Substance specific to pd-1

Substances comprising a substance that recognizes PD-1, a substance that recognizes a membrane protein co-existing with PD-1 on a cell membrane, and a linker.

Skip to: Description  ·  Claims  · Patent History  ·  Patent History
Description
TECHNICAL FIELD

[0001] The invention is related to a substance comprising a substance that recognizes PD-1, a substance that recognizes a membrane protein co-existing with PD-1 on a cell membrane, and a linker.

BACKGROUND

[0002] The immune system acquired the mechanism that can respond to various foreign antigens. The mechanism brings about the diversity of an antigen receptor by recombination of V, (D) and J fragment in T cells and B cells. Although this mechanism brought a result that produces self-reactive lymphocytes simultaneously, these self-reactive lymphocytes are removed by the negative selection in the thymus or bone marrow, and are further controlled by the self-tolerance mechanism of clone removal or anergy in the periphery.

[0003] Although it is thought that an autoimmune disease is developed by the breakdown of self-tolerance, the researches using various disease model mice have been made towards the elucidation of the mechanism of pathogenesis of the disease. However, there are many still unknown matters about the etiology of an autoimmune disease and the molecular mechanism of self-tolerance. In such a situation, existence of the mouse which shows the symptoms of an autoimmune disease with a single gene deficit is very important, when trying to study the etiology of an autoimmune disease from a molecular biological viewpoint. CTLA4-/-mouse which causes lethal systemic lymphocytes infiltration (Waterhouse P. et. al., Science 270:985˜988, 1995, TivolE.A.et.al., Immunity3: 541˜547, 1995) SHP-1 deficit mothaten mice (Shulltz L. D. et. al., Cell 73: 1445˜1454, 1993) , TGF-beta-1 knockout mouse (Shull M. M. et. al., Nature 359:693-699, 1992), lyn-/-mouse which shows the symptoms of glomerular nephritis (Hibbs M. L. et. al., Cell 83:301-311, 1995), and FCRIIB-/-mouse (Bolland S. & Ravetch J. V., Immunity 13:277-285, 2000) are the representation, and the relation of these molecules and self-tolerance is studied.

[0004] PD-1 is type I membrane protein of 55 kD belonging to an immunoglobulin family. Both mouse PD-1 and human PD-1 consist of 288 amino acids, and have signal peptide at N terminal (20 amino acid) and hydrophobic region in the middle part, which is a transmembrane region (The EMBO J. 11 (11):3887-3895, 1992); Japanese patent Publication No. 5-336973; EMBL/GenBank/DDJB Acc. No. X67914, Genomics 23:704, 1994; Japanese patent Publication No. 7-291996 (U.S. Pat. No. 5,629,204).

[0005] In a thymocyte, PD-1 is expressed during a CD4−CD8− cell differentiation to a CD4+CD8+ cell (Nishimura H. et. al., Int.Immunol. 8:773-780, 1996, Nishimura H. et. al., J. Exp. Med. 191:891-898, 2000). Moreover, in the periphery, PD-1 is expressed in T cells and B cells which were activated by the stimulus from an antigen receptor (Agata Y. et. al., Int. Immunol. 8:765-772, 1996), and in bone marrow cells including activated macrophage.

[0006] PD-1 has ITIM (Immunoreceptor tyrosine-based inhibitory motif) in its intracellular region, therefore it is considered to be a negative regulator in immune reaction. PD-1 deficit mice develop a lupus-like autoimmune disease such as glomerular nephritis and arthritis (C57BL/6 genetic background) (Nishimura H. et. al., Int. Imuunol. 10:1563-1572, 1998, Nishimura H. et. al., Immunity 11:141-151, 1999) and dilated cardiomyopathy-like disease (BALB/c genetic background) (Nishimura H. et. al., Science 291:319-322, 2001), it became clear that PD-1 is a regulator of the development of autoimmune disease, especially one of self-tolerance in the periphery.

DISCLOSURE OF THE INVENTION

[0007] It is thought that PD-1 is the regulator of various autoimmune diseases, and that it is one of the genes that cause an autoimmune disease. By controlling the function of PD-1, it thought that the medical treatment and diagnosis of suppression or enhancement of the immune function, infection, the rejection at the time of a transplant, a neoplasm, etc. could be performed, and as a result of repeating examination wholeheartedly, the inventors reached this invention concerning the substance which controls the function of PD-1.

[0008] The stimulus to lymphocytes which control immunity is transmitted mainly through T cell receptor (TCR) in the case of T cells, and B cell receptor (BCR) in the case of B cells, and the intracellular phosphorylation play an important role in its molecular mechanism.

[0009] Since it became clear that PD-1 is controlling negatively various cells responsible for immunity, such as lymphocytes and myeloid cells etc., and PD-1 has ITIM (Immunoreceptor tyrosine-based inhibitory motif) in its intracellular region, the inventors considered the molecular mechanism involved in the inhibitory signal transduction of PD-1 to be the recruit of de-phosphorylation enzymes (phosphatases). Therefore, it came to be considered by locating PD-1 near TCR or BCR that it can display the function of PD-1. The inventors confirmed that the inhibitory signal of PD-1 was transmitted with the substance in which PD-1 was cross-linked to TCR or BCR, and completed this invention.

[0010] The inventors confirmed first that the above-mentioned idea was right using anti-PD-1 antibody, and anti-BCR antibody or anti-CD3 antibody. CD3 is the membrane protein expressed on a T cell, and is one component of the complexes that constitute TCR. The divalent antibody was constructed by bridging anti-PD-1 antibody and anti-BCR or anti-CD3 antibody. In the present invention, this divalent antibody is called hybrid antibody. The inventors produced this hybrid antibody for the first time.

[0011] Moreover, the knowledge that a signal transmits by cross-linking two kind of receptors using this hybrid antibody is also acquired for the first time.

[0012] Namely, the present invention relates to,

[0013] 1. A substance comprising a substance that recognizes PD-1, a substance that recognizes a membrane protein co-existing with PD-1 on a cell membrane, and a linker,

[0014] 2. The substance according to (1), which is a divalent substance comprising a substance that recognizes PD-1, a substance that recognizes a membrane protein co-existing with PD-1 on a cell membrane, and a linker,

[0015] 3. The substance according to (1) or (2), in which a membrane protein is a protein existing on a T cell membrane or B cell membrane,

[0016] 4. The substance according to (1) or (2), which comprises a substance that recognizes PD-1, a substance that recognizes a protein constituting T cell receptor complex or a substance that recognizes a protein constituting B cell receptor complex, and a linker,

[0017] 5. The substance according to any one of (1) to (4), in which a substance that recognizes PD-1 and in which a substance that recognizes a protein is dimer to pentamer, respectively,

[0018] 6. The substance according to any one of (1) to (5), in which one of or both substance that recognizes PD-1 and substance that recognizes a protein are an antibody,

[0019] 7. The substance according to any one of (1) to (6), in which one of the two or both substance that recognizes PD-1 and substance that recognizes a protein are a Fab portion of antibody,

[0020] 8. The substance according to any one of (1) to (5), in which a linker comprises an organic compound,

[0021] 9. The substance according to any one of (1) to (5), in which a linker comprises a peptide,

[0022] 10. The substance according to any one of (1) to (5), in which a substance that recognizes PD-1 and in which a substance that recognizes a protein is a peptide, respectively,

[0023] 11. The substance according to any one of (1) to (5), in which a substance that recognizes PD-1 and in which a substance that recognizes a protein comprises two or more peptides including a heavy chain variable region and a light chain variable region of antibody, respectively,

[0024] 12. The substance according to any one of (1) to (5), in which a substance that recognizes PD-1, in which a substance that recognizes a protein, and in which a linker is a peptide, respectively,

[0025] 13. A pharmaceutical composition containing an effective dose of the substance according to (1) for the medical treatment and/or prevention of a disease in which PD-1 participates,

[0026] 14. The pharmaceutical composition according to (13), in which disease is selected from the group consisting of neurodegenerative disease, autoimmune disease, organ transplant rejection, neoplasm and infection,

[0027] 15. The pharmaceutical composition according to (14), in which neurodegenerative disease is selected from the group consisting of Parkinson's disease, parkinsonian syndrome, Huntington's disease, Machado-Joseph disease, amyotrophic lateral sclerosis and Creutzfeldt-Jakob disease,

[0028] 16. The pharmaceutical composition according to (14), in which autoimmune disease is selected from the group consisting of glomerular nephritis, arthritis, dilated cardiomyopathy-like disease, ulcerative colitis, Sjogren's syndrome, Crohn's disease, systemic lupus erythematosus, chronic rheumatoid arthritis, multiple sclerosis, Psoriasis, allergic contact dermatitis, polymyositis, scleroderma, periarteritis nodosa, rheumatic fever, vitiligo vulgaris, insulin-dependent diabetes mellitus, Behcet's Syndrome and chronic thyroiditis.

