WO2010137654A1 - Egfファミリーリガンドのアンタゴニストを成分とする医薬組成物 - Google Patents
Egfファミリーリガンドのアンタゴニストを成分とする医薬組成物 Download PDFInfo
- Publication number
- WO2010137654A1 WO2010137654A1 PCT/JP2010/059008 JP2010059008W WO2010137654A1 WO 2010137654 A1 WO2010137654 A1 WO 2010137654A1 JP 2010059008 W JP2010059008 W JP 2010059008W WO 2010137654 A1 WO2010137654 A1 WO 2010137654A1
- Authority
- WO
- WIPO (PCT)
- Prior art keywords
- antibody
- seq
- amino acid
- acid sequence
- set forth
- Prior art date
Links
Images
Classifications
-
- C—CHEMISTRY; METALLURGY
- C07—ORGANIC CHEMISTRY
- C07K—PEPTIDES
- C07K16/00—Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
- C07K16/18—Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
- C07K16/22—Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against growth factors ; against growth regulators
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K39/00—Medicinal preparations containing antigens or antibodies
- A61K39/395—Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum
- A61K39/39533—Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum against materials from animals
- A61K39/3955—Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum against materials from animals against proteinaceous materials, e.g. enzymes, hormones, lymphokines
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K45/00—Medicinal preparations containing active ingredients not provided for in groups A61K31/00 - A61K41/00
- A61K45/06—Mixtures of active ingredients without chemical characterisation, e.g. antiphlogistics and cardiaca
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61P—SPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
- A61P1/00—Drugs for disorders of the alimentary tract or the digestive system
- A61P1/04—Drugs for disorders of the alimentary tract or the digestive system for ulcers, gastritis or reflux esophagitis, e.g. antacids, inhibitors of acid secretion, mucosal protectants
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61P—SPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
- A61P1/00—Drugs for disorders of the alimentary tract or the digestive system
- A61P1/18—Drugs for disorders of the alimentary tract or the digestive system for pancreatic disorders, e.g. pancreatic enzymes
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61P—SPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
- A61P11/00—Drugs for disorders of the respiratory system
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61P—SPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
- A61P13/00—Drugs for disorders of the urinary system
- A61P13/12—Drugs for disorders of the urinary system of the kidneys
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61P—SPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
- A61P35/00—Antineoplastic agents
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61P—SPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
- A61P35/00—Antineoplastic agents
- A61P35/04—Antineoplastic agents specific for metastasis
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61P—SPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
- A61P43/00—Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00
-
- G—PHYSICS
- G01—MEASURING; TESTING
- G01N—INVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
- G01N33/00—Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
- G01N33/48—Biological material, e.g. blood, urine; Haemocytometers
- G01N33/50—Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
- G01N33/5005—Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving human or animal cells
- G01N33/5008—Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving human or animal cells for testing or evaluating the effect of chemical or biological compounds, e.g. drugs, cosmetics
- G01N33/5011—Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving human or animal cells for testing or evaluating the effect of chemical or biological compounds, e.g. drugs, cosmetics for testing antineoplastic activity
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K39/00—Medicinal preparations containing antigens or antibodies
- A61K2039/505—Medicinal preparations containing antigens or antibodies comprising antibodies
- A61K2039/507—Comprising a combination of two or more separate antibodies
-
- C—CHEMISTRY; METALLURGY
- C07—ORGANIC CHEMISTRY
- C07K—PEPTIDES
- C07K2317/00—Immunoglobulins specific features
- C07K2317/20—Immunoglobulins specific features characterized by taxonomic origin
- C07K2317/24—Immunoglobulins specific features characterized by taxonomic origin containing regions, domains or residues from different species, e.g. chimeric, humanized or veneered
-
- C—CHEMISTRY; METALLURGY
- C07—ORGANIC CHEMISTRY
- C07K—PEPTIDES
- C07K2317/00—Immunoglobulins specific features
- C07K2317/30—Immunoglobulins specific features characterized by aspects of specificity or valency
- C07K2317/31—Immunoglobulins specific features characterized by aspects of specificity or valency multispecific
-
- C—CHEMISTRY; METALLURGY
- C07—ORGANIC CHEMISTRY
- C07K—PEPTIDES
- C07K2317/00—Immunoglobulins specific features
- C07K2317/50—Immunoglobulins specific features characterized by immunoglobulin fragments
- C07K2317/56—Immunoglobulins specific features characterized by immunoglobulin fragments variable (Fv) region, i.e. VH and/or VL
- C07K2317/565—Complementarity determining region [CDR]
-
- C—CHEMISTRY; METALLURGY
- C07—ORGANIC CHEMISTRY
- C07K—PEPTIDES
- C07K2317/00—Immunoglobulins specific features
- C07K2317/70—Immunoglobulins specific features characterized by effect upon binding to a cell or to an antigen
- C07K2317/73—Inducing cell death, e.g. apoptosis, necrosis or inhibition of cell proliferation
-
- C—CHEMISTRY; METALLURGY
- C07—ORGANIC CHEMISTRY
- C07K—PEPTIDES
- C07K2317/00—Immunoglobulins specific features
- C07K2317/70—Immunoglobulins specific features characterized by effect upon binding to a cell or to an antigen
- C07K2317/76—Antagonist effect on antigen, e.g. neutralization or inhibition of binding
-
- G—PHYSICS
- G01—MEASURING; TESTING
- G01N—INVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
- G01N2333/00—Assays involving biological materials from specific organisms or of a specific nature
- G01N2333/435—Assays involving biological materials from specific organisms or of a specific nature from animals; from humans
- G01N2333/46—Assays involving biological materials from specific organisms or of a specific nature from animals; from humans from vertebrates
- G01N2333/47—Assays involving proteins of known structure or function as defined in the subgroups
- G01N2333/4701—Details
- G01N2333/4703—Regulators; Modulating activity
-
- G—PHYSICS
- G01—MEASURING; TESTING
- G01N—INVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
- G01N2333/00—Assays involving biological materials from specific organisms or of a specific nature
- G01N2333/435—Assays involving biological materials from specific organisms or of a specific nature from animals; from humans
- G01N2333/475—Assays involving growth factors
- G01N2333/485—Epidermal growth factor [EGF] (urogastrone)
Definitions
- the present invention relates to a pharmaceutical composition
- a pharmaceutical composition comprising an EGF family ligand antagonist as a component.
