antibody fragment thereof of the present invention is preferably an antibody or antibody fragment thereof that strongly recognizes human CCR8. When selecting an antibody or an antibody fragment thereof that strongly recognizes human CCR8, it is possible to select an antibody or an antibody fragment thereof based on the strength of binding activity and binding selectivity for CCR8 as an index, thereby creating an antibody that more strongly recognizes human CCR8. or an antibody fragment thereof can be selected.
 本発明のモノクローナル抗体又はその抗体断片は、CCR8に対する結合がCCR8の中和抗体と競合しないため、CCR8の中和抗体がすでに結合した試料であっても、CCR8の検出が可能である。さらに、試料に結合させたCCR8の中和抗体によっては、本発明のモノクローナル抗体又はその抗体断片のCCR8に対する結合活性をむしろ増加させうる。
 したがって、本発明のモノクローナル抗体又はその抗体断片は、抗CCR8抗体を含有する医薬を投与した、癌の患者等の被験者から採取した試料に含まれるCCR8を検出することが可能であり、さらに、抗CCR8抗体を含有する医薬に含まれる抗CCR8抗体の腫瘍内浸潤Treg細胞への結合量を測定することもできる。抗CCR8抗体を含有する医薬に含まれる抗CCR8抗体としては、上記「CCR8の中和抗体」が好ましく、特許文献4~9の実施例に記載された抗CCR8抗体がより好ましく、特許文献4の実施例に記載された抗CCR8抗体がさらに好ましく、配列番号8のアミノ酸配列を有する軽鎖可変領域、及び、配列番号9のアミノ酸配列を有する重鎖可変領域を含む抗体とCCR8に対する結合が競合しない抗CCR8抗体が特に好ましく、配列番号12のアミノ酸配列を有する軽鎖可変領域、及び、配列番号13のアミノ酸配列を有する重鎖可変領域を含む抗CCR8抗体が最も好ましい。
Since the monoclonal antibody of the present invention or its antibody fragment does not compete with the neutralizing antibody for CCR8 in binding to CCR8, detection of CCR8 is possible even in a sample to which a neutralizing antibody for CCR8 has already bound. Furthermore, depending on the CCR8-neutralizing antibody bound to the sample, the CCR8-binding activity of the monoclonal antibody of the present invention or its antibody fragment may be increased.
Therefore, the monoclonal antibody or antibody fragment thereof of the present invention can detect CCR8 contained in a sample collected from a subject, such as a cancer patient, who has been administered a medicine containing an anti-CCR8 antibody, and It is also possible to measure the amount of anti-CCR8 antibody contained in a medicament containing a CCR8 antibody that binds to intratumoral infiltrating Treg cells. As the anti-CCR8 antibody contained in the anti-CCR8 antibody-containing medicine, the above-mentioned "CCR8 neutralizing antibody" is preferable, the anti-CCR8 antibodies described in the Examples of Patent Documents 4 to 9 are more preferable, and the anti-CCR8 antibodies described in the Examples of Patent Documents 4 to 9 are more preferable. More preferred are the anti-CCR8 antibodies described in the Examples, which do not compete in binding to CCR8 with antibodies comprising a light chain variable region having the amino acid sequence of SEQ ID NO: 8 and a heavy chain variable region having the amino acid sequence of SEQ ID NO: 9. Anti-CCR8 antibodies are particularly preferred, and anti-CCR8 antibodies comprising a light chain variable region having the amino acid sequence of SEQ ID NO: 12 and a heavy chain variable region having the amino acid sequence of SEQ ID NO: 13 are most preferred.
 本発明のモノクローナル抗体又はその抗体断片は、CCR8に特異的に結合するため、ヒトから採取された試料においてCCR8を検出するために使用することができる。試料としては、特に限定されないが、例えば、血液、血漿、血清、尿、臓器、組織、骨髄、リンパ節等が挙げられる。 Since the monoclonal antibody of the present invention or its antibody fragment specifically binds to CCR8, it can be used to detect CCR8 in samples collected from humans. Examples of the sample include, but are not limited to, blood, plasma, serum, urine, organs, tissues, bone marrow, lymph nodes, and the like.
 本発明のモノクローナル抗体又はその抗体断片がCCR8検出用抗体として用いられる場合、CCR8を検出する際に、標識された検出用抗体を用いてもよいし、検出用抗体に対する標識二次抗体を用いてもよい。標識に用いる物質としては、蛍光物質、微小粒子、酵素、ビオチン、放射性標識体、磁性粒子等が例示されるが、蛍光物質が好ましい。 When the monoclonal antibody of the present invention or an antibody fragment thereof is used as an antibody for detecting CCR8, a labeled detection antibody may be used when detecting CCR8, or a labeled secondary antibody for the detection antibody may be used. Good too. Examples of substances used for labeling include fluorescent substances, microparticles, enzymes, biotin, radiolabels, magnetic particles, and the like, with fluorescent substances being preferred.
 蛍光物質としては、抗体又はその抗体断片の抗原結合性を損なわない限りにおいて特に限定されず、例えば、ローダミン系、クマリン系、オキサジン系、カルボピロニン系、シアニン系、ピロメセン系、ナフタレン系、ビフェニル系、アントラセン系、フェナントレン系、ピレン系、カルバゾール系、Cy系、EvoBlue系、フルオレセイン系又はこれらの誘導体等が挙げられる。 The fluorescent substance is not particularly limited as long as it does not impair the antigen binding property of the antibody or its antibody fragment, and includes, for example, rhodamine, coumarin, oxazine, carbopyronine, cyanine, pyromecene, naphthalene, biphenyl, Examples include anthracene, phenanthrene, pyrene, carbazole, Cy, EvoBlue, fluorescein, and derivatives thereof.
 微小粒子としては、抗体又はその抗体断片の抗原結合性を損なわない限りにおいて特に限定されず、例えば、金コロイドのような金属コロイド粒子、着色ラテックス粒子のような着色粒子等が挙げられる。 The microparticles are not particularly limited as long as they do not impair the antigen binding properties of the antibody or antibody fragment thereof, and include, for example, metal colloid particles such as colloidal gold, colored particles such as colored latex particles, and the like.
 酵素としては、抗体又はその抗体断片の抗原結合性を損なわない限りにおいて特に限定されず、ホースラディッシュ・ペルオキシダーゼ(HRP)、アルカリホスファターゼ、ペルオキシダーゼ、β-D-ガラクトシダーゼ、マイクロペルオキシダーゼ等が挙げられる。 The enzyme is not particularly limited as long as it does not impair the antigen binding property of the antibody or its antibody fragment, and examples include horseradish peroxidase (HRP), alkaline phosphatase, peroxidase, β-D-galactosidase, microperoxidase, and the like.
 放射性標識体としては、特に限定されないが、例えば、H、14C、15N、35S、90Y、99Tc、111In、125I、131I等が挙げられる。 Examples of radioactive labels include, but are not limited to, 3 H, 14 C, 15 N, 35 S, 90 Y, 99 Tc, 111 In, 125 I, 131 I, and the like.
 本発明の抗体又はその抗体断片は、タグを付加することができる。このようなタグとしては、例えば、FLAG、HA(ヘマグルチニン)、GST(グルタチオン-S-トランスフェラーゼ)、Myc、ポリヒスチジン(6×Hisタグ等)等が挙げられる。これらのタグは抗体の固相化、精製、検出等に用いることができる。タグ化された抗体又は抗体断片は、例えば、本発明の抗体又はその抗体断片をコードするポリヌクレオチドに、タグをコードするポリヌクレオチドを付加したものを作製し、動物細胞等のin vivo又は無細胞翻訳系等のin vitroのタンパク質発現システムに導入することにより製造することができる。 A tag can be added to the antibody of the present invention or an antibody fragment thereof. Examples of such tags include FLAG, HA (hemagglutinin), GST (glutathione-S-transferase), Myc, polyhistidine (6×His tag, etc.), and the like. These tags can be used for immobilization, purification, detection, etc. of antibodies. A tagged antibody or antibody fragment is produced by adding a polynucleotide encoding a tag to a polynucleotide encoding the antibody of the present invention or an antibody fragment thereof, and then in vivo in animal cells or the like or in a cell-free manner. It can be produced by introducing it into an in vitro protein expression system such as a translation system.
 本発明のCCR8の検出方法は、蛍光免疫測定法(FIA)、酵素免疫測定法(ELISAまたはEIA)、放射免疫測定法(RIA)、化学発光免疫測定法、表面プラズモン共鳴法、イムノクロマトグラフィー法等の免疫測定法により測定され、フローサイトメトリー法、免疫組織染色法又はELISA法による測定が好ましく、フローサイトメトリー法が特に好ましい。 The detection method of CCR8 of the present invention includes fluorescence immunoassay (FIA), enzyme immunoassay (ELISA or EIA), radioimmunoassay (RIA), chemiluminescence immunoassay, surface plasmon resonance, immunochromatography, etc. Flow cytometry, immunohistological staining, or ELISA is preferred, with flow cytometry being particularly preferred.
 本発明のCCR8の検出方法がフローサイトメトリー法の場合、例えば、CCR8及びTreg細胞に特異的なマーカーの発現を同時に測定する方法で、腫瘍内浸潤細胞等の試料中に含まれるTreg細胞において発現したCCR8を検出することができる。さらに、同様の方法で、試料よりCCR8を発現するTreg細胞を分画することもできる。Treg細胞に特異的なマーカーとしては、FoxP3、CD25、TIGITなどが例示されるが、FoxP3が好ましい。
 フローサイトメトリー法によりCCR8及びTreg細胞に特異的なマーカーの発現を同時に測定した場合、X軸をTreg細胞に特異的なマーカーの検出蛍光強度、Y軸をCCR8の検出蛍光強度とした場合、CCR8を発現するTreg細胞を示すプロットは右上の領域に現れる。なお、プロットの際に、X軸とY軸は上記と逆に設定してもよい。
When the method for detecting CCR8 of the present invention is a flow cytometry method, for example, it is a method of simultaneously measuring the expression of CCR8 and a marker specific to Treg cells, which is expressed in Treg cells contained in a sample such as intratumoral infiltrating cells. CCR8 can be detected. Furthermore, Treg cells expressing CCR8 can also be fractionated from a sample using a similar method. Examples of markers specific to Treg cells include FoxP3, CD25, and TIGIT, with FoxP3 being preferred.
When the expression of CCR8 and Treg cell-specific markers is simultaneously measured by flow cytometry, the X axis is the detected fluorescence intensity of the Treg cell-specific marker, and the Y axis is the detected fluorescence intensity of CCR8. A plot showing Treg cells expressing Treg cells appears in the upper right area. Note that when plotting, the X-axis and Y-axis may be set in the opposite manner to the above.
 本発明のCCR8の検出方法がフローサイトメトリー法の場合、CCR8及びTreg細胞に特異的なマーカーの発現を測定する前にゲートする細胞の表面抗原としては、測定の目的により適宜選択可能であるが、例えば、CD3、CD4、CD45等が選択されうる。 When the method for detecting CCR8 of the present invention is flow cytometry, the cell surface antigen to be gated before measuring the expression of markers specific to CCR8 and Treg cells can be selected as appropriate depending on the purpose of measurement. , for example, CD3, CD4, CD45, etc. may be selected.
 本発明のCCR8の検出方法が免疫組織染色法の場合、パラフィン切片等の組織切片の作成、免疫反応、洗浄、発色等の各工程を含め、公知の方法で行うことができる。 When the method for detecting CCR8 of the present invention is an immunohistological staining method, it can be carried out by known methods, including steps such as preparation of tissue sections such as paraffin sections, immunoreaction, washing, and color development.
 本発明のCCR8の検出方法がELISA法の場合、例えば、次のように行う。まず、一方の抗体(捕捉抗体)をELISA用プレートのウェル表面に固相化する。次いでウェル表面への非特異的な吸着を防ぐためにブロッキングを行った後、試料を添加し、試料中のCCR8を該抗体と接触させて複合体を形成させる。該抗体と結合しなかったタンパク質を洗浄により除去した後、標識した他方の抗体(検出抗体)をウェルに加え、CCR8に接触させて複合体を形成させ、該標識により検出・定量を行う。定量は、用いた標識に基づき検出したシグナルを、CCR8の標準試料を使用して作成した検量線を利用して数値化すること等、公知の方法により行うことができる。 When the CCR8 detection method of the present invention is an ELISA method, it is carried out, for example, as follows. First, one antibody (capture antibody) is immobilized on the well surface of an ELISA plate. Next, after blocking is performed to prevent nonspecific adsorption to the well surface, a sample is added, and CCR8 in the sample is brought into contact with the antibody to form a complex. After removing proteins that did not bind to the antibody by washing, the other labeled antibody (detection antibody) is added to the well, brought into contact with CCR8 to form a complex, and detection and quantification are performed using the label. Quantification can be performed by a known method, such as quantifying a signal detected based on the label used using a calibration curve prepared using a standard sample of CCR8.
 本発明のモノクローナル抗体又はその抗体断片は、癌の診断薬として使用することができる。例えば、乳癌、子宮体癌、子宮頸癌、卵巣癌、前立腺癌、肺癌、胃癌、膵臓癌、頭頚部癌、食道癌、膀胱癌、メラノーマ、大腸癌、腎癌、非ホジキンリンパ腫、尿路上皮癌、肉腫、血球癌(白血病、リンパ腫等)、胆管癌、胆のう癌、甲状腺癌、精巣癌、胸腺癌、肝臓癌等の診断薬として使用することができ、好ましくは乳癌、卵巣癌、肺癌、胃癌、膵臓癌、頭頚部癌、食道癌、メラノーマ、大腸癌、腎癌、尿路上皮癌の診断薬として使用することができる。 The monoclonal antibody of the present invention or its antibody fragment can be used as a diagnostic agent for cancer. For example, breast cancer, endometrial cancer, cervical cancer, ovarian cancer, prostate cancer, lung cancer, stomach cancer, pancreatic cancer, head and neck cancer, esophageal cancer, bladder cancer, melanoma, colorectal cancer, kidney cancer, non-Hodgkin lymphoma, urothelial cancer. It can be used as a diagnostic agent for cancer, sarcoma, blood cell cancer (leukemia, lymphoma, etc.), cholangiocarcinoma, gallbladder cancer, thyroid cancer, testicular cancer, thymus cancer, liver cancer, etc., preferably breast cancer, ovarian cancer, lung cancer, etc. It can be used as a diagnostic agent for gastric cancer, pancreatic cancer, head and neck cancer, esophageal cancer, melanoma, colon cancer, renal cancer, and urothelial cancer.
 なお、本明細書記載の「癌」は、卵巣癌、胃癌等の上皮性の悪性腫瘍のみならず、慢性リンパ性白血病やホジキンリンパ腫等の造血器がんを含む非上皮性の悪性腫瘍も意味するものとし、本明細書において、「がん(cancer)」、「癌(carcinoma)」、「腫瘍(tumor)」、「新生物(neoplasm)」等の用語は互いに区別されず、相互に交換可能である。 Note that "cancer" as used herein refers not only to epithelial malignant tumors such as ovarian cancer and gastric cancer, but also to non-epithelial malignant tumors including hematopoietic cancers such as chronic lymphocytic leukemia and Hodgkin's lymphoma. In this specification, terms such as "cancer," "carcinoma," "tumor," and "neoplasm" are not distinguished from each other and are used interchangeably. It is possible.
 本発明のモノクローナル抗体又はその抗体断片は、抗CCR8抗体を含有する医薬の投与の適否を判断するための診断薬としても使用することができる。「抗CCR8抗体を含有する医薬」の抗CCR8抗体としては、上記「CCR8の中和抗体」が好ましく、特許文献4~9の実施例に記載された抗CCR8抗体がより好ましく、特許文献4の実施例に記載された抗CCR8抗体がさらに好ましく、配列番号8のアミノ酸配列を有する軽鎖可変領域、及び、配列番号9のアミノ酸配列を有する重鎖可変領域を含む抗体とCCR8に対する結合が競合しない抗CCR8抗体が特に好ましく、配列番号12のアミノ酸配列を有する軽鎖可変領域、及び、配列番号13のアミノ酸配列を有する重鎖可変領域を含む抗CCR8抗体が最も好ましい。
 「抗CCR8抗体を含有する医薬」は抗癌剤として有用であり、例えば、乳癌、子宮体癌、子宮頸癌、卵巣癌、前立腺癌、肺癌、胃癌、膵臓癌、頭頚部癌、食道癌、膀胱癌、メラノーマ、大腸癌、腎癌、非ホジキンリンパ腫、尿路上皮癌、肉腫、血球癌(白血病、リンパ腫等)、胆管癌、胆のう癌、甲状腺癌、精巣癌、胸腺癌、肝臓癌等の癌、好ましくは乳癌、卵巣癌、肺癌、胃癌、膵臓癌、頭頚部癌、食道癌、メラノーマ、大腸癌、腎癌、尿路上皮癌を治療及び/又は予防するための医薬として非常に有用である。
The monoclonal antibody of the present invention or its antibody fragment can also be used as a diagnostic agent to determine the suitability of administering a drug containing an anti-CCR8 antibody. As the anti-CCR8 antibody of the "medicine containing an anti-CCR8 antibody", the above-mentioned "CCR8 neutralizing antibody" is preferable, the anti-CCR8 antibodies described in the Examples of Patent Documents 4 to 9 are more preferable, and the anti-CCR8 antibodies described in the Examples of Patent Documents 4 to 9 are more preferable. More preferred are the anti-CCR8 antibodies described in the Examples, which do not compete in binding to CCR8 with antibodies comprising a light chain variable region having the amino acid sequence of SEQ ID NO: 8 and a heavy chain variable region having the amino acid sequence of SEQ ID NO: 9. Anti-CCR8 antibodies are particularly preferred, and anti-CCR8 antibodies comprising a light chain variable region having the amino acid sequence of SEQ ID NO: 12 and a heavy chain variable region having the amino acid sequence of SEQ ID NO: 13 are most preferred.
"Pharmaceuticals containing anti-CCR8 antibodies" are useful as anticancer agents, such as breast cancer, endometrial cancer, cervical cancer, ovarian cancer, prostate cancer, lung cancer, stomach cancer, pancreatic cancer, head and neck cancer, esophageal cancer, and bladder cancer. , melanoma, colorectal cancer, kidney cancer, non-Hodgkin's lymphoma, urothelial cancer, sarcoma, blood cell cancer (leukemia, lymphoma, etc.), bile duct cancer, gallbladder cancer, thyroid cancer, testicular cancer, thymus cancer, liver cancer, etc. Preferably, it is very useful as a medicine for treating and/or preventing breast cancer, ovarian cancer, lung cancer, gastric cancer, pancreatic cancer, head and neck cancer, esophageal cancer, melanoma, colon cancer, kidney cancer, and urothelial cancer.