[0029] A substance that recognizes PD-1 said by the present invention maybe a substance which recognizes PD-1 specifically, for example, anti-PD-1 antibody, the fragment of anti-PD-1 antibody, PD-1 in itself, the fragment of PD-1, ligand of PD-1 (PD-L1 (Freeman G. J. et. al., J. Exp. Med. 192:1027-1034, 2000), PD-L2, PD-H3), the fragment thereof and a low molecule organic compound, etc.

[0030] In more concretely, it is anti-PD-1 antibody produced by the hybridoma strain “J43”, which was deposited on May 30, 2001 in International Patent Organism Depositary, National Institute of Advanced Industrial Science and Technology, AIST Tsukuba Central 6, 1-1, Higashi 1-Chome Tsukuba-shi, Ibaraki-ken 305-8566 Japan as Accession number FERM P-18356, and transferred to international deposition on Jul. 16, 2002 as international deposition Accession number FERM BP-8118.

[0031] Preferably, although it is the Fab portion of the antibody, it is not limited to this.

[0032] A substance that recognizes a membrane protein co-existing with PD-1 on a cell membrane said by the present invention may be a substance which recognizes a membrane protein specifically, for example, a substance which specifically recognizes a complex that constitute T cell receptor (TCR) (TCR complex) etc. expressed on T cells, or a substance which specifically recognizes a complex that constitute B cell receptor (BCR) (BCR complex) etc. expressed on B cells, such as the fragment of a protein constituting TCR complex, anti-TCR antibody, the fragment of anti-TCR antibody, the fragment of a protein constituting BCR complex, anti-BCR antibody and the fragment of anti-BCR antibody.

[0033] Preferably, although it is the Fab portion of an anti-TCR antibody or the Fab portion of an anti-BCR antibody, it is not limited to these.

[0034] Both an anti-TCR antibody and an anti-BCR antibody are available commercially. For example, anti-CD3 antibody (&agr;-CD3&egr; mAb, manufactured by Pharmingen) as an anti-TCR antibody, and anti-IgG (H+L) polyclonal antibody (manufactured by Zymed) as an anti-BCR antibody are available, respectively.

[0035] A linker said by the present invention may be a substance which can connect the above mentioned substance recognizing PD-1 and the substance recognizing a membrane protein co-existing with PD-1 on a cell membrane if a suitable distance can be maintained. More specifically, it may be a peptide, amide, etc.

[0036] A linker can be used what is marketed, for example, Phenylenedimaleimide (manufactured by Aldrich) is available.

[0037] A substance that specifically recognizes PD-1, which is the subject of the present invention, can be produced as follows, for example.

[0038] In the case that an antibody is chosen as a substance which recognizes PD-1 specifically, and that an antibody is also chosen as a substance which recognizes specifically a membrane protein co-existing with PD-1 on a cell membrane (hereinafter, simply referred to as “the membrane protein”), a substance is called hybrid antibody by this invention. The method of producing this hybrid antibody is explained.

[0039] (1) Animals are immunized with PD-1 or the membrane protein as an antigen,

[0040] (2) Spleen cells from the immunized animal and myeloma cells are fused,

[0041] (3) Cells producing monoclonal antibody against the antigen (PD-1 or the membrane protein) are screened among thus obtained hybridoma cells,

[0042] (4) Aimed hybridoma cells are cloned,

[0043] (5) Cloned hybridoma cells are grown,

[0044] (6) Antibodies produced are isolated and purified,

[0045] (7) A hybrid antibody can be produced by bridging thus obtained anti-PD-1 antibody and anti-membrane antibody with a linker.

[0046] Alternatively,

[0047] (8) To obtain F(ab′)2, the antibodies produced are treated with pepsin, isolated and purified,

[0048] (9) Each F(ab′)2 thus prepared is reduced, isolated and purified,

[0049] (10)A hybrid antibody can be produced by bridging each F(ab′)SH thus prepared with a linker.

[0050] In the case that both or one of the two of a substance which specifically recognizes PD-1 and a substance which specifically recognizes a membrane protein co-existing with PD-1 on a cell membrane (the membrane protein) are a low molecule organic compound,

[0051] (11) A low molecule compound which inhibits the binding of each antibody to PD-1 or the membrane protein which is a corresponding antigen, respectively, is found out by using antibody produced by the above-mentioned technique and suitable detection equipment,

[0052] (12) A substance can be produced by bridging the compound-to-compound, the compound to antibody, or the compound to Fab with a linker.

[0053] It is as follows when each step is explained more concretely.

[0054] In an immunization step (1), it is desirable to administer PD-1 or the membrane protein to an animal into the peritoneal cavity or foot pad. Moreover, the animal to be immunized will not be limited especially if it is the one from which the monoclonal antibody is obtained generally such as mouse, rat or the like. In the case of a mouse, the amount of an antigen is enough if 10-200 micrograms is administered per time.

[0055] The cell fusion of (2) is carried out by excising spleen from an immunized animal with which the antibody titer has fully risen among the animals immunized in the step (1), preparing spleen cell suspension according to a usual method, and adding polyethylene glycol (preferably PEG4000) to a mixture of the spleen cells thus obtained and myeloma cells at 37° C. Some kinds, such as P3X63Ag8, P3/NS1/1-Ag4-1, and SP-2/0-Ag-14, are known as the mouse myeloma cell, and all of them are easily available.

[0056] As the myeloma cell, HGPRT (hypoxanthine-guanine phosphoribosyl transferase)-defective cell line which cannot survive in HAT medium (a medium containing hypoxanthine, aminopterin and thymidine) is useful, and it is further preferred that it is a cell line in which the myeloma cells themselves do not secrete any antibody. Preferably, SP-2O—Ag-14 is used.

[0057] Next, the obtained cell fusion mixture is dispensed into a 96-well micro plate at a low density, and cultivated in the HAT medium. By culturing them for 1 to 2 weeks, un-fused myeloma cells, hybridomas of myeloma cells themselves, un-fused spleen cells and dybridomas of spleen cells themselves die because their surviving conditions are not satisfied, and only the hybridomas of spleen cell with myeloma cell are propagated.

[0058] In the screening of (3), whether or not a hybridoma is the one which produces an antibody against PD-1 or the membrane protein is judged by allowing each hybridoma culture supernatant to react with the immobilized antigen, and then determining amount of the antibody in the supernatant specifically adsorbed to the antigen using a labeled second antibody.

[0059] The step (4) is carried out by cloning the antibody-producing hybridoma in accordance with the soft agar culture method (Monoclonal Antibodies, 372 (1980)). In this case, it is also possible to use the limiting dilution method.

[0060] The step (5) is carried out by culturing the cloned hybridoma in usual medium and then separating and purifying fromthe culture supernatant, however, to obtain a larger amount of the antibody efficiently a method in which the hybridoma is administered into the abdominal cavity of mouse, allowed to propagate therein and then separated and purified from the ascites is used.

[0061] In the step (6), the purification can be carried out by usual methods such as salting out, ion exchange chromatography, gel filtration, hydrophobic chromatography and affinity chromatography, however, affinity chromatography using protein A-sepharose CL-4B (manufactured by Amersham Bioscience) is used more effectively.

[0062] Since a hybrid antibody of the present invention recognizes PD-1 specifically, it can be used for the purification and concentration of PD-1, for example, for affinity chromatography etc.

[0063] The step (7) can be carried out by bridging a linker such as sulfo-EMCS (N-(6-maleimidcaproxy) sulfo-succinimide sodium salt) to amide groups or SH (mercapto) groups of an antibody. First, one antibody is combined to sulfo-EMCS by amide coupling, un-reacted sulfo-EMCS is discarded by gel filtration, the maleimide groups of the sulfo-EMCS that is bound to the first antibody is reacted with SH (mercapto) groups of the other antibody that is reduced with 2-mercaptoethylamine etc., and then a substance bridged over two kinds of antibodies is size-fractionated using gel filtration.

[0064] The step (8) is carried out by digesting each antibody obtained in the step (6) with pepsin at 37° C. for 48 hours. The separation and purification of F(ab′) 2 digested with pepsin can be carried out by usual methods such as salting out, ion exchange chromatography, gel filtration, hydrophobic chromatography and affinity chromatography, however, gel filtration using Sephacryl S-200 (manufactured by Amersham Bioscience) is used more effectively.

[0065] The step (9) is carried out by reducing F(ab′)2 with 2-mercaptoethanol at 30° C. for 30 minutes. The separation and purification of the reduced FabSH can be carried out by usual methods such as salting out, ion exchange chromatography, gel filtration, hydrophobic chromatography and affinity chromatography, however, gel filtration using Sephacryl S-200 is used more effectively.

[0066] In the step (10), FabSH fraction of one antibody is combined with a linker. As a linker, a substance that is combinable with mercapto (SH) groups of FabSH may be used, for example, a reaction is performed by adding phenylene dimaleimide for 30 minutes at room temperature. Next, the reaction is followed by adding the other FabSH multiplied by 1.3 at room temperature for 4 hours. The separation and purification of bispecific substance can be carried out by usual methods such as salting out, ion exchange chromatography, gel filtration, hydrophobic chromatography and affinity chromatography, however, gel filtration using Sephacryl S-200 is used more effectively.