- ErbB receptor family and EGF family ligand Epidermal growth factor receptor (EGFR) family
- the erbB receptor has 4 members from EGFR (erbB1), erbB2, erbB3, erbB4, and the EGF family ligand has 13 members EGF, TGF ⁇ (transforming growth factor ⁇ ), HB-EGF (heparin-binding EGF-like growth factor), BTC (betacellulin), AREG (amphiregulin), EREG (epiregulin), EPGN (epigen), NRGs (neuregulin1-6) (Refer nonpatent literature 3).
- Activation of the erbB receptor is controlled by the presence of ligand (based on spatiotemporal expression control) and post-translational processing.
- the EGF family of ligands is produced as a type I membrane protein, which undergoes a process by protease from the transmembrane type and functions as a soluble ligand.
- some of the ligands activate receptors on neighboring cells while remaining in a membrane form.
- the receptor activation mechanism by EGF family ligands is the endocrine, in which the secretory peptide spreads throughout the bloodstream and acts on the receptor-expressing cells, the autocrine, in which the secretory peptide stimulates the receptor of the expressing cell itself, the secretory peptide Consists of a paracrine that activates an on-cell receptor in the vicinity, and a juxtaclein that a membrane ligand stimulates an adjacent cell receptor (see Non-Patent Document 6).
- EGF family ligands cause EGFR / EGFR homodimer formation, heterodimer formation such as EGFR / erbB2, EGFR / erbB4, etc., activates protein tyrosine kinases and causes receptor autophosphorylation. Phosphorylated receptors attract several types of intracellular substrates and are consequently converted into cell growth stimuli and other cellular activities.
- EGF family ligands show different affinities and elaborate receptor binding specificities for each erbB receptor. Surprisingly, despite the fact that they bind to the same receptor, individual EGF family ligands cause various non-identical physiological responses in cells (see Non-Patent Document 11).
- TGF ⁇ and AREG stimulate DNA synthesis to the same extent in MDCK cells. AREG causes E-cadherin redistribution and morphological changes in MDCK cells, but not TGF ⁇ . In human mammary epithelial cell MCF10A, AREG enhances cell motility and invasive ability more than EGF. AREG stimulates NF ⁇ B signaling and interleukin-1 secretion, but not EGF. TGF ⁇ transmits signals through effector molecules different from EGF. AREG, EGF, and TGF ⁇ bind selectively to EGFR among erbB family receptors. The above differences are not explained by the binding saturation of EGFR simply due to affinity strength.
- mice deficient in AREG, BTC, EGF, EREG, and TGF ⁇ each show several characteristic phenotypes, but are viable.
- HB-EGF is an exception to this, and its gene deficiency causes damage to the heart and lungs, leading to embryonic lethality.
- AREG-deficient mice have impaired liver regeneration.
- TGF ⁇ -deficient mice abnormalities were observed in closing the hair follicles and wrinkles.
- the difference in affinity of EGF member ligands for the four receptors is one factor that determines the ligand signaling specificity.
- the physiological difference of the ligand is due to the difference in the conformation of the receptor that occurs when the ligand binds to the receptor, the change in the phosphorylation site of the receptor tyrosine residue due to the difference in the conformation, It is also believed that this may be due to a difference in coupling with a signal coupling factor (see Non-Patent Document 11).
- ErbB receptor activation is important for determining cell fate, for example, affecting proliferation, survival and differentiation. Therefore, abnormal erbB receptor activation plays an important role in human tumorigenesis. In addition, erbB receptor stimulation by function-selective ligands may be involved in human tumor growth.
- Cancer and erbB receptor signaling EGFR mutations and increased expression in human epithelial cancer have been reported previously, and it is clear that enhanced activity of EGFR itself or its downstream signals has an important role in cancer development and progression Has been.
- Increased tyrosine phosphorylation activity of mutated EGFR, activation of EGFR by ligand through autocrine loop, amplification of receptor chromosomal gene, various cancers such as breast cancer, prostate cancer, pancreatic cancer, colon cancer, lung cancer, head and neck cancer This is a common feature found in cancer. It has been experimentally shown to induce canceration traits by introducing activated EGFRs into non-tumor cell lines.
- EGFR on cancer cells is often constitutively activated by the presence of EGF ligands present in the tumor.
- EGF ligands present in the tumor.
- cancers expressing TGF ⁇ , AREG, and HB-EGF are associated with poor prognosis and drug resistance.
- K-RAS constitutive activation of the EGFR signal pathway induces EREG expression and enhances tumorigenicity (see Non-Patent Document 1).
- EGFRs are attractive target candidates for molecular targeted therapy.
- EGFR and erbB2 have been confirmed to be useful as targets for therapeutic agents in treating colorectal cancer, lung cancer, head and neck cancer, breast cancer, and gastric cancer.
- the use of anti-EGFR antibodies is an essential therapy for patients with metastatic colorectal cancer. This is based on evidence of prolonged response-free progression and high reactivity.
- the signal network of EGFR family receptors is not only essential for the growth and survival of cancer cells, but also has an essential role in maintaining the homeostasis of the living body that controls the growth and differentiation of normal cells. Inhibition of EGF receptors suppresses signals essential for maintaining homeostasis in the living body, resulting in serious side effects.