 本発明のモノクローナル抗体を含有するキットは、CCR8検出用キットとして使用することができる。該キットは、本発明のモノクローナル抗体又はその抗体断片を含み、さらに、ゲート用抗体、標識二次抗体、構成試薬(希釈液、洗浄液、反応停止液等)、蛍光検出試薬、CCR8標準品、使用説明書等を含んでいてもよい。 A kit containing the monoclonal antibody of the present invention can be used as a CCR8 detection kit. The kit contains the monoclonal antibody of the present invention or an antibody fragment thereof, and further includes a gating antibody, a labeled secondary antibody, constituent reagents (diluting solution, washing solution, reaction stop solution, etc.), a fluorescence detection reagent, a CCR8 standard product, and usage. It may also include instructions, etc.
 構成試薬に含まれるpH緩衝成分としては、特に限定されないが、例えば、リン酸、ホウ酸、酢酸、クエン酸、ギ酸、カコジル酸等の酸又はこれらの塩;グリシン等のアミノ酸;トリスヒドロキシアミノメタン(Tris)、HEPES、MES等のグッドバッファー等が挙げられる。 The pH buffer component contained in the constituent reagents is not particularly limited, but includes, for example, acids such as phosphoric acid, boric acid, acetic acid, citric acid, formic acid, and cacodylic acid, or salts thereof; amino acids such as glycine; trishydroxyaminomethane; (Tris), HEPES, MES, and other good buffers.
 構成試薬に含まれる界面活性剤としては、特に限定されないが、非イオン性界面活性剤が好ましく、エステルエーテル型、エステル型、エーテル型のいずれも用いることができる。より具体的にはTween20、Tween40、Tween80、Triton-X100、ポリオキシエチレン(60)ソルビタンモノステアレート、ポリオキシエチレン(65)ソルビタントリステアレート、ポリオキシエチレン(80)ソルビタンモノオレエート、ポリオキシエチレンアルキルフェニルエーテル、ノニデットP-40、CHAPS等が挙げられる。これらの界面活性剤は、いずれかを単独で用いても良いし、2種以上を組み合わせても良い。 The surfactant contained in the constituent reagents is not particularly limited, but nonionic surfactants are preferred, and any of the ester ether type, ester type, and ether type can be used. More specifically, Tween20, Tween40, Tween80, Triton-X100, polyoxyethylene (60) sorbitan monostearate, polyoxyethylene (65) sorbitan tristearate, polyoxyethylene (80) sorbitan monooleate, polyoxy Examples include ethylene alkylphenyl ether, Nonidet P-40, CHAPS, and the like. These surfactants may be used alone or in combination of two or more.
 本発明は、本発明のモノクローナル抗体の軽鎖可変領域又は重鎖可変領域をコードするポリヌクレオチドを包含する。本発明は、さらに、該ポリヌクレオチドを含む発現ベクターを包含する。 The present invention includes polynucleotides encoding the light chain variable region or heavy chain variable region of the monoclonal antibody of the present invention. The invention further includes expression vectors containing the polynucleotides.
 該ポリヌクレオチドは、本発明のモノクローナル抗体の軽鎖可変領域又は重鎖可変領域をコードする限り、特に限定されず、複数のデオキシリボ核酸(DNA)又はリボ核酸(RNA)等のヌクレオチドからなる重合体である。天然以外の塩基を含んでいてよい。本発明のポリヌクレオチドは、抗体を遺伝子工学的な手法により製造するために使用することができる。また本発明のモノクローナル抗体と同等な機能を有する抗体をスクリーニングするために、プローブとして用いることもできる。即ち本発明のモノクローナル抗体をコードするポリヌクレオチド、又はその一部をプローブとして用い、ハイブリダイゼーション、遺伝子増幅技術(例えばPCR)等の技術により、該ポリヌクレオチドとストリンジェントな条件下でハイブリダイズし、かつ本発明のモノクローナル抗体と同等の活性を有する抗体をコードするDNAを得ることができる。このようなDNAも本発明のポリヌクレオチドに含まれる。 The polynucleotide is not particularly limited as long as it encodes the light chain variable region or heavy chain variable region of the monoclonal antibody of the present invention, and may be a polymer consisting of multiple nucleotides such as deoxyribonucleic acid (DNA) or ribonucleic acid (RNA). It is. It may contain non-natural bases. The polynucleotide of the present invention can be used to produce antibodies by genetic engineering techniques. It can also be used as a probe to screen for antibodies having functions equivalent to the monoclonal antibodies of the present invention. That is, using a polynucleotide encoding the monoclonal antibody of the present invention, or a portion thereof as a probe, hybridize with the polynucleotide under stringent conditions by a technique such as hybridization or gene amplification technique (e.g. PCR), Moreover, DNA encoding an antibody having an activity equivalent to that of the monoclonal antibody of the present invention can be obtained. Such DNA is also included in the polynucleotide of the present invention.
 ハイブリダイゼーション技術(Sambrook,J et al., Molecular Cloning 2nd ed., 9.47-9.58, Cold Spring Harbor Lab. press, 1989)は当業者によく知られた技術である。ハイブリダイゼーションの条件としては、例えば、低ストリンジェントな条件が挙げられる。低ストリンジェントな条件とは、ハイブリダイゼーション後の洗浄において、例えば42℃、0.1×SSC、0.1%SDSの条件であり、好ましくは50℃、0.1×SSC 、0.1%SDSの条件である。より好ましいハイブリダイゼーションの条件としては、高ストリンジェントな条件が挙げられる。高ストリンジェントな条件とは、例えば65℃、5×SSC及び0.1%SDSの条件である。これらの条件において、温度を上げる程に高い相同性を有するポリヌクレオチドが効率的に得られることが期待できる。但し、ハイブリダイゼーションのストリンジェンシーに影響する要素としては温度や塩濃度等複数の要素が考えられ、当業者であればこれら要素を適宜選択することで同様のストリンジェンシーを実現することが可能である。 Hybridization techniques (Sambrook, J et al., Molecular Cloning 2nd ed., 9.47-9.58, Cold Spring Harbor Lab. press, 1989) are well known to those skilled in the art. Examples of hybridization conditions include low stringency conditions. Low stringency conditions include, for example, 42°C, 0.1x SSC, 0.1% SDS in washing after hybridization, preferably 50°C, 0.1x SSC, 0.1%. This is a condition of SDS. More preferable hybridization conditions include highly stringent conditions. High stringency conditions are, for example, 65° C., 5×SSC, and 0.1% SDS. Under these conditions, it can be expected that polynucleotides with higher homology can be obtained more efficiently as the temperature is raised. However, multiple factors such as temperature and salt concentration can be considered as factors that affect the stringency of hybridization, and those skilled in the art will be able to achieve similar stringency by appropriately selecting these factors. .
 これらハイブリダイゼーション技術や遺伝子増幅技術により得られるポリヌクレオチドがコードする、本発明のモノクローナル抗体と機能的に同等な抗体は、通常、これら抗体とアミノ酸配列において高い相同性を有する。本発明のモノクローナル抗体には、本発明のモノクローナル抗体と機能的に同等であり、かつ該抗体のアミノ酸配列と高い相同性を有する抗体も含まれる。高い相同性とは、アミノ酸レベルにおいて、通常、少なくとも75%以上の同一性、好ましくは85%以上の同一性、さらに好ましくは95%以上の同一性を指す。ポリペプチドの相同性を決定するには、文献(Wilbur, W. J. and Lipman, D. J. Proc. Natl. Acad. Sci. USA, (1983), 80, 726-730)記載のアルゴリズムに従えばよい。 Antibodies that are functionally equivalent to the monoclonal antibodies of the present invention and encoded by polynucleotides obtained by these hybridization techniques or gene amplification techniques usually have high homology with these antibodies in their amino acid sequences. The monoclonal antibodies of the present invention also include antibodies that are functionally equivalent to the monoclonal antibodies of the present invention and have high homology with the amino acid sequence of the antibodies. High homology generally refers to at least 75% identity, preferably 85% or more identity, and more preferably 95% or more identity at the amino acid level. To determine polypeptide homology, an algorithm described in the literature (Wilbur, W. J. and Lipman, D. J. Proc. Natl. Acad. Sci. USA, (1983), 80, 726-730) was used. Just follow.
 本発明のモノクローナル抗体又はその抗体断片は、腫瘍内浸潤Treg細胞除去作用や腫瘍細胞除去作用を有していてもよい。本発明のモノクローナル抗体が腫瘍内浸潤Treg細胞除去作用を有しているか否かは、例えば、特許文献2の実施例記載の方法にて測定することができる。 The monoclonal antibody of the present invention or an antibody fragment thereof may have an effect of removing intratumoral infiltrating Treg cells or a tumor cell. Whether or not the monoclonal antibody of the present invention has an effect of removing tumor-infiltrating Treg cells can be determined, for example, by the method described in Examples of Patent Document 2.
 本発明のモノクローナル抗体又はその抗体断片は、医薬組成物にも有用である。本発明のモノクローナル抗体又はその抗体断片を含有する医薬組成物は、経口的又は非経口的に全身あるいは局所的に投与することができる。非経口的投与としては、例えば、点滴などの静脈内注射、筋肉内注射、腹腔内注射、皮下注射、鼻腔内投与、吸入などを選択することができる。 The monoclonal antibody or antibody fragment thereof of the present invention is also useful in pharmaceutical compositions. Pharmaceutical compositions containing the monoclonal antibodies of the present invention or antibody fragments thereof can be administered orally or parenterally, systemically or locally. As parenteral administration, for example, intravenous injection such as drip, intramuscular injection, intraperitoneal injection, subcutaneous injection, intranasal administration, inhalation, etc. can be selected.
 本発明のモノクローナル抗体又はその抗体断片を含有する医薬組成物は、CCR8関連疾患の治療及び/又は予防のための医薬として非常に有用である。特に、CCR8発現Treg細胞の腫瘍内浸潤が生じている癌を治療及び/又は予防するための医薬として非常に有用である。例えば、乳癌、子宮体癌、子宮頸癌、卵巣癌、前立腺癌、肺癌、胃癌、膵臓癌、頭頚部癌、食道癌、膀胱癌、メラノーマ、大腸癌、腎癌、非ホジキンリンパ腫、尿路上皮癌、肉腫、血球癌(白血病、リンパ腫等)、胆管癌、胆のう癌、甲状腺癌、精巣癌、胸腺癌、肝臓癌等の癌、好ましくは乳癌、卵巣癌、肺癌、胃癌、膵臓癌、頭頚部癌、食道癌、メラノーマ、大腸癌、腎癌、尿路上皮癌を治療及び/又は予防するための医薬として非常に有用である。 A pharmaceutical composition containing the monoclonal antibody or antibody fragment thereof of the present invention is very useful as a medicament for the treatment and/or prevention of CCR8-related diseases. In particular, it is very useful as a medicament for treating and/or preventing cancer in which CCR8-expressing Treg cells infiltrate into tumors. For example, breast cancer, endometrial cancer, cervical cancer, ovarian cancer, prostate cancer, lung cancer, stomach cancer, pancreatic cancer, head and neck cancer, esophageal cancer, bladder cancer, melanoma, colorectal cancer, kidney cancer, non-Hodgkin lymphoma, urothelial cancer. Cancer, sarcoma, blood cell cancer (leukemia, lymphoma, etc.), bile duct cancer, gallbladder cancer, thyroid cancer, testicular cancer, thymus cancer, liver cancer, etc., preferably breast cancer, ovarian cancer, lung cancer, stomach cancer, pancreatic cancer, head and neck cancer. It is very useful as a medicine for treating and/or preventing cancer, esophageal cancer, melanoma, colon cancer, renal cancer, and urothelial cancer.
 以下に本発明の実施例を挙げて本発明をさらに詳しく説明するが、本発明はこれらにより限定されるものではない。 The present invention will be explained in more detail with reference to Examples below, but the present invention is not limited thereto.
実施例1:抗ヒトCCR8抗体産生ラットハイブリドーマの作製
(1)ハイブリドーマの作製
 免疫原であるヒトCCR8発現Rat-1細胞は、pQCXIP(クロンテック社)に
ヒトCCR8遺伝子をクローニングした発現ベクターをRat-1細胞にトランスフェクションしたのちに、ピューロマイシン(1μg/ml)で一ヶ月間薬剤選択することにより作製した。
 ヒトCCR8発現Rat-1細胞をラットに一回免疫し、約2週間後にリンパ節を回収して常法によりハイブリドーマを作製した。
Example 1: Preparation of rat hybridoma producing anti-human CCR8 antibody (1) Preparation of hybridoma Human CCR8-expressing Rat-1 cells, which are the immunogen, were prepared using an expression vector in which the human CCR8 gene was cloned into pQCXIP (Clontech). The cells were transfected, followed by drug selection with puromycin (1 μg/ml) for one month.
Rats were immunized once with human CCR8-expressing Rat-1 cells, and about two weeks later, lymph nodes were collected and hybridomas were produced by a conventional method.
(2)クローンの選抜
1)CCR8結合活性評価
 上記(1)で得られた4種のハイブリドーマ(No.3、No.12、No.30、No.37)の上清を用いて、ヒトCCR8発現293細胞及び、親細胞の293細胞に対する結合を評価した。二次抗体としてAlexa647標識抗ラットIgG抗体(サーモフィッシャーサイエンティフィック社製)を使用した。ハイブリドーマの上清とヒトCCR8発現293細胞又は親細胞の293細胞を混合し、4℃で20分間反応させ、FACSバッファーで3回洗浄した。二次抗体を30000倍希釈で添加し、4℃で20分間反応させ、FACSバッファーで3回洗浄した。DAPI(4’,6-ジアミジノ-2-フェニルインドール)を100ng/mlとなるように添加し、フローサイトメトリーで細胞に結合した蛍光値(平均値)を定量した。
 その結果を表1に示す。No.30、No.3、No.12、No.37のハイブリドーマ上清の順にヒトCCR8に対する結合が強かった。また、親細胞に対する結合はNo.3が最も結合が低く、S/N比(シグナル対ノイズ比)はNo.3がもっとも高かった。
 次に、抗ヒトCCR8抗体(L263G8、BioLegend社)に対する競合阻害を評価した。上記4種のハイブリドーマ上清又は比較対照としてWO2020/138489号公報の実施例に記載された2-7B抗体を産生するハイブリドーマ(No.2)上清をヒトCCR8発現293細胞に添加し、4℃で30分間静置した。陽性コントロールとして、ハイブリドーマ上清の代わりにGIT培地で希釈した未標識抗ヒトCCR8抗体(L263G8、BioLegend社)を2μg/mlの濃度で使用した。PE標識抗ヒトCCR8抗体(L263G8、BioLegend社)を0.2μg/mlの濃度で添加し、30分間4℃静置した。その後、FACSバッファーで3回洗浄した。DAPIを100ng/mlとなるように添加し、フローサイトメトリーで細胞に結合した蛍光値(平均値)を定量した。陰性コントロールとしてGIT培地を添加した場合の蛍光平均値を100%とし、陽性コントロールの蛍光平均値を0%として、各試験抗体の蛍光平均値から、阻害率(%)を算出した。
 その結果を表1及び図1に示す。No.2のハイブリドーマ上清はL263G8抗体に対する競合阻害活性を示したのに対し、上記(1)で得られた4種のハイブリドーマ上清はL263G8抗体に対する競合阻害活性をほとんど示さなかった。
(2) Clone selection 1) CCR8 binding activity evaluation Using the supernatants of the four hybridomas (No. 3, No. 12, No. 30, and No. 37) obtained in (1) above, human CCR8 Binding to expressing 293 cells and parental cells to 293 cells was evaluated. Alexa647-labeled anti-rat IgG antibody (manufactured by Thermo Fisher Scientific) was used as a secondary antibody. The hybridoma supernatant and human CCR8-expressing 293 cells or parental 293 cells were mixed, reacted at 4°C for 20 minutes, and washed three times with FACS buffer. A secondary antibody was added at a 30,000-fold dilution, reacted at 4°C for 20 minutes, and washed three times with FACS buffer. DAPI (4',6-diamidino-2-phenylindole) was added at a concentration of 100 ng/ml, and the fluorescence value (average value) bound to the cells was quantified by flow cytometry.
The results are shown in Table 1. No. 30, No. 3.No. 12, No. The 37 hybridoma supernatants showed the strongest binding to human CCR8. In addition, binding to parent cells was No. 3 has the lowest coupling, and the S/N ratio (signal to noise ratio) is No. 3 was the highest.
Next, competitive inhibition against anti-human CCR8 antibody (L263G8, BioLegend) was evaluated. The above four hybridoma supernatants or the hybridoma supernatant (No. 2) producing the 2-7B antibody described in the Examples of WO2020/138489 as a comparison were added to human CCR8-expressing 293 cells, and the mixture was incubated at 4°C. It was left standing for 30 minutes. As a positive control, an unlabeled anti-human CCR8 antibody (L263G8, BioLegend) diluted in GIT medium was used at a concentration of 2 μg/ml instead of the hybridoma supernatant. PE-labeled anti-human CCR8 antibody (L263G8, BioLegend) was added at a concentration of 0.2 μg/ml, and the mixture was allowed to stand at 4° C. for 30 minutes. Thereafter, it was washed three times with FACS buffer. DAPI was added at a concentration of 100 ng/ml, and the fluorescence value (average value) bound to the cells was quantified by flow cytometry. The inhibition rate (%) was calculated from the average fluorescence value of each test antibody, with the average fluorescence value when GIT medium was added as a negative control being 100% and the average fluorescence value of the positive control being 0%.
The results are shown in Table 1 and FIG. No. The hybridoma supernatant of No. 2 exhibited competitive inhibitory activity against the L263G8 antibody, whereas the four hybridoma supernatants obtained in (1) above showed almost no competitive inhibitory activity against the L263G8 antibody.
Figure JPOXMLDOC01-appb-T000001
Figure JPOXMLDOC01-appb-T000001