[0067] The step (11) can be carried out by using the antibody obtained at the step (6) without modification, or by using the antibody with appropriate labeling (for example, biotin-conjugated or FITC-conjugated etc.) in accordance with a usual method. In the case that ELISA method is used, an antigen is immobilized by a usual method, and then an antibody is added. Next, when enzyme-conjugated second antibody and biotin-conjugated antibody are used, enzyme-conjugated streptavidin is added, then the specific binding between antigen and antibody is measured in the presence of chromophore-producing substance by using absorptiometer. By using this assay system a low molecule that specifically recognizes PD-1 or the membrane protein can be obtained.

[0068] In the step (12), when one of the two is an antibody or Fab, the obtained low molecule can be combined with the antibody or Fab by introducing the suitable functional group in it. For example, when maleimide groups is introduced, it is possible to make it combine with mercapto groups of an antibody or Fab. Moreover, when both substances are low molecules, it is possible to synthesize a molecule containing both ones.

[0069] In the case that an antibody is chosen as a substance which recognizes PD-1 specifically, that an antibody is also chosen as a substance which recognizes specifically a membrane protein co-existing with PD-1 on a cell membrane, and that both antibodies are included in the same peptide, the substance is called bispecific antibody in the present invention. The method of producing this bispecific antibody is explained.

[0070] (1) An antibody gene is isolated from hybridoma cells producing antibodies against PD-1 and the membrane protein, respectively,

[0071] (2) A variable domain of an antibody gene against PD-1 and a variable domain of an antibody gene against the membrane protein are connected using a linker DNA, the connected DNA fragment is inserted to an expression vector, and cells are transfected with the expression vector and propagated,

[0072] (3) Bispecific antibody can be prepared by separating and purifying the protein thus produced.

[0073] It is as follows when each step is explained more concretely.

[0074] The step (1) consists of the processes, which isolates RNA from hybridoma cells, and which isolates cDNA encoding an antibody or its partial peptide.

[0075] The process which isolates total RNA or mRNA from hybridoma cells can be carried out in accordance with known methods (hereinafter, if unstated especially, a known method is the method described in Molecular Cloning (Sambrook J., Fritsch E. F. and Maniatis T., Cold Spring Harbor Laboratory Press, 1989) or Current Protocol in Molecular Biology (Ausubel F. M. et al., John Wiley & Sons and Inc.).

[0076] The cloning of a cDNA encoding an antibody gene or its partial peptide of the present invention can be performed by an amplification using Polymerase Chain Reaction (hereinafter simply referred to as “PCR method”) with synthesized DNA primers having partial nucleotide sequence encoding an antibody of the present invention, or by a selection using hybridization of cDNAs inserted into a suitable vector with a labeled DNA fragment or synthetic DNA encoding an antibody of the present invention partially or entirely. Hybridization can be carried out in accordance with known methods. An antibody gene may be amplified directly by using Reverse Transcriptase Polymerase Chain Reaction (hereinafter simply referred to as “RT-PCR method”) from total RNA or mRNA.

[0077] (2) Bispecific antibody of the present invention may be prepared by:

[0078] i) A method of peptide synthesis, or

[0079] ii) A method of production using recombinant DNA technology, preferably, by the method described in ii) in an industrial production.

[0080] Examples of expression system (host-vector system) for the production of peptide by using recombinant DNA technology are the expression systems using bacteria, yeast, insect cells and mammalian cells.

[0081] For example, in an E. Coli expression system, the initiation codon (ATG) is added to 5′-end of the nucleotide sequence, for example, shown in SEQ ID NO:28, then the expression vector is prepared by connecting the obtained DNA to the downstream of a suitable promoter (e.g., trp promoter, lac promoter, &lgr; PL promoter, and T7 promoter), and by inserting it into a vector (e.g., pBR322, pUC18and pUC19) which functions in an E. coli strain.

[0082] Then, an E. coli strain (e.g., E. coli strain DH1, E. coli strain JM 109 and E. coli strain HB101) which is transformed with the expression vector described above may be cultured in an appropriate medium to obtain the desired peptide. When a signal peptide of bacteria (e.g., signal peptide of pel B) is utilized, the desired peptide maybe released in periplasm. Furthermore, a fusion protein with other peptide may be produced easily.

[0083] In a mammalian cell expression system, an expression vector is prepared by inserting a DNA, for example, having the nucleotide sequence shown in SEQ ID NO: 28, into the downstream of a proper promoter (e.g., SV40 promoter, LTR promoter and metallothione in promoter) in a proper vector (e.g., retro virus vector, papilloma virus vector, vaccinia virus vector and SV40vector). Next, suitable mammalian cells (e.g., monkey COS-1 cells, COS-7 cells, Chinese hamster CHO cells, mouse L cells, 293 cells etc.) are transfected with the expression vector thus obtained, and then the transfected cells are cultured in an appropriate medium, the aimed peptide can be secreted into the culture medium.

[0084] A transformation of E. coli can be carried out in accordance with the method, for example, described in Proc. Natl. Acad. Sci. (USA) 69:2110, 1972 and Gene 17: 107, 1982.

[0085] A transfection of mammalian cells can be carried out in accordance with the method, for example, described in Saiboukougaku supl.8:263 (New experimental protocol for cell technology), Shujun-sha, 1995 and Virology 52:456, 1973.

[0086] It is known that a bispecific antibody can be prepared directly by using recombinant DNA technology. For example, Alt et al. (FEBS Letter 454:90, 1999) reported the generation of a bispecific antibody (it is referred to a single-chain diabody) directed against carcinoembryonic antigen and E. coli beta-galactosidase. In said fragment, a heavy chain variable domain (VH) of one antibody is connected to a light chain variable domain (VL) of the other antibody with a short linker, which prevent the pairing of these two continuous domains on the same chain. Therefore, the VH and VL domains of this fragment are obliged to pair with each complementary VL and VH domain on other chain, and thereby form two antigen binding sites.

[0087] It is preferred that a peptide linker contains 3 to 12 amino acid residues, but it is not limited particularly to its amino acid sequence (Hudson et al., J. Immunol. Met. 231:177, 1999).

[0088] (3) The peptide thus obtained can be purified by usual methods such as salting out, ion exchange chromatography, gel filtration, hydrophobic chromatography and affinity chromatography.

[0089] Since bispecific antibody of the present invention also recognizes PD-1 specifically, it can be used for the purification and concentration of PD-1, for example, for affinity chromatography etc.

Industrial Applicability

[0090] Application for Pharmaceuticals:

[0091] The greatest and important usage of a substance that specifically recognizes PD-1 by the present invention is for the medical treatment of the following disease.

[0092] A substance that specifically recognizes PD-1 by the present invention is useful for the medical treatment and/or prevention of diseases such as neurodegenerative disease (Parkinson's disease, parkinsonian syndrome, Huntington's disease, Machado-Joseph disease, amyotrophic lateral sclerosis and Creutzfeldt-Jakob disease etc.).

[0093] A substance that specifically recognizes PD-1 by the present invention is also useful for the medical treatment and/or prevention of diseases, in which PD-1 is involved and immune responses are enhanced, such as autoimmune diseases (glomerular nephritis, arthritis, dilated cardiomyopathy-like disease, ulcerative colitis, Sjogren's syndrome, Crohn's disease, systemic lupus erythematosus, chronic rheumatoid arthritis, multiple sclerosis, Psoriasis, allergic contact dermatitis, polymyositis, scleroderma, periarteritis nodosa, rheumatic fever, vitiligo vulgaris, insulin-dependent diabetes mellitus, Behcet's Syndrome and chronic thyroiditis etc.), organ transplant rejection, and allergy.

[0094] A substance that specifically recognizes PD-1 by the present invention is also useful for the medical treatment and/or prevention of diseases, in which PD-1 is involved and immune responses are reduced, such as neoplasm and infections.

[0095] For the above mentioned usage, administration of the substance that specifically recognizes PD-1 by the present invention can be carried out in systemic or local, generally peroral or parenteral ways.

[0096] The dosage to be administered depends upon age, body weight, symptom, desired therapeutic effect, route of administration, and duration of the treatment etc. In human adults, one dose per person is generally between 0.1 mg and 100 mg by oral administration up to several times per day, or between 0.01 mg and 30 mg by parenteral administration (preferably intravenous administration) up to several times per day, or continuous administration between 1 and 24 hrs. per day into vein.

[0097] As mentioned above, the doses to be used depend upon various conditions. Therefore, there are cases in which doses lower than or greater than the ranges specified above may be used.

[0098] The compounds of the present invention may be administered as inner solid compositions or inner liquid compositions for oral administration, or as injections, liniments or suppositories etc. for parenteral administration.

[0099] Examples of Inner solid compositions for oral administration include compressed tablets, pills, capsules, dispersible powders and granules etc. Examples of capsules include hard capsules and soft capsules.