- side effects of the anti-EGFR antibody hypersensitivity, lung injury, skin symptom, photosensitivity, fever and myocardial injury have been reported (see Non-Patent Document 5).
- skin toxicity is seen in many cases because EGFR signaling plays an important role in the skin. Moderate to severe toxicity may cause a change in dosage and schedule, and treatment may need to be interrupted.
- hypomagnesemia occurs frequently by administration of anti-EGFR antibody (see Non-Patent Document 8). In some cases, treatment must be interrupted due to serious side effects. This hypomagnesemia has been shown to be associated with magnesium consumption in the distal convoluted tubule. It has been shown that when the kidney EGFR is not sufficiently stimulated, the epithelial magnesium channel TRPM6 is not sufficiently activated and magnesium is consumed (see Non-Patent Document 9). It has been shown that EGF is involved in activation of renal EGFR.
- the activity of the EGFR family receptor by the EGF family ligand is very complicated. Although an approach of selecting an EGF family ligand as a target for a cancer drug can be considered, an approach targeting a receptor is currently preferred. The reason is that there are multiple ligands that activate EGFR and erbB4, no activation ligand is known for erbB2, and there may be unidentified activation ligands.
- the present inventor has conducted intensive research to solve the above-mentioned problems, and has identified a combination of EGFR ligand inhibition that is a target for inhibiting the growth of cancer cells. More specifically, it has been found that EREG antagonists and TGF ⁇ antagonists are useful as cytostatic agents.
- the present invention relates to a pharmaceutical composition comprising an antagonist of an EGF family ligand as a component, more specifically, [1] A pharmaceutical composition comprising an EREG antagonist and a TGF ⁇ antagonist as active ingredients, [2] The pharmaceutical composition according to [1], wherein the pharmaceutical composition is a cell growth inhibitor, [3] The pharmaceutical composition according to [1], wherein the pharmaceutical composition is an anticancer agent, [4] The pharmaceutical composition according to any one of [1] to [3], wherein the EREG antagonist is an anti-EREG antibody, [5] The pharmaceutical composition according to any one of [1] to [4], wherein the anti-EREG antibody is an antibody according to any one of the following (a) to (x): (A) an antibody comprising a heavy chain comprising CDR1 having the amino acid sequence set forth in SEQ ID NO: 61, CDR2 having the amino acid sequence set forth in SEQ ID NO: 62, and CDR3 having the amino acid sequence set forth in SEQ ID NO: 63; (B) an antibody compris
- the present invention also provides the following.
- a method for inhibiting cell proliferation comprising a step of administering a pharmaceutical composition of the present invention (for example, an EREG antagonist and a TGF ⁇ antagonist) to a subject.
- a method for treating cancer comprising a step of administering a pharmaceutical composition of the present invention (for example, an EREG antagonist and a TGF ⁇ antagonist) to a subject.
- EGF (R) cells proliferate in response to EREG.
- the growth of EGF (R) is promoted by co-culture with EREG / DG. The accelerated proliferation is canceled by the addition of anti-EREG antibody and anti-EGFR antibody.
- EGF (R) growth is promoted by co-culture with DLD-1. Accelerated proliferation is canceled by the addition of both anti-EREG and anti-TGF ⁇ antibodies, or the addition of anti-EGFR antibodies.
- the proliferation of DLD-1 is suppressed by the addition of both anti-EREG antibody and anti-TGF ⁇ antibody, or the addition of anti-EGFR antibody.
- the horizontal axis indicates the concentration of the topoisomerase inhibitor Campto added to the culture solution.
- the upper panel shows the EGF domain sequence alignment of human EREG and human TGF ⁇ (SEQ ID NOs: 35 and 36, respectively).
- the lower panel shows the amino acid residues conserved in EREG and TGF ⁇ mapped onto the EREG conformation (PDB: 1K37). * In the 3D structure diagram indicates the region where amino acids conserved in EREG and TGF ⁇ are gathered.
- Anti-EREG chimeric antibody EP19 binds to EREG and TGF ⁇ .
- EREG on the cell surface increases with increasing cell density in Caki1.
- EREG expression is also induced by adding soluble EREG at low density.
- the upper panel shows the results of flow cytometric analysis of EREG protein (bold line) and EGFR protein (dashed line) on the cell membrane.
- the lower panel is a photograph showing cell confluency.
- AREG protein and EREG protein on the surface of DLD-1 cells were detected by flow cytometry.
- Mouse # 2-20 antibody binds to TGF ⁇ .
- EGF (R) cells proliferate upon addition of TGF ⁇ .
- B EGF (R) cell proliferation by TGF ⁇ is neutralized by addition of mouse # 2-20 antibody.
- Biotinylated TGF ⁇ binds to soluble EGFR protein.
- Binding of biotinylated TGF ⁇ to soluble EGFR protein is inhibited by mouse # 2-20 antibody and goat polyclonal anti-TGF ⁇ antibody.
- the recombinant antibody binds to TGF ⁇ .
- the recombinant antibody inhibits the binding of biotinylated TGF ⁇ to the EGFR protein.
- the present invention provides a pharmaceutical composition comprising an EREG antagonist and a TGF ⁇ antagonist as active ingredients.
- an EREG antagonist refers to a substance that inhibits the binding of EREG and EGFR by binding to EREG and inhibits the signaling of EREG through EGFR or a substance that reduces the expression level of EREG. .
- the EREG antagonist is not particularly limited and may be any substance as long as it has the above-described activity.
- Examples of the substance that inhibits the binding of EREG and EGFR by binding to EREG and inhibits the signal transduction of EREG via EGFR include, for example, an antibody having antagonistic activity against EREG.
- Examples of substances that reduce the expression level of EREG include, for example, siRNA and antisense oligonucleotides against the EREG gene.
- the EREG antagonist is preferably an antagonist against human EREG.
- the sequence of human EREG is already known (GenBank Accession No: NM_001432, etc.).