2)中和活性評価
 上記(1)で得られた4種のハイブリドーマ上清を用いて、ヒトCCL1によるヒトCCR8を介したヒトCCR8発現293細胞内カルシウム流入活性に対する阻害活性評価を実施した。ヒトCCR8発現293細胞をトリプシンで剥離し、常法に従いFluo3-AM(同人化学社)で37℃、60分間細胞を処理することで、細胞にFluo3を取り込ませた。その後DMEM/10%FBSで2回洗浄した。600μLのDMEM/10%FBSに懸濁した上記ラベルされた細胞に各ハイブリドーマ上清300μLを添加し、30分間室温で静置した。陰性コントロールはGIT培地を使用した。なお、比較対照とする中和活性を有する抗CCR8抗体を産生するハイブリドーマとして、WO2020/138489号公報の実施例に記載された2-7B抗体を産生するハイブリドーマ(No.2)上清を用いた。その後ヒトCCL1(BioLegend社)を終濃度5nMになるように添加し、F-7000形分光蛍光光度計(HITACHI)を用いて励起波長: 508.0 nm、 蛍光波長: 527.0 nmで、経時的に細胞内カルシウム濃度を5分間測定した。定量化方法は、ヒトCCL1を添加後3秒後の値をバックグラウンド値とし、カルシウムの蛍光値の最大値をMaxとし、以下の計算式で%阻害を算出した。
100-(各ITM抗体max-各ITM抗体のBG)/(ネガコンのMax-ネガコンのBG)×100 (%)
 その結果を表2及び図2に示す。なお、No.2及びNo.3に対応する陰性コントロールは左列下のコントロールが対応し、No.30、No.12及びNo.37に対応する陰性コントロールは右列下のコントロールが対応している。No.3、No.30、No.12、No.37のハイブリドーマ上清はいずれもヒトCCL1に対する中和活性をほとんど示さず、この順に中和活性が上昇した。No.3の上清がもっともヒトCCL1に対する中和活性が低かった。
2) Evaluation of neutralizing activity Using the four types of hybridoma supernatants obtained in (1) above, an evaluation of the inhibitory activity of human CCL1 against human CCR8-mediated calcium influx activity into human CCR8-expressed 293 cells was performed. Human CCR8-expressing 293 cells were detached with trypsin, and the cells were treated with Fluo3-AM (Dojin Kagakusha) for 60 minutes at 37°C to incorporate Fluo3 into the cells. Thereafter, it was washed twice with DMEM/10% FBS. 300 μL of each hybridoma supernatant was added to the labeled cells suspended in 600 μL of DMEM/10% FBS, and the cells were allowed to stand at room temperature for 30 minutes. GIT medium was used as a negative control. As a comparison control hybridoma producing an anti-CCR8 antibody with neutralizing activity, the supernatant of a hybridoma (No. 2) producing the 2-7B antibody described in the Examples of WO2020/138489 was used. . Thereafter, human CCL1 (BioLegend) was added to a final concentration of 5 nM, and the mixture was incubated at an excitation wavelength of 508.0 nm and a fluorescence wavelength of 527.0 nm using an F-7000 spectrofluorometer (HITACHI) over time. Intracellular calcium concentration was measured for 5 minutes. In the quantification method, the value 3 seconds after addition of human CCL1 was taken as the background value, the maximum value of the fluorescence value of calcium was taken as Max, and % inhibition was calculated using the following formula.
100 - (Max of each ITM antibody - BG of each ITM antibody) / (Max of negative control - BG of negative control) x 100 (%)
The results are shown in Table 2 and FIG. In addition, No. 2 and no. The negative control corresponding to No. 3 corresponds to the control at the bottom of the left column. 30, No. 12 and no. The negative control corresponding to No. 37 corresponds to the control at the bottom of the right column. No. 3.No. 30, No. 12, No. All 37 hybridoma supernatants showed almost no neutralizing activity against human CCL1, and the neutralizing activity increased in this order. No. The supernatant of No. 3 had the lowest neutralizing activity against human CCL1.
Figure JPOXMLDOC01-appb-T000002
Figure JPOXMLDOC01-appb-T000002