[0100] In such inner solid compositions, one or more of the active compound(s) remains intact, or is/are admixed with excipients (lactose, mannitol, glucose, microcrystalline cellulose and starch etc.), connecting agents (hydroxypropyl cellulose, polyvinylpyrrolidone, magnesium metasilicate aluminate, etc.), disintegrating agents (cellulose calcium glycolate etc.), lubricating agents (magnesium stearate etc.), stabilizing agents, assisting agents for dissolving (glutamic acid, asparaginic acid etc.) etc. to prepare pharmaceuticals by known methods. The pharmaceuticals may, if desired, be coated with coating agent (sugar, gelatin, hydroxypropyl cellulose or hydroxypropylmethyl cellulose phthalate etc.), or be coated with two or more films. Further, coating may include capsules of absorbable materials such as gelatin.

[0101] Inner liquid compositions for oral administration may contain pharmaceutically acceptable water-agents, suspensions, emulsions, syrups and elixirs etc. In such liquid compositions, one or more of the active compound(s) is/are resolved, suspended or emulsified in inert diluent (s) commonly used in the art (purified water, ethanol or mixture thereof etc.). Such liquid compositions may also comprise wetting agents, suspending agents, emulsifying agent, sweetening agents, flavoring agents, perfuming agents, preserving agents and buffer agents etc.

[0102] Injections for parenteral administration include solutions, suspensions and emulsions and solid injections, which are dissolved or suspended in solvent when it is used. In such compositions, one or more active compound(s) is/are dissolved, suspended or emulsified in a solvent. Solvents include distilled water for injection, physiological salt solution, plant oil, propylene glycol, polyethylene glycol, alcohol such as ethanol, and mixture thereof etc. Such compositions may comprise additional stabilizing agents, assisting agents for dissolving (glutamic acid, asparaginic acid, POLYSOLBATE80 (resistered trade mark) etc.), suspending agents, emulsifying agents, soothing agent, buffer agents, preserving agents etc. They may be manufactured or prepared by sterilization or by aseptic manipulation in a final process. They may also be manufactured in the form of sterile solid compositions such as freeze-dried compositions, and can be dissolved in sterile water or some other sterile solvent for injection immediately before use.

[0103] Other compositions for parenteral administration include liquids for external use, ointments, endermic liniments, aerosols, spray compositions, suppositories and pessaries for vaginal administration etc., which comprise one or more of the active compound(s) and may be prepared by known methods.

[0104] Spray compositions may comprise additional substances other than inert diluents generally used: e.g. stabilizing agents such as sodium hydrogen sulfate, buffer agents to give isotonicity, isotonic buffer such as sodium chloride, sodium citrate and citric acid. For preparation of such spray compositions, for example, the methods described in the U.S. Pat. No. 2,868,691 and 3,095,355 may be used.

[0105] Since PD-1 is involved in immune responses, the substance that specifically recognizes PD-1 by the present invention can also be used for the screening of substances, which are involved in immune responses, by measuring the expression of PD-1.

Effect of the Invention

[0106] A substance that specifically recognizes PD-1 by the present invention comprises a substance that recognizes PD-1, a substance that recognizes a membrane protein co-existing with PD-1 on a cell membrane, and a linker, is a superior substance recognizable both PD-1 and the membrane protein specifically and transmittable the signal of PD-1.

BRIEF DESCRIPTION OF THE DRAWINGS

[0107] FIG. 1 shows the results of FACS analysis using anti-PD-1/anti-TCR hybrid Fab antibodies;

[0108] FIG. 2 shows the effect of anti-PD-1/anti-TCR hybrid Fab antibodies on activated T cells;

[0109] FIG. 3 shows the inhibitory effect of anti-PD-1/anti-BCR hybrid Fab antibodies on the production of IL-2 by anti-BCR antibody stimulation;

[0110] FIG. 4 shows the effect of anti-PD-1/anti-BCR hybrid Fab antibodies on SHP-2 recruitment after anti-BCR antibody stimulation;

[0111] FIG. 5 shows the plasmid J43-2C11scDb-pSec/hygro B for the expression of J43-2C11 bispecific antibody; and

[0112] FIG. 6 shows the effect of J43-2C11 bispecific antibodies on the production of IFN-r by activated mouse spleen T cells.

BEST MODE FOR CARRYING OUT THE INVENTION

[0113] The present invention is more specifically explained by means of the following examples, but is not limited only to these examples.

[0114] Hereinafter, a substance in which anti-PD-1 antibody and anti-T cell receptor antibody are linked by a linker simply referred to as “anti-PD-1/anti-TCR hybrid antibody”, and a substance in which anti-PD-1 antibody and anti-B cell receptor antibody are linked by a linker “anti-PD-1/anti-BCR hybrid antibody”, respectively. A substance in which the FabSH portion of anti-PD-1 antibody and the FabSH portion of anti-T cell receptor antibody are linked by a linker hereinafter simply referred to as “anti-PD-1/anti-TCR hybrid Fab antibody”, and a substance in which the FabSH portion of anti-PD-1 antibody and the FabSH portion of anti-B cell receptor antibody are linked by a linker “anti-PD-1/anti-BCR hybrid Fab antibody”, respectively.

EXAMPLE 1 (1) Preparation of Anti-PD-1/Anti-TCR Hybrid Antibodies

[0115] (1-A) Introduction of Maleimide to Anti-Mouse CD3&egr; Monoclonal Antibodies

[0116] Anti-mouse CD3&egr; monoclonal antibodies were substituted with Sodium phosphate (0.1 M, pH7.0) and Nacl (50 mM), then 200 times quantity of sulfo-EMCS (manufactured by Dojin Chemical) were added and incubated at 20° C. for 1 hour. Then the reaction mixture was size-fractionated by gel filtration using Sephacril S-300 [Sodium phosphate (0.1 M, pH7.0)] and major peak fractions were collected by monitoring the absorbency at 280 nm. The protein content was calculated from the absorbency at 280 nm simultaneously.

[0117] (1-B) Reduction of Anti-PD-1 Antibodies

[0118] Antibodies against mouse PD-1 (produced by the hybridoma cells named (J43) and deposited as international deposition Accession number FERM BP-8118) were substituted with Sodium phosphate (0.1 M, pH6.0), added with 2-mercaptoethylamine (final concentration of 10 mM) and EDTA (final concentration of 1 mM), and reduced at 37° C. for 90 minutes. Then the reaction mixture was size-fractionated by gel filtration using Sephacryl S-300 [Sodium phosphate (0.1 M, pH6.0)] and major peak fractions (single chain fractions) were collected by monitoring the absorbency at 280 nm. The protein content was calculated from the absorbency at 280 nm simultaneously. (1-C) Bridging of Maleimide-conjugated anti-mouse CD3&egr; monoclonal antibodies and Reduced anti-PD-1 antibodies

[0119] Maleimide-conjugated anti-mouse CD3&egr; monoclonal antibodies and Reduced anti-PD-1 antibodies were mixed at the rate of 1:4 and incubated at 15° C. for 18 hours. Then the reaction mixture was size-fractionated by gel filtration using Sephacryl S-300 [Sodium phosphate (0.1 M, pH7.0)] and major peak fractions were collected by monitoring the absorbency at 280 nm. The protein content was calculated from the absorbency at 280 nm simultaneously.

(2) Preparation of Anti-PD-1/Anti-TCR Hybrid Fab Antibodies

[0120] (2-A) Preparation of F(ab′)2 Fraction of Anti-PD-1 Antibodies

[0121] Antibodies against mouse PD-1 (produced by the hybridoma cells named (J43) and deposited as international deposition Accession number FERM BP-8118) were substituted with pepsin-buffer [(sodium acetate 0.1 M, pH4.5), NaCl (0.1 M)], added with pepsin (final concentration of 0.2 mg/ml), and digested for 48 hours at 37° C. Then the reaction mixture was size-fractionated by gel filtration using Sephacryl S-200 [Tris-HCl (0.2 M, pH8.0), EDTA (10 mM)] and major peak fractions (F(ab′)2 fraction) were collected by monitoring the absorbency at 280 nm. The protein content was calculated from the absorbency at 280 nm simultaneously.

[0122] (2-B) Preparation of FabSH Fraction of Anti-PD-1 Antibodies

[0123] 2-mercaptoethanol (final concentration of 20 mM) was added to reduce the F(ab′)2 fraction at 30° C. for 30 minutes. After cooling the reaction mixture on ice, it was size-fractionated by gel filtration using Sephacryl S-200 [sodium acetate (50 mM, pH6.3), EDTA (1 mM)] and major peak fractions (FabSH fraction) were collected by monitoring the absorbency at 280 nm. The protein content was calculated from the absorbency at 280 nm simultaneously.

[0124] (2-C) Preparation of F(ab′)2 Fraction of Anti-Mouse CD3&egr; Monoclonal Antibodies

[0125] Anti-mouse CD3&egr; monoclonal antibodies (manufactured by Pharmingen) were substituted with pepsin-buffer [(sodium acetate 0.1 M, pH4.5), NaCl (0.1 M)], added with pepsin (final concentration of 0.2 mg/ml), and digested for 48 hours at 37° C. Then the reaction mixture was size-fractionated by gel filtration using Sephacryl S-200 [Tris-HCl (0.2 M, pH8.0), EDTA (10 mM)] and major peak fractions (F(ab′)2 fraction) were collected by monitoring the absorbency at 280 nm. The protein content was calculated from the absorbency at 280 nm simultaneously.