- the EREG antagonist may target soluble EREG, or may target transmembrane EREG. Further, both may be targeted.
- the EREG antagonist may be any substance, and for example, an antagonist described in WO2008 / 047723, a commercially available anti-human EREG antibody (R & D® Systems, AF1195) or the like can be used.
- the antibody described in any of the following can be mentioned as a preferred embodiment;
- the TGF ⁇ antagonist refers to a substance that inhibits the binding between TGF ⁇ and EGFR by binding to TGF ⁇ and inhibits the signal transduction of TGF ⁇ via EGFR or reduces the expression level of TGF ⁇ .
- the TGF ⁇ antagonist is not particularly limited and may be any substance as long as it has the above-described activity.
- Examples of the substance that inhibits the binding of TGF ⁇ and EGFR by binding to TGF ⁇ and inhibits the signal transduction of TGF ⁇ via EGFR include, for example, an antibody having antagonistic activity against TGF ⁇ .
- Examples of substances that reduce the expression level of TGF ⁇ include siRNA and antisense oligonucleotides against the TGF ⁇ gene.
- the TGF ⁇ antagonist is preferably an antagonist against human TGF ⁇ .
- the sequence of human TGF ⁇ is already known (GenBank Accession No: NM_003236, M31172, etc.).
- the TGF ⁇ antagonist may target soluble TGF ⁇ or may target transmembrane TGF ⁇ . Further, both may be targeted.
- the TGF ⁇ antagonist may be any substance, and for example, an antagonist described in WO2004 / 076622 or a commercially available anti-TGF ⁇ antibody (GeneTex, GTX16811; R & D Systems, AB-239-NA) can be used.
- an antibody described in any of the following can be mentioned as a preferred embodiment;
- A an antibody comprising a heavy chain comprising CDR1 having the amino acid sequence set forth in SEQ ID NO: 47, CDR2 having the amino acid sequence set forth in SEQ ID NO: 48, CDR3 having the amino acid sequence set forth in SEQ ID NO: 49,
- B an antibody comprising a light chain comprising CDR1 having the amino acid sequence set forth in SEQ ID NO: 50, CDR2 having the amino acid sequence set forth in SEQ ID NO: 51, and CDR3 having the amino acid sequence set forth in SEQ ID NO: 52;
- C an antibody comprising the heavy chain of (a) and the light chain of (b),
- D An antibody that recognizes the same epitope as that recognized by the antibody of (c).
- the EREG antagonist and the TGF ⁇ antagonist contained in the pharmaceutical composition containing the EREG antagonist and the TGF ⁇ antagonist of the present invention as active ingredients may be the same substance or different substances. That is, a substance having both EREG antagonist activity and TGF ⁇ antagonist activity may be used as an active ingredient, or a substance having only EREG antagonist activity and a substance having only TGF ⁇ antagonist activity may be used as active ingredients. Furthermore, a substance having both EREG antagonist activity and TGF ⁇ antagonist activity and a substance having only EREG antagonist activity, or a substance having both EREG antagonist activity and TGF ⁇ antagonist activity and a substance having only TGF ⁇ antagonist activity are effective. It may be an ingredient.
- the present invention provides antibodies that bind to EREG and TGF ⁇ .
- the antibody that binds to EREG and TGF ⁇ is an antibody that binds to human EREG and human TGF ⁇ .
- Human EREG and human TGF ⁇ are as described above. Examples of the binding to EREG include binding to soluble EREG, binding to transmembrane EREG, binding to soluble EREG and transmembrane EREG, and the like. Examples of the binding to TGF ⁇ include binding to soluble TGF ⁇ , binding to transmembrane TGF ⁇ , binding to soluble TGF ⁇ and transmembrane TGF ⁇ , and the like.
- the antibody that binds to EREG and TGF ⁇ of the present invention is not particularly limited, but is preferably an antagonist antibody having antagonist activity against EREG or TGF ⁇ , more preferably an antagonist antibody having antagonist activity against EREG and TGF ⁇ . .
- the antibody that binds to human EREG and TGF ⁇ is not particularly limited, but it is preferable to recognize a common region having high homology in EREG and TGF ⁇ .
- a region As an example of such a region, the region from the 56th Gln to the 102nd Leu of the amino acid sequence described in SEQ ID NO: 4 (human EREG) in human EREG, SEQ ID NO: 26 (human TGF ⁇ in human TGF ⁇ )
- SEQ ID NO: 26 human TGF ⁇ in human TGF ⁇
- the region from the 41st Val to the 87th Leu of the amino acid sequence described in (1) can be exemplified.
- the region from the 56th Gln to the 102nd Leu of the amino acid sequence described in SEQ ID NO: 4 (human EREG) or SEQ ID NO: 26 an antibody that recognizes the region from the 41st Val to the 87th Leu of the amino acid sequence described in human (TGF ⁇ ) can be mentioned, and a more preferred example is the amino acid sequence described in SEQ ID NO: 4 (human EREG).
- Examples include an antibody that recognizes a region from the 56th Gln to the 102nd Leu and a region from the 41st Val to the 87th Leu of the amino acid sequence described in SEQ ID NO: 26 (human TGF ⁇ ).
- the base sequence of human EREG is shown in SEQ ID NO: 3, and the base sequence of human TGF ⁇ is shown in SEQ ID NO: 25.
- antibody that binds to EREG and TGF ⁇ of the present invention include the antibodies described in any of (a) to (d) below.
- A an antibody comprising a heavy chain comprising CDR1 having the amino acid sequence set forth in SEQ ID NO: 10, CDR2 having the amino acid sequence set forth in SEQ ID NO: 12, and CDR3 having the amino acid sequence set forth in SEQ ID NO: 14;
- B an antibody comprising a light chain comprising CDR1 having the amino acid sequence set forth in SEQ ID NO: 16, CDR2 having the amino acid sequence set forth in SEQ ID NO: 18 and CDR3 having the amino acid sequence set forth in SEQ ID NO: 20;
- C an antibody comprising the heavy chain of (a) and the light chain of (b),
- D An antibody that recognizes the same epitope as that recognized by the antibody of (c).