 上記(1)で得られた4種のハイブリドーマのうちで、No.3クローンはヒトCCR8の結合活性が2番目に強く、S/N比が最も高く、ヒトCCL1によるヒトCCR8を介した細胞内カルシウム流入に対する中和活性が最も低い抗体であった。以上より、中和活性を有さない抗CCR8抗体を産生するハイブリドーマとして、もっとも優れているNo.3クローンを選抜した。 Among the four hybridomas obtained in (1) above, No. The three clones were antibodies with the second strongest binding activity to human CCR8, the highest S/N ratio, and the lowest neutralizing activity against intracellular calcium influx via human CCR8 by human CCL1. From the above, No. 1 is the best hybridoma that produces anti-CCR8 antibodies without neutralizing activity. Three clones were selected.
実施例2:抗体配列の決定
 No.3クローンが産生する3-3F抗体について、ハイブリドーマ細胞より、常法に従って抗体の軽鎖可変領域及び重鎖可変領域のアミノ酸配列を決定した(表3)。
Example 2: Determination of antibody sequence No. Regarding the 3-3F antibody produced by the three clones, the amino acid sequences of the light chain variable region and heavy chain variable region of the antibody were determined from hybridoma cells according to a conventional method (Table 3).
Figure JPOXMLDOC01-appb-T000003
Figure JPOXMLDOC01-appb-T000003