[0126] (2-D) Preparation of FabSH Fraction of Anti-Mouse CD3&egr; Monoclonal Antibodies

[0127] 2-mercaptoethanol (final concentration of 20 mM) was added to reduce the F(ab′)2 fraction at 30° C. for 30 minutes. After cooling the reaction mixture on ice, it was size-fractionated by gel filtration using Sephacryl S-200 [sodium acetate (50 mM, pH6.3), EDTA (1 mM)] and major peak fractions (FabSH fraction) were collected by monitoring the absorbency at 280 nm. The protein content was calculated from the absorbency at 280 nm simultaneously.

[0128] (2-E) Bridging of FabSH Fraction of Anti-PD-1 Antibodies and FabSH Fraction of Anti-Mouse CD3&egr; Monoclonal Antibodies

[0129] Phenylenedimaleimide (manufactured by Aldrich) (final concentration of 4 mM) was added to the FabSH fraction of anti-PD-1 antibodies prepared in step (2-A), and incubated for30minutes at room temperature to prepare J43 Fabmal fraction. The J43 Fabmal fraction and the FabSH fraction of anti-mouse CD3&egr; monoclonal antibodies were mixed at the rate of 1:1.3 and incubated for 4 hours at room temperature. Next, an appropriate amount of Tris-HCl (1 M, pH8.0) were added to make the pH of the reaction mixture 8.0, 2-mercaptoethanol (final concentration of 20 mM) was added and incubated at 30° C. for 30 minutes. Then iodoacetoamide (manufactured by Sigma) was added (final concentration of 25 mM) and incubated for additional 10 minutes at room temperature under light shielding.

[0130] Finally, the reaction mixture was size-fractionated by gel filtration using Sephacryl S-200 [sodium acetate (50 mM, pH6.3), EDTA (1 mM)] and major peak fractions (BsAb fraction) were collected by monitoring the absorbency at 280 nm. The protein content was calculated from the absorbency at 280 nm simultaneously.

EXAMPLE 2 Confirmation of Reactivity of Anti-PD-1/Anti-TCR Hybrid Fab Antibodies on Cell Surface Antigen (PD-1 and CD3)

[0131] 1×106 cells were recovered from mPD-1/A 20IIA1.6 cells as a PD-1 positive and CD3 negative cell, and naive T cells prepared from mouse spleen cells as a PD-1 negative and CD3 positive cell, respectively. The cells were added with 91 &mgr;l of FACS buffer (PBS(−) containing 0.5% BSA, EDTA (2 mM) and 0.01% NaN3), 5 &mgr;l of mouse serum and 4 &mgr;l of hybrid antibodies (2 &mgr;g) and incubated for 30 minutes on ice. After washing with PBS(−) once, the cells were added with each 2 &mgr;l (1 &mgr;g) of second antibodies, fill upped to final 100 &mgr;l with FACS buffer, and incubated for 30 minutes on ice. Then they were analyzed by using FACS can. The results were shown in FIG. 1 (in the following figures, hybrid Fab antibody simply referred to as “HFAb”).

[0132] Analysis whether the prepared anti-PD-1/anti-TCR hybrid Fab antibodies actually react with the cell surface antigens by using FACSsort (manufactured by Becton Dickinson) showed the results that the antibodies reacted with both surface antigens.

EXAMPLE 3 Effect of Anti-PD-1/Anti-TCR Hybrid Fab Antibodies on Activated T cells

[0133] (A) Preparation of Spleen T Cells

[0134] Spleen was excised from BALB/c mouse. After red blood cells were hemolyzed, the spleen cells were washed once with PBS(−) and suspended in medium RPMI1640 (10% FCS, antibiotics) (1×108 cells/ml). Next, T cells were prepared by using nylon fiber column (manufactured by WAKO) for T cell separation equilibrated with the medium.

[0135] (B) Effect of Anti-PD-1/Anti-TCR Hybrid Fab Antibodies on Activated Spleen T Cells

[0136] Ninety six-well plates were coated with 0.5 &mgr;g/ml and 5 &mgr;g/ml of anti-CD3 antibodies (clone KT3, manufactured by Immuntech) at 37° C. for 3 hours. T cells (2×105 cells/well/200 &mgr;l ) were seeded on the plates, anti-PD-1/anti-TCR hybrid antibodies (0.03, 0.1, 0.3, 1, 3 &mgr;g/100 ml) were added, and incubated in a CO2 incubator (at 37° C.). After 72 hours, cytokine (IFN-r, IL-2, IL-4 and IL-10) concentrations in the recovered culture supernatants were measured by using assay kit (manufactured by R & D System).

[0137] As the results shown in FIG. 2, after 72 hours when 3 &mgr;g of anti-PD-1/anti-TCR hybrid Fab antibodies were used, the results were obtained that the stimulation by the antibodies both in 0.5 &mgr;g/ml and 5 &mgr;g/ml suppressed the production of IFN-r, IL-4 and IL-10 dominantly.

EXAMPLE 4 Preparation of Anti-PD-1/Anti-BCR Hybrid Fab Antibodies

[0138] (A) Preparation of F(ab′)2 Fraction of Anti-PD-1 Antibodies

[0139] Antibodies against mouse PD-1 (the same antibodies as used in Example 1) were substituted with pepsin-buffer [(sodium acetate 0.1 M, pH4.5), NaCl (0.1 M)], added with pepsin (manufactured by SIGMA) (final concentration of 0.2 mg/ml), and digested at 37° C. for 48 hours. Then the reaction mixture was size-fractionated by gel filtration using Sephacryl S-200 (manufactured by AmarshamParmacia) [Tris-HCl (0.2 M, pH8.0), EDTA (10 mM)] and major peak fractions (F(ab′)2 fraction) were collected by monitoring the absorbency at 280 nm. The protein content was calculated from the absorbency at 280 nm simultaneously.

[0140] (B) Preparation of FabSH Fraction of Anti-PD-1 Antibodies

[0141] 2-mercaptoethanol (final concentration of 20 mM) was added to reduce the F(ab′)2 fraction at 30° C. for 30 minutes. After cooling the reaction mixture on ice, it was size-fractionated by gel filtration using Sephacryl S-200 [sodium acetate (50 mM, pH6.3), EDTA (1 mM)] and major peak fractions (FabSH fraction) were collected by monitoring the absorbency at 280 nm. The protein content was calculated from the absorbency at 280 nm simultaneously.

[0142] (C) Preparation of F(ab′)2 Fraction of Anti-IgG (H+L) Polyclonal Antibodies

[0143] Rabbit anti-mouse IgG (H+L) polyclonal antibodies (manufactured by Zymed) were substituted with pepsin-buffer [(sodium acetate 0.1 M, pH4.5), NaCl (0.1 M)], added with pepsin (final concentration of 0.2 mg/ml), and digested at 37° C. for 48 hours. Then the reaction mixture was size-fractionated by gel filtration using Sephacryl S-200 [Tris-HCl (0.2 M, pH8.0), EDTA (10 mM)] and major peak fractions (F(ab′)2 fraction) were collected by monitoring the absorbency at 280 nm. The protein content was calculated from the absorbency at 280 nm simultaneously.

[0144] (D) Preparation of FabSH Fraction of Anti-IgG (H+L) Polyclonal Antibodies

[0145] 2-mercaptoethanol (final concentration of 20 mM) was added to reduce the F(ab′)2 fraction at 30° C. for 30 minutes. After cooling the reaction mixture on ice, it was size-fractionated by gel filtration using Sephacryl S-200 [sodium acetate (50 mM, pH6.3), EDTA (1 mM)] and major peak fractions (FabSH fraction) were collected by monitoring the absorbency at 280 nm. The protein content was calculated from the absorbency at 280 nm simultaneously.

[0146] (E) Bridging of FabSH Fraction of Anti-PD-1 Antibodies and FabSH Fraction of Anti-IgG (H+L) Polyclonal Antibodies

[0147] Phenylenedimaleimide (manufactured by Aldrich) (final concentration of 4 mM) was added to the FabSH fraction of J43 anti-PD-1 antibodies, and incubated for 30 minutes at room temperature to prepare J43 Fabmalfraction. The J43 Fabmal fraction and the FabSH fraction of anti-IgG (H+L) polyclonal antibodies were mixed at the rate of 1:1.3 and incubated for 4 hours at room temperature. Next, an appropriate amount of Tris-HCl (1 M, pH8.0) were added to make the pH of the reaction mixture 8.0, 2-mercaptoethanol (final concentration of 20 mM) was added and incubated at 30° C. for 30 minutes. Then iodoacetoamide (manufactured by SIGMA) was added (final concentration of 25 mM) and incubated for additional 10 minutes at room temperature under light shielding. Finally, the reaction mixture was size-fractionated by gel filtration using Sephacryl S-200 [sodium acetate (50 mM, pH6.3), EDTA (1 mM)] and major peak fractions (BsAb fraction) were collected by monitoring the absorbency at 280 nm. The protein content was calculated from the absorbency at 280 nm simultaneously.