- the base sequence of heavy chain CDR1 is shown in SEQ ID NO: 9
- the base sequence of CDR2 is shown in SEQ ID NO: 11
- the base sequence of CDR3 is shown in SEQ ID NO: 13.
- the base sequence of the light chain CDR1 is shown in SEQ ID NO: 15
- the base sequence of CDR2 is shown in SEQ ID NO: 17
- the base sequence of CDR3 is shown in SEQ ID NO: 19.
- An antibody that recognizes the same epitope as that recognized by a certain antibody can be obtained, for example, by the following method.
- test antibody shares an epitope with a certain antibody by competition for the same epitope.
- Competition between antibodies is detected by a cross-blocking assay or the like.
- a competitive ELISA assay is a preferred cross-blocking assay.
- the anti-EREG antibody of the present invention is added after preincubating the EREG protein coated on the well of the microtiter plate in the presence or absence of the candidate competitive antibody. Is done.
- the amount of anti-EREG antibody of the present invention bound to the EREG protein in the well is indirectly correlated with the binding ability of a candidate competitive antibody (test antibody) that competes for binding to the same epitope. That is, the greater the affinity of the test antibody for the same epitope, the lower the binding amount of the above-described antibody to the well coated with EREG protein and / or the binding amount to the well coated with TGF ⁇ protein. Or conversely, the greater the affinity of the test antibody for the same epitope, the greater the amount of binding of the test antibody to the well coated with EREG protein and / or the amount of binding to the well coated with TGF ⁇ protein.
- the amount of antibody bound to the well can be easily measured by labeling the antibody in advance.
- biotin-labeled antibodies can be measured by using an avidin peroxidase conjugate and an appropriate substrate.
- a cross-blocking assay using an enzyme label such as peroxidase is particularly referred to as a competitive ELISA assay.
- the antibody can be labeled with another labeling substance that can be detected or measured. Specifically, radiolabels or fluorescent labels are known.
- any antibody bound to the well can be measured by a labeled antibody that recognizes any constant region.
- the antibodies bound to the wells can be measured by the antibodies that identify the respective classes.
- the candidate antibody is at least 20%, preferably at least 20-50%, more preferably at least 50%, compared to the binding activity obtained in a control test carried out in the absence of the candidate competing antibody. If the binding can be blocked, the candidate competing antibody is an antibody that binds to substantially the same epitope as described above or competes for binding to the same epitope.
- the antibody that recognizes the same epitope recognized by the antibody of (c) of the present invention may be an antibody that recognizes an epitope in EREG that is recognized by the antibody of (c), or the antibody of (c) It may be an antibody that recognizes an epitope in TGF ⁇ recognized by.
- the antibody recognizes both the epitope in EREG and the epitope in TGF ⁇ recognized by the antibody (c).
- an antibody that recognizes the same epitope as that recognized by the antibody of (c) is preferably an antibody that competes with the antibody of (c) or TGF ⁇ by binding to a well coated with EREG protein in the cross-blocking assay described above.
- the antibody used in the present invention may be bound to the target protein, and its origin, type and shape are not limited. Specifically, known antibodies such as non-human animal antibodies (eg, mouse antibodies, rat antibodies, camel antibodies), human antibodies, chimeric antibodies, and humanized antibodies can be used. In the present invention, it can be used as a monoclonal or polyclonal antibody. A preferred antibody is a monoclonal antibody.
- the antibody used in the present invention is preferably an antibody that specifically binds to the target protein.
- the antibody used in the present invention can be obtained as a polyclonal or monoclonal antibody using known means.
- a monoclonal antibody derived from a mammal is particularly preferable.
- Mammal-derived monoclonal antibodies include those produced by hybridomas and those produced by hosts transformed with expression vectors containing antibody genes by genetic engineering techniques.
- Monoclonal antibody-producing hybridomas can be basically prepared using known techniques as follows. First, EREG protein or TGF ⁇ is used as a sensitizing antigen and immunized according to a normal immunization method. Immune cells obtained from an immunized animal are fused with a known parent cell by a conventional cell fusion method to obtain a hybridoma. Further, from this hybridoma, a hybridoma producing an antibody can be selected by screening cells producing the target antibody by a usual screening method.
- EREG protein or TGF ⁇ protein used as a sensitizing antigen for antibody acquisition can be obtained. That is, after a suitable host cell is transformed by inserting a gene sequence encoding EREG or TGF ⁇ into a known expression vector, the target human EREG protein or human TGF ⁇ protein is extracted from the host cell or culture supernatant. It can be purified by known methods. Purified natural EREG protein or TGF ⁇ protein can also be used as well.
- a fusion protein obtained by fusing a desired partial polypeptide of EREG protein or TGF ⁇ protein with a different polypeptide can also be used as an immunogen.
- an Fc fragment of an antibody, a peptide tag, or the like can be used.
- a vector expressing a fusion protein can be prepared by fusing genes encoding two or more desired polypeptide fragments in frame and inserting the fusion gene into the expression vector as described above. .
- the method for preparing the fusion protein is described in Molecular® Cloning® 2nd® ed. (Sambrook, Jet et al., Molecular Cloning® 2nd® ed., 9.47-9.58, Cold® Spring® Labor® Lab.® press, 1989).
- the EREG protein or TGF ⁇ protein purified in this way can be used as a sensitizing antigen used for immunization against mammals.
- a partial peptide of EREG or TGF ⁇ can also be used as a sensitizing antigen.
- the following peptides can be used as the sensitizing antigen.