実施例3:精製抗体の中和活性評価
 樹立したハイブリドーマを無血清培地で培養し、その培養上清をProteinGアフィニティー精製及びゲル濾過精製を行うことで、精製抗体を得た。この精製3-3F抗体について、下記の方法で中和活性を測定した。なお、比較対照とする中和活性を有する抗CCR8抗体として、市販抗ヒトCCR8抗体であるL263G8抗体ならびに、WO2020/138489号公報の実施例に記載された2C7抗体、10A11抗体及び2-7B抗体を用いた。
 あらかじめCa2+指示薬を取り込ませたヒトCCR8発現293細胞に対して培地で希釈した抗体希釈液を添加し、FLIPRにより200nMのヒトCCL1(バイオレジェンド社製)添加によるCa2+流入を測定した。ヒトCCL1非添加時のシグナルを阻害率100%、ヒトCCL1添加及び抗体非添加時のシグナルを阻害率0%として阻害率を計算し、50%の阻害率を示す抗体濃度をIC50とした。少なくとも3回評価を行い、IC50をAverage±SDとして表記した。
 その結果、精製した3-3F抗体にも中和活性が認められなかった(表4)。
Example 3: Evaluation of neutralizing activity of purified antibody The established hybridoma was cultured in a serum-free medium, and the culture supernatant was purified by Protein G affinity and gel filtration to obtain a purified antibody. The neutralizing activity of this purified 3-3F antibody was measured by the method described below. In addition, as anti-CCR8 antibodies with neutralizing activity for comparison, L263G8 antibody, which is a commercially available anti-human CCR8 antibody, and 2C7 antibody, 10A11 antibody, and 2-7B antibody described in the Examples of WO2020/138489 were used. Using.
A diluted antibody solution diluted with a medium was added to human CCR8-expressing 293 cells into which a Ca 2+ indicator had been incorporated in advance, and Ca 2+ influx due to the addition of 200 nM human CCL1 (manufactured by Biolegend) was measured by FLIPR. The inhibition rate was calculated by setting the signal when human CCL1 was not added as an inhibition rate of 100%, and the signal when human CCL1 was added and the antibody was not added as an inhibition rate of 0%, and the antibody concentration showing a 50% inhibition rate was defined as IC50. Evaluations were performed at least three times and IC50 was expressed as Average±SD.
As a result, no neutralizing activity was observed in the purified 3-3F antibody (Table 4).
Figure JPOXMLDOC01-appb-T000004
Figure JPOXMLDOC01-appb-T000004