EXAMPLE 5 Effect of Anti-PD-1/Anti-BCR Hybrid Fab Antibodies on B Cell Line

[0148] (A) Generation of A20IIA1.6 (B cell line) that was Forced to Express Mouse PD-1

[0149] (1) Construction of expression plasmid for mouse PD-1

[0150] DNA fragments of mPD1-flag digested with EcoRI were inserted into the EcoRI site of a commercially available expression vector to construct the expression plasmid mPD1-pA.

[0151] (2) Transfection

[0152] A20IIA1.6 cells (1×107) in 325 &mgr;l of ice-cold RPMI1640 medium containing 15% FCS and the PD-1 expression plasmid linearized with ScaI in 10 &mgr;l of distilled water were incorporated into a Cuvette for electroporation (Gene Pulser Cuvette 0.4 cm electrode gap, 50, BIO RAD), and pulsed under the condition of 250V/960 &mgr;F (Gene Pulser, BIO RAD). After leaving at rest for 10 minutes at room temperature, the cells were suspended in 30 ml of medium (RPMI1640 containing 10% FBS, 50 &mgr;l of 2-mercaptoethanol, penicillin and streptomycin), diluted thirtyfold further, and dispensed onto 96-well plates (103/100 &mgr;l/well). After 48 hours, the selection was initiated using final 3 &mgr;M of Puromycin to establish the cell line expressing mouse PD-1.

[0153] (B) Effect of Anti-PD-1/Anti-BCR Hybrid Fab Antibodies on A20IIA1.6 Cells that was Forced to Express Mouse PD-1

[0154] A20IIA1.6 cells that was forced to express mouse PD-1 were seeded on 96-well plates (5×105 cells/100 &mgr;l). Anti-PD-1/anti-BCR hybrid Fab antibodies (0, 1, 3, 10 &mgr;g/100 &mgr;l) were added, 10 minutes later 100 &mgr;l of anti-mouse IgG (H+L) F(ab′)2 (manufactured by Zymed) (final concentration of 0.3, 1, 3 &mgr;g/ml) were dispensed and cultivated for 12 hours in a CO2 incubator (at 37° C.). The culture supernatants were recovered and the concentrations of IL-2 in the culture supernatants were measured by using mouse IL-2 assay kit (manufactured by R & G System). The results were shown in FIG. 3.

[0155] The studies using various doses of anti-PD-1/anti-BCR hybrid Fab antibodies and anti-BCR antibodies F(ab′)2 showed the suppressive effects of the hybrid Fab antibodies in all cases, regardless of the concentration of anti-BCR antibodies F(ab′)2.

[0156] (C) Confirmation of SHP-2 Recruitment

[0157] Anti-PD-1/anti-BCR hybrid Fab antibodies (0, 1, 3, 10 &mgr;g/100 &mgr;l) were added to 3×106 of A20IIA1.6 cells (B cell line) that was forced to express mouse PD-1. After 10 minutes, 100 &mgr;l of anti-mouse IgG (H+L) F(ab′)2 were added and incubated for 5 minutes at room temperature. After discarding the supernatants by centrifugation, the cells were suspended in 200 &mgr;l of lysis buffer (composition: Tris-HCl (20 mM, pH7.4), NaCl (150 mM), Na2EDTA (1 mM), EGTA (1 mM), 1% Triton-X100, sodium pyrophosphate (2.5 mM), &bgr;-sodium glycerophosphate (1 mM), Na3VO4 (1 mM), leupeptin (1 &mgr;g/ml) and PMSF (1 mM) and left at rest on ice. After 30 minutes, the supernatants were recovered by centrifugation, 20 &mgr;l of protein G-cepharose beads (manufactured by Amersham Bioscience) added, and incubated at 4° C. for 30 minutes. After recovering the supernatants by centrifugation, 20 &mgr;l of protein G-cepharose beads bound beforehand with anti-FLAG antibodies (manufactured by SIGMA) were added and mixed over night at 4° C.

[0158] The beads were washed five times with 400 &mgr;l of lysis buffer, added with 20 &mgr;l of lysis buffer and 20 &mgr;l of 2× SDS sample buffer, and boiled at 100° C. for 5 minutes. After discarding the beads by centrifugation, 15 &mgr;l of supernatants were subjected to 4-20% SDS-PAGE. After electrophoresis the gels were substituted with blotting buffer and transferred onto PVDF membrane (manufactured by BIO RAD). Then the membrane was blocked with Block Ace (manufactured by Dainippon Pharmaceuticals) for 1 hour at room temperature.

[0159] The membrane was incubated with anti-SHP-2 antibody (manufactured by SANTA CRUZ) diluted 1/200 for 1 hour at room temperature, then washed three times with TBS-T for 10 minutes. Next, the membrane was incubated with HRP-conjugated anti-rabbit Ig antibody (manufactured by Amersham Bioscience) diluted 1/2000 for 1 hour at room temperature, then washed three times with TBS-T for 10 minutes. Finally the membrane was emitted light by using ECT plus detection kit (manufactured by Amersham Bioscience) and analyzed using luminoimager LAS1000 plus (manufactured by FUJI Film).

[0160] Since anti-PD-1/anti-BCR hybrid Fab antibodies suppressed the production of IL-2 from anti-BCR antibody stimulated B cells, the evaluation whether the effects were caused by the recruitment of phosphatase SHP-2 to ITIM of PD-1was carried out. As shown in FIG. 4, by adjusting the amount of samples with that of PD-1 as a control, the determination of the quantity of SHP-2 recruitment resulted in the obvious recruitment of SHP-2, in 1 and 10 &mgr;g, as compared with control hybrid antibodies.

EXAMPLE 6 cDNA Cloning of Anti-Mouse PD-1 Antibody J43

[0161] (1) Preparation of Anti-Mouse PD-1 Antibody J43

[0162] Anti-mouse PD-1 antibody producing hybridoma (J43) cells were cultured in Hybridoma SFM medium (manufactured by Invitrogen) at 37° C. under 5% CO2, a few days after the culture supernatants of hybridoma cells were recovered. IgG fraction was purified from the culture supernatants recovered by using HiTrap Protein G (manufactured by Amersham Bioscience).

[0163] (2) Peptide Sequencing

[0164] J43 IgG was subjected to 10-20% SDS-PAGE. After electrophoresis the IgG was electrically transferred from the gel onto PVDF membrane (manufactured by BIO RAD). The membrane transferred was stained with coomassie, the membrane fraction containing the light chain of J43 IgG was removed, and amino terminal 15 residues of the light chain were determined by using peptide sequencer Procise492 (manufactured by Applied Biosystems)(SEQ ID NO:1).

[0165] (3) Extraction of RNA

[0166] 5×106 hybridoma cells were lysed with 1 ml of TRIzol (manufactured by Invitrogen). Total RNA was prepared according to the direction of attached document. mRNA was purified from total RNA thus prepared by using Oligotex-MAG mRNA Purification Kit (manufactured by Takara Shuzo).

[0167] (4) Cloning of Light Chain cDNA (3′RACE) Degenerated primer (primer No.1) was designed based on the amino terminal sequence (YELTQPPSASVNVGE) of the light chain determined by peptide sequencing. 3′RACE was carried out by using 3′-Full RACE Core Set (manufactured by Takara Shuzo) under the following conditions. 1 Primer No. 1 5′-ta(c/t) ga(a/g) ct(g/a/t/c) ac(g/a/t/c) ca(a/g) (SEQ ID NO:2) cc(g/a/t/c) cc-3′ 1) Synthesis of first strand cDNA 10 × RNA polymerase chain reaction (PCR) buffer 2 J43 mRNA (50 ng/&mgr;l) 2 Magnesium chloride (25 mM) 4 dNTP mixture (each 10 mM) 2 AMV Reverse Transcriptase XL (5 U/&mgr;l) 1 Oligo dT-3sites Adapter primer (2.5 pmol/&mgr;l) 1 Ribonuclease (RNase) inhibitor (40 U/&mgr;l) 0.5 dH2O 7.5 15 &mgr;l 30° C. 10 min. → 50° C. → 30 min. → 95° C. 5 min. → 4° C. 5 min. 2) Polymerase chain reaction (PCR) First strand cDNA 1 Primer No. 1 (20 pmol/&mgr;l) 1 Anchorprimer 1 dH2O 22 One shot LA PCR Mix™ 25 50 &mgr;l 95° C. 5 min. → (94° C. 29 sec., 50° C. 20 sec., 72° C. 60 sec.) × 30 cycles

[0168] The PCR products were subjected to electrophoresis using 1% Agarose gel, then the gel was stained by EtBr. DNA fragment was recovered from the gel by using MinElute Gel Extraction Kit (manufactured by Quiagen), the DNA fragment recovered was ligated to pGEM-T Easy Vector (manufactured by Promega) by using DNA Ligation Kit ver.2 (manufactured by Takara Shuzo). E. coli DH5&agr; was transformed with the ligated plasmid. Finally, the plasmid was purified from E. coli and J43 IgG light chain cDNA was sequenced (SEQ ID NO:3). The deduced amino acid sequence of the cDNA is shown in sequence listing (SEQ ID NO:4).