- Peptides obtained by chemical synthesis from the amino acid sequence of human EREG or TGF ⁇ protein Peptides obtained by incorporating a part of the EREG gene or TGF ⁇ gene into an expression vector for expression Peptide obtained by degrading EREG protein or TGF ⁇ protein with proteolytic enzyme
- the region and size of EREG or TGF ⁇ used as a partial peptide are not limited.
- a region from the 56th Gln to the 102nd Leu of the amino acid sequence described in SEQ ID NO: 4 (human EREG) or SEQ ID NO: 26 Mention may be made of a polypeptide comprising a region from the 41st Val to the 87th Leu of the amino acid sequence described in human (TGF ⁇ ).
- the mammal immunized with the sensitizing antigen is not particularly limited.
- an immunized animal in consideration of compatibility with a parent cell used for cell fusion.
- rodent animals are preferred as immunized animals. Specifically, mice, rats, hamsters, or rabbits can be used as immunized animals.
- monkeys and the like can be used as immunized animals.
- the above animals can be immunized with a sensitizing antigen.
- mammals can be immunized by injecting a sensitizing antigen intraperitoneally or subcutaneously. Specifically, the sensitizing antigen is administered to mammals several times every 4 to 21 days.
- the sensitizing antigen is diluted with PBS (Phosphate-Buffered Saline) or physiological saline at an appropriate dilution ratio and used for immunization.
- a sensitizing antigen can be administered with an adjuvant. For example, it can be mixed with Freund's complete adjuvant and emulsified to give a sensitizing antigen.
- An appropriate carrier can be used for immunization with the sensitizing antigen.
- a partial peptide having a small molecular weight is used as a sensitizing antigen, it is desirable to immunize the sensitizing antigen peptide by binding it to a carrier protein such as albumin or keyhole limpet hemocyanin.
- immune cells are collected from the mammal and subjected to cell fusion.
- spleen cells can be used.
- Mammalian myeloma cells are used as the cells fused with the immune cells.
- the myeloma cell is preferably provided with an appropriate selection marker for screening.
- a selectable marker refers to a trait that can (or cannot) survive under certain culture conditions.
- Known selection markers include hypoxanthine-guanine-phosphoribosyltransferase deficiency (hereinafter abbreviated as HGPRT deficiency) or thymidine kinase deficiency (hereinafter abbreviated as TK deficiency).
- HGPRT deficiency hypoxanthine-guanine-phosphoribosyltransferase deficiency
- TK deficiency thymidine kinase deficiency
- Cells having HGPRT or TK deficiency have hypoxanthine-aminopterin-thymidine sensitivity (hereinafter abbreviated as HAT sensitivity).
- HGPRT-deficient and TK-deficient cells can be selected in media containing 6 thioguanine, 8 azaguanine (hereinafter abbreviated as 8AG), or 5 'bromodeoxyuridine, respectively.
- 8AG 8 azaguanine
- Normal cells die because they incorporate these pyrimidine analogs into the DNA, but cells deficient in these enzymes cannot survive these pyrimidine analogs and can survive in selective media.
- a selectable marker called G418 resistance confers resistance to 2-deoxystreptamine antibiotics (gentamicin analogs) by a neomycin resistance gene.
- Various myeloma cells suitable for cell fusion are known. For example, the following myeloma cells can be used for the production of the monoclonal antibody in the present invention.
- P3 P3x63Ag8.653
- P3x63Ag8U.1 Current Topics in Microbiology and Immunology (1978) 81, 1-7)
- NS-1 Kohler. G. and Milstein, C. Eur. J. Immunol. (1976) 6, 511-519
- MPC-11 Margulies. DH et al., Cell (1976) 8, 405-415
- SP2 / 0 Shulman, M. et al., Nature (1978) 276, 269-270
- FO de St. Groth, S. F. etal., J. Immunol.
- the cell fusion can be performed in a normal nutrient culture medium in the presence of a cell fusion promoter.
- a cell fusion promoter for example, polyethylene glycol (PEG), Sendai virus (HVJ) or the like can be used.
- an auxiliary agent such as dimethyl sulfoxide can be added as desired in order to increase the fusion efficiency.
- the usage ratio of immune cells and myeloma cells can be set arbitrarily.
- the number of immune cells is preferably 1 to 10 times that of myeloma cells.
- the culture medium used for the cell fusion for example, RPMI1640 culture medium suitable for growth of the myeloma cell line, MEM culture medium, and other normal culture liquids used for this type of cell culture can be used.
- serum supplements such as fetal calf serum (FCS) can be added to the culture medium.
- a predetermined amount of the immune cells and myeloma cells are mixed well in the culture solution, and a target PEG (hybridoma) is formed by mixing a PEG solution preheated to about 37 ° C.
- the In the cell fusion method for example, PEG having an average molecular weight of about 1000 to 6000 can be usually added at a concentration of 30 to 60% (w / v).
- cell fusion agents and the like that are undesirable for the growth of hybridomas are removed by sequentially adding the appropriate culture medium listed above, and then centrifuging to remove the supernatant.
- the hybridoma obtained in this manner can be selected by using a selective culture solution corresponding to the selection marker possessed by the myeloma used for cell fusion.
- a selective culture solution corresponding to the selection marker possessed by the myeloma used for cell fusion.
- cells having HGPRT or TK deficiency can be selected by culturing in a HAT culture solution (a culture solution containing hypoxanthine, aminopterin and thymidine). That is, when HAT-sensitive myeloma cells are used for cell fusion, cells that have succeeded in cell fusion with normal cells can be selectively proliferated in the HAT culture solution.
- the culture using the HAT culture solution is continued for a time sufficient for cells other than the target hybridoma (non-fusion cells) to die.
- the target hybridoma can be selected by culturing for several days to several weeks. Subsequently, by carrying out the usual limiting dilution method, screening and single cloning of the hybridoma producing the target antibody can be performed.