実施例4:CCR8に対する結合解析
(1)CCR8に対する結合活性評価
 ヒトCCR8発現Ramos細胞及びその親株(Ramos細胞)に対する3-3F抗体の結合及び、ヒトCCR8に対する中和抗体であるOP-1抗体(WO2020/138489号公報の実施例に記載されたヒト化10A11抗体(軽鎖可変領域:IGKV4-1 N53Q+G29R(配列番号12)/重鎖可変領域:IGHV3-15 T94R(配列番号13)))存在下の3-3F抗体の結合について評価した。
 本実施例では、3-3F抗体の蛍光非標識体およびAlexa647蛍光標識体を用いた。蛍光非標識体の検出には二次抗体としてAPC蛍光標識抗ラットIgG抗体(Jackson ImmunoResearch Laboratories社製)を使用した。比較対照のため、市販抗ヒトCCR8抗体であるL263G8抗体のAPC蛍光標識体および433H抗体(BDバイオサイエンス社製)のBV421蛍光標識体を使用した。ヒトCCR8発現Ramos細胞およびRamos細胞を6.0×10細胞ずつマイクロチューブへ分注し、(A)3-3F抗体の蛍光非標識体を5μg/mLを添加し、4℃で30分間反応させた。アイソタイプ抗体としてはRat IgG2a抗体(未標識、 100倍希釈)を使用した。その後、培養液(10%FBS)で洗浄後、APC蛍光標識抗ラットIgG抗体を加え、4℃で30分間反応させた。(B)L263G8抗体のAPC蛍光標識体(100倍希釈)を添加し、4℃で30分間反応させた。アイソタイプ抗体としてはAPC-mouse IgG2a抗体(100倍希釈)を使用した。(C)433H抗体のBV421蛍光標識体(100倍希釈)を添加し、4℃で30分間反応させたアイソタイプ抗体としてはBV421-mouse IgG2a抗体(100倍希釈)を使用した。(A)及び(B)の反応後、培養液(10%FBS)で洗浄して、Cellstain-DAPI solution(10000倍希釈、同仁化学研究所社製)を加えて、フローサイトメトリーで測定した。(C)の反応後、eBioscience Fixable Viability Dye eFluor 780溶液(10000倍希釈、Thermo Fisher Scientific社製)を加えて、4℃で10分間反応させた後、培養液(10%FBS)で洗浄して、培養液(10%FBS)で再懸濁後、フローサイトメトリーで測定した。また、競合試験においては、マイクロチューブに分注したヒトCCR8発現Ramos細胞にOP-1抗体を各種濃度(1,10,100μg/mL)で添加し、4℃で30分間反応させ、培養液(10%FBS)で洗浄後に上記(A)、(B)及び(C)の抗体反応を行った。
 その結果、ヒトCCR8発現Ramos細胞において3-3F抗体、L263G8抗体及び433H抗体の結合が認められた(図3)。また、OP-1抗体存在下での各抗体の反応性についても確認したところ、3-3F抗体については、OP-1抗体存在下で結合の減弱が認めらなかった。(図4)
Example 4: Binding analysis to CCR8 (1) Evaluation of binding activity to CCR8 Binding of 3-3F antibody to human CCR8-expressing Ramos cells and its parent strain (Ramos cells), and OP-1 antibody (neutralizing antibody to human CCR8) In the presence of the humanized 10A11 antibody (light chain variable region: IGKV4-1 N53Q+G29R (SEQ ID NO: 12)/heavy chain variable region: IGHV3-15 T94R (SEQ ID NO: 13)) described in the Examples of WO2020/138489. The binding of the 3-3F antibody was evaluated.
In this example, a non-fluorescently labeled 3-3F antibody and a fluorescently labeled Alexa647 antibody were used. APC fluorescently labeled anti-rat IgG antibody (manufactured by Jackson ImmunoResearch Laboratories) was used as a secondary antibody to detect the non-fluorescently labeled substance. For comparison purposes, APC fluorescently labeled L263G8 antibody and BV421 fluorescently labeled 433H antibody (manufactured by BD Biosciences), which are commercially available anti-human CCR8 antibodies, were used. Dispense human CCR8-expressing Ramos cells and Ramos cells at 6.0 x 10 5 cells into microtubes, add 5 μg/mL of (A) non-fluorescent labeled 3-3F antibody, and react at 4°C for 30 minutes. I let it happen. Rat IgG2a antibody (unlabeled, 100-fold diluted) was used as the isotype antibody. Thereafter, after washing with culture medium (10% FBS), APC fluorescently labeled anti-rat IgG antibody was added and reacted at 4°C for 30 minutes. (B) APC fluorescently labeled L263G8 antibody (100-fold dilution) was added and reacted at 4°C for 30 minutes. APC-mouse IgG2a antibody (100-fold dilution) was used as the isotype antibody. (C) BV421 fluorescently labeled 433H antibody (100 times diluted) was added and reacted at 4° C. for 30 minutes. BV421-mouse IgG2a antibody (100 times diluted) was used as the isotype antibody. After the reactions in (A) and (B), the cells were washed with culture solution (10% FBS), Cellstain-DAPI solution (10,000-fold dilution, manufactured by Dojindo Laboratories) was added, and the cells were measured by flow cytometry. After the reaction in (C), eBioscience Fixable Viability Dye eFluor 780 solution (10,000-fold dilution, manufactured by Thermo Fisher Scientific) was added, reacted at 4°C for 10 minutes, and then washed with culture medium (10% FBS). , resuspended in culture medium (10% FBS) and measured by flow cytometry. In the competition test, OP-1 antibody was added at various concentrations (1, 10, 100 μg/mL) to human CCR8-expressing Ramos cells dispensed into microtubes, reacted at 4°C for 30 minutes, and culture medium ( After washing with 10% FBS), the antibody reactions in (A), (B), and (C) above were performed.
As a result, binding of the 3-3F antibody, L263G8 antibody, and 433H antibody was observed in human CCR8-expressing Ramos cells (FIG. 3). Furthermore, when the reactivity of each antibody in the presence of OP-1 antibody was confirmed, no decrease in binding was observed for 3-3F antibody in the presence of OP-1 antibody. (Figure 4)
(2)ケモカイン受容体における結合選択性評価
 表5の各ケモカイン受容体について、それぞれ対応するアクセッション番号のcDNA配列を組み込んだ発現プラスミドを2又は6μg用いて、Lipofectamine LTX又はLipofectamine 3000(いずれもThermo Fisher Scientific社)により293c18細胞にトランスフェクションを行い、37℃、5%COで72時間培養して、ケモカイン受容体発現細胞を得た。
 CCR3発現細胞については、配列番号16に示すヒトCCR3のcDNA配列を用い、上記と同様の方法で発現細胞を得た。
 なお、ヒトCCR8発現細胞については、以下の方法で安定発現細胞株を得た。pQCXIPベクターのEcoRI/BamHIサイトにヒトCCR8のcDNA(配列番号17)をクローニングし、pQCXIP-hCCR8発現ベクターを作製した。この発現ベクター5μgをlipofectamine3000によりHEK293T細胞(TAKARA)に形質転換し、2日後よりピューロマイシン2μg/mlで薬剤選択した。薬剤存在下で3週間、37℃、5%COで培養し、生き残った薬剤耐性HEK293T細胞の細胞表面上に発現するヒトCCR8を、PE標識抗CCR8抗体(BioLegend)を用いてフローサイトメーターにより発現確認し、安定発現細胞hCCR8/HEK293Tを樹立した。
 上記ケモカイン受容体発現細胞に死細胞染色試薬(Zombie NIR Fixable Viability Dye、BioLegend社)を2000倍希釈で添加後、検出抗体としてAF647標識3-3F抗体又はAF647標識rat IgG2a抗体(#407512)を500ng/mLの濃度で添加し、4℃で30分間静置した。その後Stain Buffer(BD Pharmingen)で2回洗浄した。Novocyte3000を用いて各抗体の蛍光を測定し、それぞれの陰性コントロール抗体のヒストグラムとオーバーレイした。なお、各ケモカイン受容体発現細胞におけるケモカイン受容体の発現確認のため、上記と同様の方法(ただし、検出抗体として、各ケモカイン受容体の抗体(表6)又は対応する陰性コントロール抗体(表7)を40倍希釈で添加)で解析したところ、各ケモカイン受容体発現細胞においてケモカイン受容体が正常に発現していることを確認した。
 その結果、CCR8についてはAF647標識3-3F抗体のピークが陰性コントロール抗体のピークからシフトしたが、その他のケモカイン受容体について両ピークが重なったことから、AF647標識3-3F抗体のCCR8以外のケモカイン受容体への結合は認められなかった(図5)。







(2) Evaluation of binding selectivity in chemokine receptors For each chemokine receptor in Table 5, 2 or 6 μg of an expression plasmid incorporating the cDNA sequence with the corresponding accession number was used to inject Lipofectamine LTX or Lipofectamine 3000 (both with Thermo (Fisher Scientific), 293c18 cells were transfected and cultured at 37° C. and 5% CO 2 for 72 hours to obtain chemokine receptor-expressing cells.
CCR3-expressing cells were obtained in the same manner as above using the human CCR3 cDNA sequence shown in SEQ ID NO: 16.
In addition, regarding human CCR8 expressing cells, a stable expressing cell line was obtained by the following method. Human CCR8 cDNA (SEQ ID NO: 17) was cloned into the EcoRI/BamHI site of the pQCXIP vector to create a pQCXIP-hCCR8 expression vector. 5 μg of this expression vector was transformed into HEK293T cells (TAKARA) using lipofectamine 3000, and after 2 days drug selection was performed using 2 μg/ml of puromycin. Human CCR8 expressed on the cell surface of surviving drug-resistant HEK293T cells cultured in the presence of drugs for 3 weeks at 37°C and 5% CO was detected using a flow cytometer using a PE-labeled anti-CCR8 antibody (BioLegend). Expression was confirmed and stable expressing cell hCCR8/HEK293T was established.
After adding a dead cell staining reagent (Zombie NIR Fixable Viability Dye, BioLegend) to the chemokine receptor-expressing cells at a 2000-fold dilution, 500 ng of AF647-labeled 3-3F antibody or AF647-labeled rat IgG2a antibody (#407512) was added as a detection antibody. /mL and left at 4°C for 30 minutes. Thereafter, it was washed twice with Stain Buffer (BD Pharmingen). Fluorescence of each antibody was measured using Novocyte 3000 and overlaid with the histogram of each negative control antibody. In addition, to confirm the expression of chemokine receptors in each chemokine receptor-expressing cell, the same method as above (however, as a detection antibody, the antibody for each chemokine receptor (Table 6) or the corresponding negative control antibody (Table 7)) (added at a 40-fold dilution), it was confirmed that chemokine receptors were normally expressed in each chemokine receptor-expressing cell.
As a result, for CCR8, the peak of AF647-labeled 3-3F antibody shifted from the peak of the negative control antibody, but for other chemokine receptors, both peaks overlapped. No binding to the receptor was observed (Figure 5).