[0169] (5) Cloning of Heavy Chain cDNA (5′RACE)

[0170] To perform 5′RACE, primers for constant region were designed based upon the reported information of hamster IgG heavy chain cDNA sequence (GenBank Accession No.U17166). 5′RACE was carried out by using 5′-Full RACE Core Set (manufactured by Takara Shuzo) under the following conditions. 2 Primer No. 2 5′-ccc aag agg tca gga gtt gga-3′ (5′ phosphorylated) (SEQ ID NO:5) Primer No. 3 5′-ttg acc agg cat ccc agg gtc-3′ (SEQ ID NO:6) Primer No. 4 5′-cgt aag ctg gaa ctc tgg agc-3′ (SEQ ID NO:7) Primer No. 5 5′-tgg ttg tgc tgt cac agg cag-3′ (SEQ ID NO:8) Primer No. 6 5′-tgc aca cct tcc cat ctg tcc t-3′ (SEQ ID NO:9) 1) Synthesis of first strand cDNA J43 total RNA (2 &mgr;g/&mgr;l) 2 10 × RNA polymerase chain reaction (PCR) buffer 1.5 Ribonuclease (RNase) inhibitor (40 U/&mgr;l) 0.5 AMV Reverse Transcriptase XL (5 U/&mgr;l) 1 Primer No. 2 (100 pmol/&mgr;l) 2 dH2O 8 15 &mgr;l 30° C. 10 min. → 50° C. → 40 min. → 30° C. 2 min. 2) Degeneration of hybrid RNA First strand cDNA 15 5 × Hybrid RNA Degeneration buffer 15 dH2O 45 Ribonuclease H (RNaseH) 1 76 &mgr;l 30° 1 hour After the reaction, ethanol precipitation was carried out. 3) Cyclization of single chain cDNA by ligation 5 × RNA (ssDNA) Ligation buffer 8 40% Polyethylene glycol (PEG) #6000 20 dH2O 12 Pellet after ethanol precipitation T4 RNA ligase 1 41 &mgr;l 15° C. 15 hours 4) First PCR Sample after ligation 1 Primer No. 3 (5 pmol/&mgr;l) 2 Primer No. 4 (5 pmol/&mgr;l) 2 dH2O 20 One shot LA PCR Mix™ 25 50 &mgr;l 94° C. 3 min. → (94° C. → 30 sec., 52° C. 30 sec, 72° C. 120 sec.) × 25 cycles 5) Second PCR Product of first PCR 2 Primer No. 5 (5 pmol/&mgr;l) 2 Primer No. 6 (5 pmol/&mgr;l) 2 dH2O 20 One shot LA PCR Mix™ 25 50 &mgr;l 94° C. 3 min. → (94° C. 30 sec., 52° C. 30 sec, 72° C. 120 sec.) × 30 cycles

[0171] The PCR products were subjected to electrophoresis using 1% Agarose gel, then the gel was stained by EtBr. DNA fragment was recovered from the gel by using MinElute Gel Extraction Kit (manufactured by Quiagen), the DNA fragment recovered was ligated to pGEM-T Easy Vector (manufactured by Promega) by using DNA Ligation Kit ver.2 (manufactured by Takara Shuzo). E. coli DH5&agr; was transformed with the ligated plasmid. Finally, the plasmid was purified from E. coli and J43 IgG heavy chain cDNA was sequenced (SEQ ID NO:10). The deduced amino acid sequence of the cDNA is shown in sequence listing (SEQ ID NO:11)

EXAMPLE 7 cDNA Cloning of Anti-Mouse CD3&egr; Antibody

[0172] (1) Preparation of RNA

[0173] Anti-mouse CD3&egr; antibody producing hybridoma (145-2C11: manufactured by Pharmingen) cells were cultured in Hybridoma SFM medium (manufactured by Invitrogen) at 37° C. under 5% CO2, a few days after 5×106 hybrisoma cells were lysed with 1 ml of TRIzol (manufactured by Invitrogen). Total RNA was prepared according to the direction of attached document.

[0174] (1) Preparation of cDNA Library

[0175] cDNA was synthesized from 2.5 &mgr;g of total RNA extracted from hybridoma (145-2C11) cells by oligo-dT prime method using Ready-To-Go You-Prime First-Strand Beads (manufactured by Amersham Pharmacia). Operations and procedures were followed by the instructions of attached document.

[0176] (3) cDNA Cloning of Heavy and Light Chains

[0177] Based upon the reported cDNA sequence information of heavy chain variable region of hybridoma 145-2C11 (GenBank Accession No.AF000357), primers No.7 and No.8 were designed. Also, based upon the cDNA sequence information of light chain variable region of 145-2C11 (GenBank Accession No.AF000356), primers No.9 and No.10 were designed. PCR was carried out using these primers and the cDNA library from hybridoma 145-2C11 as a template. 3 Primer No. 7 5′-gag gtg cag ctg gtg gag tct-3′ (SEQ ID NO:12) Primer No. 8 5′-tga gga gac ggt gac cat ggt t-3′ (SEQ ID NO:13) Primer No. 9 5′-gac atc cag atg acc cag tct c-3′ (SEQ ID NO:14) Primer No. 10 5′-ttt gat ttc cag ctt ggt gcc ag-3′ (SEQ ID NO:15) cDNA library 2 Primer No. 7 or No. 9 (5 pmol/&mgr;l) 2 Primer No. 8 or No. 10 (5 pmol/&mgr;l) 2 dH2O 20 One shot LA PCR Mix™ 25 50 &mgr;l (94° C. 30 sec., 52° C. 30 sec., 72° C. 120 sec.) × 30 cycles

[0178] The PCR products were subjected to electrophoresis using 1% Agarose gel, then the gel was stained by EtBr. DNA fragments were recovered from the gel by using MinElute Gel Extraction Kit (manufactured by Quiagen), the DNA fragments recovered was ligated to pGEM-T Easy Vector (manufactured by Promega) by using DNA Ligation Kit ver.2 (manufactured by Takara Shuzo). E. coli DH5&agr; were transformed with the ligated plasmids. Finally, the plasmids were purified from E. coli, and both DNA were sequenced. It was confirmed that their sequences were identical to those of GenBank Accession No.AF000357and GenBank Accession No.AF000356, respectively.

EXAMPLE 8 Constraction of Expression Plasmid for J43-2C11 Bispecific Antibody

[0179] J43 IgG heavy chain cDNA and 145-2C11 IgG light chain cDNA were connected by PCR using linker No.1, No.2, primer No.11 and No.12 to prepare fragment 1 (see FIG. 5). Next, 145-2C11 IgG light chain cDNA and 145-2C11 IgG heavy chain cDNA were connected by PCR using linker No.3, No.4, primer No.13 and No.14 to prepare fragment 2 (see FIG. 5). 145-2C11 IgG heavy chain cDNA and J43 IgG light chain cDNA were connected by PCR using linker No.5, No.6, primer No.15and No.16 to prepare fragment 3 (see FIG. 5). 4 Primer No. 11 5′-ttt gaa ttc aga ggt gcg gct tct gga gtc t-3′ (SEQ ID NO:16) Primer No. 12 5′-gat cag gag ctt agg agc ttt cc-3′ (SEQ ID NO:17) Primer No. 13 5′-cag gcc agt cag gac att agc aa-3′ (SEQ ID NO:18) Primer No. 14 5′-taa tgt atg cga ccg act cca gc-3′ (SEQ ID NO:19) Primer No. 15 5′-tga ggc ctc tgg att cac ctt ca-3′ (SEQ ID NO:20) Primer No. 16 5′-aaa aaa aaa ctc gag gac cta gga cgg tga gct ggg t-3′ (SEQ ID NO:21) Linker No. 1 5′-agg gac cca agt cac tgt ctc ctc agg tgg agg cgg ttc aga (SEQ ID NO:22) cat cca gat gac cca gtc tcc at-3′ Linker No. 2 5′-tcc ctg ggt tca gtg aca gag gag tcc acc tcc gcc aag tct (SEQ ID NO:23) gta ggt cta ctg ggt cag agg ta-3′ Linker No. 3 5′-acc tgg cac caa gct gga aat caa agg tgg agg cgg ttc agg (SEQ ID NO:24) cgg agg tgg ctc tgg cgg tgg cgg atc gga ggt gca gct ggt gga gtc tgg gg-3′ Linker No. 4 5′-tgg acc gtg gtt cga cct tta gtt tcc acc tcc gcc aag tcc (SEQ ID NO:25) gcc tcc acc gag acc gcc acc gcc tag cct cca cgt cga cca cct cag acc cc-3′ Linker No. 5 5′-agg aac cat ggt cac cgt ctc ctc agg tgg agg cgg ttc ata (SEQ ID NO:26) tga gct gac tca gcc acc ttc ag-3′ Linker No. 6 5′-tcc ttg gta cca gtg gca gag gag tcc acc tcc gcc aag tat (SEQ ID NO:27) act cga ctg agt cgg tgg aag tc-3′ PCR condition for fragments 1, 2 and 3 First PCR Template 1 2 Template 2 2 Linker (100 ng/&mgr;l) 2 Linker (100 ng/&mgr;l) 2 dH2O 17 One shot LA PCR Mix™ 25 50 &mgr;l 95° C. 5 min., → (94° C. 30 sec., 40° C. 30 sec., 72° C. 60 sec.) × 20 cycles Second PCR Product of first PCR 5 Primer (5 pmol/&mgr;l) 2 Primer (5 pmol/&mgr;l) 2 dH2O 16 One shot LA PCR Mix™ 25 50 &mgr;l 95° C. 5 min., → (94° C. 30 sec., 50° C. 30 sec., 72° C. 60 sec.) × 30 cycles