- an antibody that recognizes EREG and / or TGF ⁇ can also be produced by the method described in International Publication WO03 / 104453.
- Desired antibody screening and single cloning can be suitably performed by screening methods based on known antigen-antibody reactions.
- the antigen is bound to a carrier such as beads made of polystyrene or the like, or a commercially available 96-well microtiter plate, and reacted with the culture supernatant of the hybridoma.
- a secondary antibody labeled with an enzyme is reacted. If the culture supernatant contains an antibody of interest that reacts with the sensitizing antigen, the secondary antibody binds to the carrier via this antibody. By detecting the secondary antibody that finally binds to the carrier, it can be determined whether the antibody of interest is present in the culture supernatant.
- EREG protein and / or TGF ⁇ which are practically homogeneous, including those used for immunization, can be preferably used.
- a target antibody can be obtained by sensitizing human lymphocytes with an antigen.
- human lymphocytes are first sensitized with EREG protein or TGF ⁇ protein in vitro.
- the immunized lymphocytes are then fused with an appropriate fusion partner.
- the fusion partner for example, a myeloma cell derived from human and having a permanent division ability can be used (see Japanese Patent Publication No. 1-59878).
- the antibody obtained by this method is a human antibody having binding activity to the target protein.
- the target antibody can also be obtained by administering EREG protein or TGF ⁇ protein as an antigen to a transgenic animal having all repertoires of human antibody genes.
- Antibody-producing cells of the immunized animal can be immortalized by treatment such as cell fusion with an appropriate fusion partner or Epstein-Barr virus infection. It is also possible to isolate human antibodies against the target protein from the immortalized cells thus obtained (see International Publications WO 94/25585, WO 93/12227, WO 92/91818, WO 94/6022). Further, by cloning the immortalized cells, it is possible to clone cells that produce an antibody having the desired reaction specificity. When a transgenic animal is used as an immunized animal, the immune system of the animal recognizes human EREG or human TGF ⁇ as a foreign substance. Therefore, the target human antibody can be easily obtained.
- the hybridoma producing the monoclonal antibody thus produced can be subcultured in a normal culture solution.
- the hybridoma can also be stored for a long time in liquid nitrogen.
- the hybridoma can be cultured according to a usual method, and the target monoclonal antibody can be obtained from the culture supernatant.
- a hybridoma can be administered to a mammal compatible therewith to proliferate and a monoclonal antibody can be obtained as its ascites.
- the former method is suitable for obtaining a highly pure antibody.
- an antibody encoded by an antibody gene cloned from an antibody-producing cell can also be used.
- the cloned antibody gene can be expressed as an antibody by incorporating it into an appropriate vector and introducing it into a host. Methods for isolation of antibody genes, introduction into vectors, and transformation of host cells have already been established (eg, Vandamme, A. M. et al., Eur.J. Biochem. (1990)). 192, 767-775).
- cDNA encoding a variable region (V region) of a target antibody can be obtained from a hybridoma cell that produces the target antibody.
- V region variable region
- RNA is extracted from the hybridoma.
- the following method can be used. Guanidine ultracentrifugation (Chirgwin, J. M. et al., Biochemistry (1979) 18, 5294-5299) AGPC method (Chomczynski, P. et al., Anal. Biochem. (1987) 162, 156-159)
- Extracted mRNA can be purified using mRNA “Purification” Kit (manufactured by GE Healthcare Bioscience) or the like.
- kits for extracting mRNA directly from cells such as QuickPrep mRNA Purification Kit (manufactured by GE Healthcare Bioscience) are also commercially available.
- total mRNA can also be obtained from the hybridoma.
- cDNA encoding the antibody V region can be synthesized using reverse transcriptase.
- cDNA can be synthesized by AMV Reverse Transcriptase First-strand cDNA Synthesis Kit (manufactured by Seikagaku Corporation).
- the desired cDNA fragment is purified from the obtained PCR product and then ligated with vector DNA.
- a recombinant vector is produced, introduced into Escherichia coli or the like and a colony is selected, a desired recombinant vector can be prepared from Escherichia coli that has formed the colony. Whether or not the recombinant vector has the target cDNA base sequence can be confirmed by a known method such as the dideoxynucleotide chain termination method.
- cDNA is synthesized using RNA extracted from a hybridoma cell as a template to obtain a 5'-RACE cDNA library.
- a commercially available kit such as SMART RACE cDNA cDNA amplification kit.
- the antibody gene is amplified by the PCR method using the obtained 5'-RACE ⁇ ⁇ ⁇ ⁇ ⁇ cDNA library as a template.
- Primers for amplifying mouse antibody genes can be designed based on known antibody gene sequences. These primers have different nucleotide sequences for each immunoglobulin subclass. Therefore, it is desirable to determine the subclass in advance using a commercially available kit such as IsoIStrip mouse monoclonal antibody isotyping kit (Roche Diagnostics).
- a primer capable of amplifying genes encoding ⁇ 1, ⁇ 2a, ⁇ 2b, and ⁇ 3 light chains as ⁇ chains and ⁇ chains as heavy chains is used. can do.
- a primer that anneals to a portion corresponding to a constant region close to the variable region is generally used as the 3 ′ primer.
- the primer attached to the 5'-RACE cDNA library preparation kit can be used as the 5'-side primer.
- the PCR product thus amplified can be used to reconstitute an immunoglobulin comprising a combination of heavy and light chains.
- the antibody of interest can be screened using the binding activity of the reconstituted immunoglobulin to EREG and / or TGF ⁇ as an index.