Figure JPOXMLDOC01-appb-T000005
Figure JPOXMLDOC01-appb-T000005





























Figure JPOXMLDOC01-appb-T000006
Figure JPOXMLDOC01-appb-T000006











Figure JPOXMLDOC01-appb-T000007
Figure JPOXMLDOC01-appb-T000007

実施例5:ヒト腫瘍内浸潤細胞に対する3-3F抗体の結合評価
 ヒト腫瘍内浸潤細胞に対する3-3F抗体の結合について、卵巣癌患者検体を用いて評価した。本実施例では、3-3F抗体の蛍光非標識体およびAlexa647蛍光標識体を用いた。蛍光非標識体の検出には二次抗体としてAlexa647蛍光標識抗ラットIgG抗体(Jackson ImmunoResearch Laboratories社製)を使用した。比較対照のため、陽性コントロールとして市販抗ヒトCCR8抗体であるL263G8抗体のAPC蛍光標識体を使用した。
 卵巣癌より抽出した腫瘍内浸潤細胞を1.5×10細胞ずつ96ウェルプレートへ播種し、Zombie NIR Fixable Viability Kit(Biolegend社製)を添加し室温で15分反応させた後、実施例4で用いたヒトCCR8に対する中和抗体であるOP-1抗体を各種濃度で添加し、4℃で60分間反応させた。HBSS(2%FBS+10mmol/L Hepes)で洗浄後、Human TruStain FcX(BioLegend社製)を添加し、室温で15分反応させた後、各表面抗原(CD45、CD45RA、CD3、CD4、CD8、CD25及びCCR8)に対する抗体を加えて4℃で30分間反応させた。HBSSで洗浄後、3-3F抗体の蛍光非標識体を反応させたウェルにAlexa647蛍光標識抗ラットIgG抗体を加え、4℃で30分間反応させた。HBSSで洗浄後、Foxp3/Transcription Factor Staining Buffer Set(Thermo Fisher Scientific社製)及びFoxP3に対する抗体を用いて、細胞内FoxP3染色を行った。同Buffer SetのPermeabilization Bufferで洗浄後、フローサイトメトリーで測定した。ゲート方法は、リンパ球をゲート後、CD45陽性細胞ゲート、CD3ゲート、CD4をゲートした後に、CD45RAとFoxP3の2軸で展開し、CD45RA陰性FoxP3陽性細胞(Treg細胞)を解析対象とした。
 その結果を図6に示す。蛍光標識二次抗体のみで染色した細胞を陰性対照とした際に、3-3F抗体において、Treg細胞中の61.4%が陽性シグナルとして検出された。
 また、CD4をゲートするまでを前述したゲート方法と同じ手法で行い、CD4でゲートした細胞をCCR8とFoxP3の2軸で展開し、蛍光標識体及び蛍光非標識体の染色性を比較した。
 その結果を図7に示す。FoxP3陽性CCR8陽性分画の陽性率は、蛍光非標識体2μg/mLの反応で54.1%、蛍光標識体2μg/mL反応で47.8%となり、蛍光標識体においても蛍光非標識体と同等の染色パターンを示した。また、比較対照として用いたL236G8抗体2μg/mLの反応において、FoxP3陽性CCR8陽性分画の陽性率は、46.6%であった。OP-1抗体存在下での各抗体の反応性についても確認したところ、3-3F抗体については、OP-1抗体存在下で結合の減弱が認めらなかった。比較対照として用いたL236G8抗体については、OP-1抗体存在下で結合の減弱が認められ、FoxP3陽性CCR8陽性分画の陽性率は、約半分に低下した。
Example 5: Evaluation of binding of 3-3F antibody to human tumor-infiltrating cells The binding of 3-3F antibody to human tumor-infiltrating cells was evaluated using ovarian cancer patient specimens. In this example, a non-fluorescently labeled 3-3F antibody and a fluorescently labeled Alexa647 antibody were used. Alexa647 fluorescently labeled anti-rat IgG antibody (manufactured by Jackson ImmunoResearch Laboratories) was used as a secondary antibody to detect the unlabeled fluorescent substance. For comparison, an APC fluorescent label of L263G8 antibody, which is a commercially available anti-human CCR8 antibody, was used as a positive control.
Intratumoral infiltrating cells extracted from ovarian cancer were seeded into a 96-well plate at a rate of 1.5 x 10 5 cells, and Zombie NIR Fixable Viability Kit (manufactured by Biolegend) was added and reacted at room temperature for 15 minutes, followed by Example 4. OP-1 antibody, which is a neutralizing antibody against human CCR8 used in , was added at various concentrations and allowed to react at 4°C for 60 minutes. After washing with HBSS (2% FBS + 10 mmol/L Hepes), Human TruStain FcX (manufactured by BioLegend) was added, and after reacting for 15 minutes at room temperature, each surface antigen (CD45, CD45RA, CD3, CD4, CD8, CD25 and CCR8) was added and allowed to react at 4°C for 30 minutes. After washing with HBSS, Alexa647 fluorescently labeled anti-rat IgG antibody was added to the wells in which the non-fluorescently labeled 3-3F antibody had been reacted, and the wells were allowed to react at 4°C for 30 minutes. After washing with HBSS, intracellular FoxP3 staining was performed using Foxp3/Transcription Factor Staining Buffer Set (manufactured by Thermo Fisher Scientific) and an antibody against FoxP3. After washing with the Permeabilization Buffer of the same Buffer Set, measurement was performed by flow cytometry. In the gating method, after gating lymphocytes, gating CD45-positive cells, CD3 gate, and gating CD4, two axes of CD45RA and FoxP3 were used, and CD45RA-negative FoxP3-positive cells (Treg cells) were targeted for analysis.
The results are shown in FIG. When cells stained only with a fluorescently labeled secondary antibody were used as a negative control, 61.4% of Treg cells were detected as positive signals with the 3-3F antibody.
In addition, the steps up to gating CD4 were performed using the same method as the gating method described above, and the cells gated by CD4 were developed on two axes of CCR8 and FoxP3, and the stainability of fluorescently labeled and non-fluorescently labeled cells was compared.
The results are shown in FIG. The positive rate of the FoxP3-positive CCR8-positive fraction was 54.1% in the reaction with 2 μg/mL of the unlabeled fluorescent substance and 47.8% in the reaction with 2 μg/mL of the fluorescent label; showed comparable staining patterns. Furthermore, in the reaction with 2 μg/mL of L236G8 antibody used as a comparison control, the positive rate of the FoxP3-positive CCR8-positive fraction was 46.6%. When the reactivity of each antibody in the presence of OP-1 antibody was also confirmed, no decrease in binding was observed for 3-3F antibody in the presence of OP-1 antibody. Regarding the L236G8 antibody used as a comparison control, a decrease in binding was observed in the presence of the OP-1 antibody, and the positivity rate of the FoxP3-positive CCR8-positive fraction was reduced to about half.
実施例6:3-3F抗体の腫瘍内浸潤Treg細胞中へのCCR8特異的結合の確認
 PrimeFlow RNAアッセイ技術を用い、フローサイトメトリー上で、3-3F抗体によるシグナルとCCR8のmRNA 発現を同時に検出し、3-3F抗体のCCR8特異性を確認した。
 卵巣癌患者由来腫瘍組織をTumor Dissociation Kit, human(Miltenyi Biotec社)を用いて分散し、凍結保存した。凍結した腫瘍分散細胞を融解し、Human TruStain FcX(BioLegend社)およびZombie NIR Fixable Viability Kit(Biolegend社)を添加したStain Buffer(BD Bioscience社)に懸濁し、4℃に30分間静置した。洗浄後、3-3F抗体(4μg/mL)を添加して4℃に30分間静置した。洗浄後、ヒトCD45、CD3及びCD8に対する蛍光標識抗体とAlexaFlour488標識抗ラットIgG抗体を添加して4℃に30分間静置した。洗浄後、PrimeFlow RNAアッセイキット(サーモフィッシャー社)の試薬にて固定および膜透過処理後、PE標識抗ヒトFoxp3抗体を添加して4℃に30分間静置した。洗浄後、CCR8 mRNAに対するプローブを添加して40℃に120分間静置した。洗浄後、シグナル増幅用プローブPreamplifierおよびAmplifierをそれぞれ40℃ 90分にてハイブリダイゼーション後、AlexaFlour647標識プローブを添加し、40℃にて60分間静置した。洗浄後、Novocyte Quanteonフローサイトメーターを用いて、Treg細胞(Zombie NIR陰性かつCD45・CD3・CD4・Foxp3陽性画分)における3-3F抗体シグナルとCCR8 mRNAシグナルの相関を解析した。
 その結果、3-3F抗体シグナルとCCR8 mRNAシグナルが正に相関する結果が得られた(図8)。
Example 6: Confirmation of CCR8-specific binding of 3-3F antibody to intratumoral infiltrating Treg cells Simultaneous detection of signal by 3-3F antibody and CCR8 mRNA expression on flow cytometry using PrimeFlow RNA assay technology The CCR8 specificity of the 3-3F antibody was confirmed.
Tumor tissues derived from ovarian cancer patients were dispersed using Tumor Dissociation Kit, human (Miltenyi Biotec) and cryopreserved. Frozen tumor dispersed cells were thawed and suspended in Stain Buffer (BD Bioscience) supplemented with Human TruStain FcX (BioLegend) and Zombie NIR Fixable Viability Kit (BioLegend). , and left at 4° C. for 30 minutes. After washing, 3-3F antibody (4 μg/mL) was added and left at 4° C. for 30 minutes. After washing, fluorescently labeled antibodies against human CD45, CD3, and CD8 and AlexaFlour488-labeled anti-rat IgG antibody were added and left at 4°C for 30 minutes. After washing, fixation and membrane permeabilization with the reagents of PrimeFlow RNA Assay Kit (Thermo Fisher), PE-labeled anti-human Foxp3 antibody was added, and the mixture was allowed to stand at 4°C for 30 minutes. After washing, a probe for CCR8 mRNA was added and left at 40°C for 120 minutes. After washing, signal amplification probes Preamplifier and Amplifier were each hybridized at 40°C for 90 minutes, Alexa Flour647 labeled probe was added, and the mixture was left standing at 40°C for 60 minutes. After washing, the correlation between the 3-3F antibody signal and CCR8 mRNA signal in Treg cells (Zombie NIR negative and CD45/CD3/CD4/Foxp3 positive fraction) was analyzed using a Novocyte Quanteon flow cytometer.
As a result, a positive correlation was obtained between the 3-3F antibody signal and the CCR8 mRNA signal (FIG. 8).
実施例7:マウス腫瘍内浸潤細胞に対する3-3F抗体の結合評価
 マウス腫瘍内浸潤細胞に対する3-3F抗体の結合について、マウス腎臓癌の肺転移モデルを用いて評価した。本実施例では、3-3F抗体のBB515蛍光標識体を用いた。CCR8に結合した3-3Fとは異なる抗CCR8抗体が3-3F抗体の結合に及ぼす影響を評価するために、本マウスモデルの一部には、実施例4で用いたヒトCCR8に対する中和抗体であるOP-1抗体を投与しており、その際のヒトCCR8に結合したOP-1抗体を検出する抗体として、抗ヒトIgG Fc抗体であるM1310G05抗体のBV421蛍光標識体を使用した。比較対照として市販抗ヒトCCR8抗体である433H抗体のBV421蛍光標識体を使用し、同一の試料から分けた試料について染色、測定を行った。
 WO2020/138489号公報の実施例10に記載されたBalb/CマウスのCCR8をヒト型のCCR8遺伝子で置換したノックインマウス(雌、21週齢)に培養したマウス腎臓癌細胞、Rencaを4×10個、尾静脈に注入した。その12日後にOP-1抗体を尾静脈から注入した。翌日、0.05mg/kg投与群(8匹)、0.5mg/kg投与群(6匹)、および無処置マウス(9匹)より吸入麻酔下で腫瘍細胞が転移した肺を採取した。Tumor Dissociation Kit,mouse(Miltenyi Biotec社)、およびBD Pharma Lyse(BD Biosciences社)を用い、肺組織より単細胞を分散、精製し、フローサイトメトリー用の染色を行った。染色は、まずeBioscience Fixable Viability Dye eFluor 780(eFluor 780、Thermo Fisher Scientific社)で死細胞を染色し、その後、Mouse BD Fc Block(BD Biosciences社)で処理し非特異的な抗体結合を阻害した。その後、3-3F抗体、M1310G05抗体を含む各種の表面抗原(TCRβ、CD4、CD8、CD25)に対する抗体を加えて4℃で30分間反応させた(サンプル1)。同時にすべての試料から別のサンプルをとり433H抗体を含む各種表面抗原(CD45、TCRβ、CD4、CD8、CD25)に対する抗体で同様に反応させた(サンプル2)。洗浄後、Transcription Factor Buffer Set(BD Biosciences社)を用い細胞を固定、及び膜透過処理した後、ふたたびMouse BD Fc Blockで処理した後にPE標識抗ヒトFoxp3抗体を添加し、4℃に50分間(サンプル1)、もしくは一晩(サンプル2)静置し染色を行った。洗浄後、MACSQuantAnalyzer(Miltenyi Biotec社)を用いて、Treg細胞(サンプル1ではeFluor 780、CD8陰性、かつTCRβ、CD4、CD25、Foxp3陽性分画、サンプル2ではeFluor 780、CD8陰性、かつCD45、TCRβ、CD4、CD25、Foxp3陽性分画)における3-3F抗体のシグナルとM1310G05抗体シグナルの相関を解析した(図9)。各抗体の示す蛍光シグナルの指標として、Treg細胞中に占める陽性細胞の率とその陽性細胞集団の示すMFI(Mean Fluorescence Intensity)を掛け合わせた値(%xMFI)を用いた。また、比較対照として433H抗体のシグナルとM1310G05抗体シグナルの相関を解析した(図10)。
 その結果、OP-1抗体が結合したTreg細胞に対する3-3F抗体の結合量は正に相関することが判明した(図9)。一方、市販の抗CCR8抗体である433H抗体の結合量は、OP-1抗体の結合量が増大するにつれ減少する事が確認された(図10)。
Example 7: Evaluation of binding of 3-3F antibody to mouse tumor-infiltrating cells The binding of 3-3F antibody to mouse tumor-infiltrating cells was evaluated using a mouse kidney cancer lung metastasis model. In this example, a BB515 fluorescently labeled 3-3F antibody was used. In order to evaluate the effect of an anti-CCR8 antibody different from 3-3F binding to CCR8 on the binding of the 3-3F antibody, a portion of this mouse model was prepared using the neutralizing antibody against human CCR8 used in Example 4. OP-1 antibody, which is an anti-human IgG Fc antibody, was administered, and the BV421 fluorescent label of M1310G05 antibody, which is an anti-human IgG Fc antibody, was used as an antibody to detect the OP-1 antibody bound to human CCR8. As a comparison, a BV421 fluorescent label of 433H antibody, which is a commercially available anti-human CCR8 antibody, was used, and samples separated from the same sample were stained and measured.
Renca, a mouse kidney cancer cell cultured in a knock-in mouse (female, 21 weeks old) in which CCR8 of Balb/C mouse described in Example 10 of WO2020/138489 was replaced with human CCR8 gene, was cultured at 4×10. Five were injected into the tail vein. Twelve days later, OP-1 antibody was injected through the tail vein. The next day, the lungs to which tumor cells had metastasized were collected from the 0.05 mg/kg administration group (8 mice), the 0.5 mg/kg administration group (6 mice), and the untreated mice (9 mice) under inhalation anesthesia. Single cells were dispersed and purified from lung tissue using a Tumor Dissociation Kit, mouse (Miltenyi Biotec), and BD Pharma Lyse (BD Biosciences), and stained for flow cytometry. For staining, dead cells were first stained with eBioscience Fixable Viability Dye eFluor 780 (eFluor 780, Thermo Fisher Scientific), and then Mouse BD Fc Block (BD Biosc to inhibit non-specific antibody binding. Thereafter, antibodies against various surface antigens (TCRβ, CD4, CD8, CD25), including 3-3F antibody and M1310G05 antibody, were added and reacted at 4°C for 30 minutes (Sample 1). At the same time, another sample was taken from all the samples and reacted in the same manner with antibodies against various surface antigens (CD45, TCRβ, CD4, CD8, CD25), including the 433H antibody (Sample 2). After washing, cells were fixed and permeabilized using Transcription Factor Buffer Set (BD Biosciences), treated again with Mouse BD Fc Block, PE-labeled anti-human Foxp3 antibody was added, and incubated at 4°C for 50 minutes ( Sample 1) or overnight (sample 2) was left standing and stained. After washing, Treg cells (eFluor 780, CD8 negative, and TCRβ, CD4, CD25, Foxp3 positive fraction for sample 1, eFluor 780, CD8 negative, and CD45, TCR fraction for sample 2) were analyzed using MACSQuantAnalyzer (Miltenyi Biotec). β , CD4, CD25, Foxp3 positive fraction) and the correlation between the 3-3F antibody signal and the M1310G05 antibody signal was analyzed (FIG. 9). As an index of the fluorescence signal exhibited by each antibody, the value obtained by multiplying the ratio of positive cells among Treg cells by the MFI (Mean Fluorescence Intensity) of the positive cell population (% x MFI) was used. Furthermore, as a comparison, the correlation between the 433H antibody signal and the M1310G05 antibody signal was analyzed (FIG. 10).
As a result, it was found that the binding amount of 3-3F antibody to Treg cells bound to OP-1 antibody was positively correlated (FIG. 9). On the other hand, it was confirmed that the binding amount of 433H antibody, which is a commercially available anti-CCR8 antibody, decreased as the binding amount of OP-1 antibody increased (FIG. 10).
 本発明のモノクローナル抗体又はその抗体断片は、生体試料においてCCR8を検出するために使用することができる。さらに、本発明のモノクローナル抗体又はその抗体断片を含む医薬組成物は、CCR8関連疾患の治療又は予防のための医薬として非常に有用である。 The monoclonal antibody or antibody fragment thereof of the present invention can be used to detect CCR8 in a biological sample. Furthermore, a pharmaceutical composition containing the monoclonal antibody of the present invention or an antibody fragment thereof is very useful as a medicament for treating or preventing CCR8-related diseases.