[0180] The DNA fragments 1, 2 and 3 and plasmid pBluescriptII SK(+) (manufactured by StrateGene) were digested with restriction enzymes EcoRI/KpnI, KpnI/SphI, SphI/XhoI, EcoRI/XhoI, respectively. After electrophoresis on 1% agarose gel, the DNA fragments were purified from the gel using MinElute Gel Extraction Kit. Next, these three fragments and plasmid pBluescriptII SK(+) (manufactured by StrateGene) were connected using DNA Ligation Kit ver.2 (manufactured by Takara Shuzo), then E. coli DH5&agr; were transformed with the connected plasmid. Plasmid J43-2C11scDb-pBluescriptII SK(+) was prepared from E. coli, then the nucleotide sequence of the insert was determined (SEQ ID NO:28). The deduced amino acid sequence is shown in sequence listing (SEQ ID NO:29).

[0181] J43-2C11scDb-pBluescriptII SK(+) was digested with restriction enzymes BamHI and XhoI, the BamHI-XhoI fragment out of BamHI-XhoI and BamHI-BamHI fragments generated by the digestion was connected with BamHI/XhoI digested mammalian expression vector pSecTag2/Hygro B (manufactured by Invitrogen). Next, the pSecTag2/Hygro B connected with BamHI-XhoI fragment was digested again with restriction enzyme BamHI, and connected with the other BamHI-BamHI fragment. E. coli DH5&agr; were transformed with the connected plasmid. Finally, Plasmid J43-2C11scDb-pSec/Hygro B was prepared from E. coli, then the nucleotide sequence of thus prepared J43-2C11bispecific antibody was determined (SEQ ID NO:30). The deduced amino acid sequence is shown in sequence listing (SEQ ID NO:31)

EXAMPLE 9 Expression of J43-2C11 Bispecific Antibody

[0182] 6×106 293 T cells were suspended in 20 ml of medium (DMEM containing 10% FBS) and seeded into 150 mm dish coated with Type Ic collagen. Next day, the cells were washed with 10 ml of DMEM and transfected with J43-2C11scDb-pSec/Hygro B by using LipofectAMINE-plus. After 3 hours, 5 ml of DMEM containing 40% FBS were added to the cells. At day 2, the cells were washed with 10 ml of DMEM, added 20 ml of new DMEM, and the culture supernatant was recovered at day 4.

[0183] The cells were discarded by centrifugation, then the supernatant was filtrated using 0.22 &mgr;m PVDF filter. The supernatant was enclosed in dialysis tube, dialyzed against PBS containing 40% PEG20000, and concentrated. The concentrated supernatant was purified using HiTrap chelating HP column (manufactured by Amersham Farmacia). To purify further, the antibodies were purified by gel-filtration using Hiprep 16/60 Sephacryl S-200 High Resolution (manufactured by Amersham Farmacia).

EXAMPLE 10 Suppression of T Cell Activation

[0184] Spleen was removed from BALB/c mouse, and cells were prepared using CellStrainer (70 &mgr;m Nyron). The cells were recovered by centrifugation, and red blood cells were hemolyzed by the addition of hemolysis buffer [NH4Cl (0.8%), KCO3 (0.1%) and EDTA (1 mM)]. The cells were washed with PBS (−) once, T cells were enriched by using mouse CD3+T cell enrichment column kit (manufactured by R&D), and suspended in medium (RPMI1640 containing 10% FBS) in the proportion of 5×106 cells/ml. The T cells thus prepared were seeded on 96-well plates (2×105 cells/100 &mgr;l /well), which were coated preliminarily with 5 &mgr;g/ml of anti-CD3 antibodies (clone KT3) at 37° C. for 3 hours, added with J43-2C11 bispecific antibody diluted in the medium (0.01, 0.03, 0.1, 0.3, 1 and 3 &mgr;g/100 &mgr;l ), and cultivated for 72 hours at 37° C. under 5% CO2. After 72 hours, the culture supernatants were recovered, and the concentrations of IFN-r in the supernatants were determined by using Quantikine Immunoassay Kit (manufactured by R&D).

[0185] As shown in FIG. 6, J43-2C11 bispecific antibody suppressed dose dependently the production of IFN-r from activated mouse spleen T cells in vitro.

Claims

1. A substance comprising a substance that recognizes PD-1, a substance that recognizes a membrane protein co-existing with PD-1 on a cell membrane, and a linker.

2. The substance according to claim 1, which is a divalent substance comprising a substance that recognizes PD-1, a substance that recognizes a membrane protein co-existing with PD-1 on a cell membrane, and a linker.

3. The substance according to claim 1, in which a membrane protein is a protein existing on a T cell membrane or B cell membrane.

4. The substance according to claim 1, which comprises a substance that recognizes PD-1, a substance that recognizes a protein constituting T cell receptor complex or a substance that recognizes a protein constituting B cell receptor complex, and a linker.

5. The substance according to claim 1, in which a substance that recognizes PD-1 and in which a substance that recognizes a protein is dimer to pentamer, respectively.

6. The substance according to claim 1, in which one of or both substance that recognizes PD-1 and substance that recognizes a protein are an antibody.

7. The substance according to claim 1, in which one of the two or both substance that recognizes PD-1 and substance that recognizes a protein are a Fab portion of antibody.

8. The substance according to claim 1, in which a linker comprises an organic compound.

9. The substance according to claim 1, in which a linker comprises a peptide.

10. The substance according to claim 1, in which a substance that recognizes PD-1 and in which a substance that recognizes a protein is a peptide, respectively.

11. The substance according to claim 1, in which a substance that recognizes PD-1 and in which a substance that recognizes a protein comprises two or more peptides including a heavy chain variable region and a light chain variable region of antibody, respectively.

12. The substance according to claim 1, in which a substance that recognizes PD-1, in which a substance that recognizes a protein, and in which a linker is a peptide, respectively.

13. A pharmaceutical composition containing an effective dose of the substance according to claim 1 for the medical treatment and/or prevention of a disease in which PD-1 participates.

14. The pharmaceutical composition according to claim 13, in which disease is selected from the group consisting of neurodegenerative disease, autoimmune disease, organ transplant rejection, neoplasm and infection.

15. The pharmaceutical composition according to claim 14, in which neulodegenerative disease is selected from the group consisting of Parkinson's disease, parkinsonian syndrome, Huntington's disease, Machado-Joseph disease, amyotrophic lateral sclerosis and Creutzfeldt-Jakob disease.

16. The pharmaceutical composition according to claim 14, in which autoimmune disease is selected from the group consisting of glomerular nephritis, arthritis, dilated cardiomyopathy-like disease, ulcerative colitis, Sjogren's syndrome, Crohn's disease, systemic lupus erythematosus, chronic rheumatoid arthritis, multiple sclerosis, Psoriasis, allergic contact dermatitis, polymyositis, scleroderma, periarteritis nodosa, rheumatic fever, vitiligo vulgaris, insulin-dependent diabetes mellitus, Behcet's Syndrome and chronic thyroiditis.

17. A method for the medical treatment and/or prevention of a disease in which PD-1 participates which comprises administering to a subject in need thereof an effective amount of the substance according to claim 1.

18. The method according to claim 17, in which disease is selected from the group consisting of neurodegenerative disease, autoimmune disease, organ transplant rejection, neoplasm and infection.

19. The method according to claim 18, in which neurodegenerative disease is selected from the group consisting of Parkinson's disease, parkinsonian syndrome, Huntington's disease, Machado-Joseph disease, amyotrophic lateral sclerosis and Creutzfeldt-Jakob disease.

20. The method according to claim 18, in which autoimmune disease is selected from the group consisting of glomerular nephritis, arthritis, dilated cardiomyopathy-like disease, ulcerative colitis, Sjogren's syndrome, Crohn's disease, systemic lupus erythematosus, chronic rheumatoid arthritis, multiple sclerosis, Psoriasis, allergic contact dermatitis, polymyositis, scleroderma, periarteritis nodosa, rheumatic fever, vitiligo vulgaris, insulin-dependent diabetes mellitus, Behcet's Syndrome and chronic thyroiditis.

Patent History
Publication number: 20040241745
Type: Application
Filed: Feb 2, 2004
Publication Date: Dec 2, 2004
Inventors: Tasuku Honjo (Kyoto-shi), Shiro Shibayama (Mishima-gun Osaka), Masayoshi Matsuo (Mishima-gun Osaka), Takao Yoshida (Mishima-gun)
Application Number: 10485466