Claims (27)

  1. 配列番号2のアミノ酸配列からなるCDR1、
    配列番号3のアミノ酸配列からなるCDR2及び
    配列番号4のアミノ酸配列からなるCDR3を含む軽鎖可変領域並びに
    配列番号5のアミノ酸配列からなるCDR1、
    配列番号6のアミノ酸配列からなるCDR2及び
    配列番号7のアミノ酸配列からなるCDR3を含む重鎖可変領域
    を含む、CCR8に結合するモノクローナル抗体又はその抗体断片。
    CDR1 consisting of the amino acid sequence of SEQ ID NO: 2,
    A light chain variable region comprising CDR2 consisting of the amino acid sequence of SEQ ID NO: 3 and CDR3 consisting of the amino acid sequence of SEQ ID NO: 4, and CDR1 consisting of the amino acid sequence of SEQ ID NO: 5,
    A monoclonal antibody or antibody fragment thereof that binds to CCR8 and includes a heavy chain variable region including CDR2 consisting of the amino acid sequence of SEQ ID NO: 6 and CDR3 consisting of the amino acid sequence of SEQ ID NO: 7.
  2. 配列番号8のアミノ酸配列を有する軽鎖可変領域、及び、配列番号9のアミノ酸配列を有する重鎖可変領域を含む、請求項1記載の抗体又はその抗体断片。 The antibody or antibody fragment thereof according to claim 1, comprising a light chain variable region having the amino acid sequence of SEQ ID NO: 8 and a heavy chain variable region having the amino acid sequence of SEQ ID NO: 9.
  3. さらに、配列番号10のアミノ酸配列を有する軽鎖定常領域、及び、配列番号11のアミノ酸配列を有する重鎖定常領域を有する、請求項1記載の抗体又はその抗体断片。 The antibody or antibody fragment thereof according to claim 1, further comprising a light chain constant region having the amino acid sequence of SEQ ID NO: 10 and a heavy chain constant region having the amino acid sequence of SEQ ID NO: 11.
  4. 請求項1~3のいずれかに記載の抗体又はその抗体断片を含有する組成物を使用する、CCR8の検出方法。 A method for detecting CCR8 using a composition containing the antibody or antibody fragment thereof according to any one of claims 1 to 3.
  5. 癌診断のための、請求項4記載の方法。 5. The method according to claim 4, for cancer diagnosis.
  6. 抗CCR8抗体を含有する医薬を投与した被験者から採取した試料に含まれるCCR8を検出する、請求項4記載の方法。 5. The method according to claim 4, wherein CCR8 contained in a sample collected from a subject to whom a medicine containing an anti-CCR8 antibody has been administered is detected.
  7. 抗CCR8抗体を含有する医薬に含まれる抗CCR8抗体が中和活性を有する、請求項6記載の方法。 7. The method according to claim 6, wherein the anti-CCR8 antibody contained in the anti-CCR8 antibody-containing medicament has neutralizing activity.
  8. 抗CCR8抗体を含有する医薬に含まれる抗CCR8抗体が、配列番号8のアミノ酸配列を有する軽鎖可変領域、及び、配列番号9のアミノ酸配列を有する重鎖可変領域を含む抗体とCCR8に対する結合が競合しない、請求項6記載の方法。 The anti-CCR8 antibody contained in the anti-CCR8 antibody-containing medicine has the ability to bind to CCR8 with an antibody comprising a light chain variable region having the amino acid sequence of SEQ ID NO: 8 and a heavy chain variable region having the amino acid sequence of SEQ ID NO: 9. 7. The method of claim 6, which is non-conflicting.
  9. 抗CCR8抗体を含有する医薬に含まれる抗CCR8抗体が、配列番号12のアミノ酸配列を有する軽鎖可変領域、及び、配列番号13のアミノ酸配列を有する重鎖可変領域を含むモノクローナル抗体又はその断片である、請求項6記載の方法。 The anti-CCR8 antibody contained in the anti-CCR8 antibody-containing medicine is a monoclonal antibody or a fragment thereof comprising a light chain variable region having the amino acid sequence of SEQ ID NO: 12 and a heavy chain variable region having the amino acid sequence of SEQ ID NO: 13. 7. The method of claim 6, wherein:
  10. 被験者が癌の患者である、請求項6記載の方法。 7. The method according to claim 6, wherein the subject is a cancer patient.
  11. 請求項1~3のいずれかに記載の抗体又はその抗体断片を含有する組成物を含む、CCR8の検出キット。 A CCR8 detection kit comprising a composition containing the antibody or antibody fragment thereof according to any one of claims 1 to 3.
  12. 癌診断のための、請求項11記載のキット。 The kit according to claim 11, for cancer diagnosis.
  13. 抗CCR8抗体を含有する医薬を投与した被験者から採取した試料に含まれるCCR8を検出する、請求項11記載のキット。 The kit according to claim 11, which detects CCR8 contained in a sample collected from a subject who has been administered a medicine containing an anti-CCR8 antibody.
  14. 抗CCR8抗体を含有する医薬に含まれる抗CCR8抗体が中和活性を有する、請求項13記載のキット。 The kit according to claim 13, wherein the anti-CCR8 antibody contained in the anti-CCR8 antibody-containing medicament has neutralizing activity.
  15. 抗CCR8抗体を含有する医薬に含まれる抗CCR8抗体が、配列番号8のアミノ酸配列を有する軽鎖可変領域、及び、配列番号9のアミノ酸配列を有する重鎖可変領域を含む抗体とCCR8に対する結合が競合しない、請求項13記載のキット。 The anti-CCR8 antibody contained in the anti-CCR8 antibody-containing medicine has the ability to bind to CCR8 with an antibody comprising a light chain variable region having the amino acid sequence of SEQ ID NO: 8 and a heavy chain variable region having the amino acid sequence of SEQ ID NO: 9. 14. The kit of claim 13, which is non-competitive.
  16. 抗CCR8抗体を含有する医薬に含まれる抗CCR8抗体が、配列番号12のアミノ酸配列を有する軽鎖可変領域、及び、配列番号13のアミノ酸配列を有する重鎖可変領域を含むモノクローナル抗体又はその断片である、請求項13記載のキット。 The anti-CCR8 antibody contained in the anti-CCR8 antibody-containing medicine is a monoclonal antibody or a fragment thereof comprising a light chain variable region having the amino acid sequence of SEQ ID NO: 12 and a heavy chain variable region having the amino acid sequence of SEQ ID NO: 13. 14. The kit of claim 13.
  17. 被験者が癌の患者である、請求項13記載のキット。 The kit according to claim 13, wherein the subject is a cancer patient.
  18. 請求項1~3のいずれかに記載の抗体又はその抗体断片を含有する、癌の診断薬。 A diagnostic agent for cancer, comprising the antibody or antibody fragment thereof according to any one of claims 1 to 3.
  19. 請求項1~3のいずれかに記載の抗体又はその抗体断片を含有する、抗CCR8抗体を含有する医薬の投与の適否を判断するための診断薬。 A diagnostic agent containing the antibody or antibody fragment thereof according to any one of claims 1 to 3, for determining the suitability of administration of a medicament containing an anti-CCR8 antibody.
  20. 抗CCR8抗体を含有する医薬が抗癌剤である、請求項19記載の診断薬。 The diagnostic agent according to claim 19, wherein the medicament containing the anti-CCR8 antibody is an anticancer agent.
  21. 抗CCR8抗体を含有する医薬に含まれる抗CCR8抗体が、配列番号12のアミノ酸配列を有する軽鎖可変領域、及び、配列番号13のアミノ酸配列を有する重鎖可変領域を含むモノクローナル抗体又はその断片である、請求項19記載の診断薬。 The anti-CCR8 antibody contained in the anti-CCR8 antibody-containing medicine is a monoclonal antibody or a fragment thereof comprising a light chain variable region having the amino acid sequence of SEQ ID NO: 12 and a heavy chain variable region having the amino acid sequence of SEQ ID NO: 13. The diagnostic agent according to claim 19.
  22. 請求項1~3のいずれかに記載の抗体又はその抗体断片を含有する組成物を使用する、抗CCR8抗体を含有する医薬の投与の適否を判断する方法。 A method for determining the suitability of administration of a medicament containing an anti-CCR8 antibody, using a composition containing the antibody or antibody fragment thereof according to any one of claims 1 to 3.
  23. 抗CCR8抗体を含有する医薬が抗癌剤である、請求項22記載の方法。 23. The method according to claim 22, wherein the medicament containing the anti-CCR8 antibody is an anticancer drug.
  24. 抗CCR8抗体を含有する医薬に含まれる抗CCR8抗体が、配列番号12のアミノ酸配列を有する軽鎖可変領域、及び、配列番号13のアミノ酸配列を有する重鎖可変領域を含むモノクローナル抗体又はその断片である、請求項22記載の方法。 The anti-CCR8 antibody contained in the anti-CCR8 antibody-containing medicine is a monoclonal antibody or a fragment thereof comprising a light chain variable region having the amino acid sequence of SEQ ID NO: 12 and a heavy chain variable region having the amino acid sequence of SEQ ID NO: 13. 23. The method of claim 22, wherein:
  25. 請求項1~3のいずれかに記載の抗体又はその抗体断片を含有する組成物を使用する、抗CCR8抗体を含有する医薬に含まれる抗CCR8抗体の腫瘍内浸潤Treg細胞への結合量を測定する方法。 Using a composition containing the antibody or antibody fragment thereof according to any one of claims 1 to 3, measuring the amount of binding of an anti-CCR8 antibody contained in a medicament containing an anti-CCR8 antibody to intratumoral infiltrating Treg cells. how to.
  26. 請求項1又は2記載の抗体の軽鎖可変領域又は重鎖可変領域をコードするポリヌクレオチド。 A polynucleotide encoding the light chain variable region or heavy chain variable region of the antibody according to claim 1 or 2.
  27. 請求項26記載のポリヌクレオチドを含む発現ベクター。 An expression vector comprising the polynucleotide according to claim 26.
PCT/JP2023/017821 2022-05-13 2023-05-12 Novel anti-ccr8 antibodies for detecting ccr8 WO2023219147A1 (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
JP2022-079190 2022-05-13
JP2022079190 2022-05-13

Publications (1)

Publication Number Publication Date
WO2023219147A1 true WO2023219147A1 (en) 2023-11-16

Family

ID=88730362

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/JP2023/017821 WO2023219147A1 (en) 2022-05-13 2023-05-12 Novel anti-ccr8 antibodies for detecting ccr8

Country Status (1)

Country Link
WO (1) WO2023219147A1 (en)

Citations (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
JP2001523446A (en) * 1997-11-13 2001-11-27 シェーリング コーポレイション Th2 cell depletion, compositions, methods
WO2018181064A1 (en) * 2017-03-27 2018-10-04 国立大学法人北海道大学 Anti-pd-l1 antibody for detecting pd-l1
JP2020511932A (en) * 2016-06-20 2020-04-23 カイマブ・リミテッド Anti-PD-L1 and IL-2 cytokine
JP2020511937A (en) * 2016-12-07 2020-04-23 ジェネンテック, インコーポレイテッド Anti-TAU antibody and method of use
WO2020138489A1 (en) * 2018-12-27 2020-07-02 塩野義製薬株式会社 Novel anti-ccr8 antibody

Patent Citations (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
JP2001523446A (en) * 1997-11-13 2001-11-27 シェーリング コーポレイション Th2 cell depletion, compositions, methods
JP2020511932A (en) * 2016-06-20 2020-04-23 カイマブ・リミテッド Anti-PD-L1 and IL-2 cytokine
JP2020511937A (en) * 2016-12-07 2020-04-23 ジェネンテック, インコーポレイテッド Anti-TAU antibody and method of use
WO2018181064A1 (en) * 2017-03-27 2018-10-04 国立大学法人北海道大学 Anti-pd-l1 antibody for detecting pd-l1
WO2020138489A1 (en) * 2018-12-27 2020-07-02 塩野義製薬株式会社 Novel anti-ccr8 antibody

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
SUZUKI HIROYUKI, SAITO MASAKI, ASANO TEIZO, TANAKA TOMOHIRO, KITAMURA KAISHI, KUDO YUMA, KANEKO MIKA K., KATO YUKINARI: "C 8 Mab-3: An Anti-Mouse CCR8 Monoclonal Antibody for Immunocytochemistry", MONOCLONAL ANTIBODIES IN IMMUNODIAGNOSIS AND IMMUNOTHERAPY, MARY ANN LIEBERT, INC. PUBLISHERS, US, vol. 41, no. 2, 1 April 2022 (2022-04-01), US , pages 110 - 114, XP093109349, ISSN: 2167-9436, DOI: 10.1089/mab.2022.0002 *

Similar Documents

Publication Publication Date Title
JP6894086B2 (en) New anti-CCR8 antibody
KR102536145B1 (en) Anti-pd-1 antibodies and uses thereof
TWI825086B (en) Her3 antigen-binding molecules
JP2022548057A (en) Cancer treatment and prevention using HER3 antigen binding molecules
AU2014273966B2 (en) Oncostatin M receptor antigen binding proteins
WO2020151761A1 (en) Bispecific antibody binding to pd-l1 and ox40
WO2020151762A1 (en) Novel bispecific antibody molecule and bispecific antibody simultaneously combining pd-l1 and lag-3
WO2016171242A1 (en) Detection of epha2
CN109988240B (en) anti-GPC-3 antibodies and uses thereof
EP2285403A2 (en) Human monoclonal antibodies against human chemokine receptor ccr7
AU2020351274A1 (en) Single-domain antibodies directed against LILRB2
KR20220024211A (en) Anti-CD47 Antibodies and Their Uses
CN113880951A (en) Coagulation Factor XI (FXI) binding proteins
WO2023219147A1 (en) Novel anti-ccr8 antibodies for detecting ccr8
JP2024513644A (en) VEGFA binding molecule
JP7082112B2 (en) Anti-GPR20 antibody
WO2020156507A1 (en) Novel anti-pd-l1 antibodies and use thereof
JPWO2016171107A1 (en) Detection of FGFR2
CN113880950A (en) Coagulation Factor XI (FXI) binding proteins
CN113880949A (en) Coagulation Factor XI (FXI) binding proteins
WO2023145844A1 (en) Anti-human cxcl1 antibody
WO2023190465A1 (en) Human anti-sema7a antibody
CN117304323A (en) Binding moieties that bind Claudin18.2 and uses thereof

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: 23803621

Country of ref document: EP

Kind code of ref document: A1