- Antibodies that bind to EREG can be screened, for example, as follows. (1) contacting an EREG with an antibody comprising a V region encoded by cDNA obtained from a hybridoma; (2) detecting the binding between EREG and the antibody, and (3) Step of selecting an antibody that binds to EREG
- a method for detecting the binding between an antibody and EREG is known. Specifically, a test antibody is reacted with EREG immobilized on a carrier, and then a labeled antibody that recognizes the antibody is reacted. When the labeled antibody on the carrier is detected after washing, the binding of the test antibody to EREG can be proved.
- an enzyme active protein such as peroxidase or ⁇ -galactosidase, or a fluorescent substance such as FITC can be used.
- a fixed specimen of cells expressing EREG can also be used.
- an antibody against TGF ⁇ it is more preferable that the binding of the antibody to TGF ⁇ is specific.
- An antibody that binds to TGF ⁇ can be obtained, for example, by the same method as for EREG described above.
- the binding of the antibody to EREG and TGF ⁇ may be specific to these two proteins, other EGFR ligands, etc. It may bind to the protein.
- Antibodies that bind to EREG and TGF ⁇ can be obtained, for example, by combining the above-described methods of EREG and TGF ⁇ .
- a panning method using a phage vector can also be used.
- an antibody gene is obtained from a polyclonal antibody-expressing cell group as a library of heavy and light chain subclasses
- a screening method using a phage vector is advantageous.
- a gene encoding the variable region of the heavy chain and the light chain can be converted into a single chain Fv (scFv) by linking with an appropriate linker sequence. If a gene encoding scFv is inserted into a phage vector, a phage expressing scFv on the surface can be obtained.
- the DNA encoding scFv having the target binding activity can be recovered.
- scFv having the desired binding activity can be concentrated.
- the polynucleotide encoding the antibody may encode the full length of the antibody or may encode a part of the antibody.
- a part of an antibody refers to any part of an antibody molecule.
- an antibody fragment may be used as a term indicating a part of an antibody.
- a preferred antibody fragment in the present invention contains a complementarity determination region (CDR) of an antibody. More preferably, the antibody fragment of the present invention includes all three CDRs constituting the variable region.
- the cDNA is digested with a restriction enzyme that recognizes restriction enzyme sites inserted at both ends of the cDNA.
- a preferred restriction enzyme recognizes and digests a base sequence that is unlikely to appear in the base sequence constituting the antibody gene.
- a restriction enzyme that provides a sticky end is preferred.
- An antibody expression vector can be obtained by inserting the cDNA encoding the V region of the anti-EREG antibody digested as described above into an appropriate expression vector.
- a chimeric antibody can be obtained by fusing the gene encoding the antibody constant region (C region) and the gene encoding the V region in-frame.
- the term “chimeric antibody” means that the origins of the constant region and the variable region are different. Accordingly, in addition to a heterologous chimeric antibody such as mouse-human, a human-human homologous chimeric antibody is also included in the chimeric antibody of the present invention.
- a chimeric antibody expression vector can also be constructed by inserting the V region gene into an expression vector having a constant region in advance.
- a restriction enzyme recognition sequence for a restriction enzyme that digests the V region gene is placed on the 5 ′ side of an expression vector holding a DNA encoding a desired antibody constant region (C region).
- a chimeric antibody expression vector is constructed by digesting both with the same combination of restriction enzymes and fusing them in frame.
- the antibody gene can be incorporated into an expression vector so as to be expressed under the control of the expression control region.
- An expression control region for expressing an antibody includes, for example, an enhancer and a promoter.
- a recombinant host cell expressing the target antibody can be obtained by transforming an appropriate host cell with this expression vector.
- DNAs encoding antibody heavy chains (H chains) and light chains (L chains) can be incorporated into separate expression vectors.
- An antibody molecule having an H chain and an L chain can be expressed by co-transfecting a vector incorporating the H chain and the L chain into the same host cell at the same time.
- DNA encoding the H chain and L chain may be incorporated into a single expression vector to transform host cells (see International Publication WO94 / 11523).
- a host and an expression vector for producing an antibody by isolating an antibody gene once and introducing it into a suitable host are known. Any of these expression systems can be applied to the present invention.
- animal cells, plant cells, or fungal cells can be used. Specifically, the following cells can be exemplified as animal cells that can be used in the present invention.
- Mammalian cells CHO, COS, myeloma, BHK (baby hamster kidney), Hela, Vero, etc.
- Amphibian cells Xenopus oocytes, etc.
- Insect cells sf9, sf21, Tn5, etc.
- an antibody gene expression system using cells from the genus Nicotiana such as Nicotiana tabacum is known.
- Callus cultured cells can be used for transformation of plant cells.
- Yeast Saccharomyces genus such as Saccharomyces serevisiae, Pichia genus fungus such as methanol-utilizing yeast (Pichia pastoris): Aspergillus genus such as Aspergillus niger
- antibody gene expression systems using prokaryotic cells are also known.
- bacterial cells such as E. coli and Bacillus subtilis can be used in the present invention.
- an expression vector containing the target antibody gene is introduced by transformation.
- a desired antibody can be obtained from the culture of the transformed cells.
- transgenic animals can also be used for the production of recombinant antibodies. That is, the antibody can be obtained from an animal into which a gene encoding the target antibody has been introduced.
- an antibody gene can be constructed as a fusion gene by inserting in frame into a gene that encodes a protein that is uniquely produced in milk.
- a protein secreted into milk for example, goat ⁇ -casein can be used.
- the DNA fragment containing the fusion gene into which the antibody gene has been inserted is injected into a goat embryo, and the injected embryo is introduced into a female goat.
- the desired antibody can be obtained as a fusion protein with milk protein from milk produced by a transgenic goat (or its offspring) born from a goat that has received the embryo.
- hormones can be used as appropriate in transgenic goats to increase the amount of milk containing the desired antibody produced from the transgenic goat (Ebert, KM et al., Bio / Technology (1994) 12, 699-702). ).
- An animal antibody-derived C region can be used as the C region of the recombinant antibody of the present invention.
- C ⁇ 1, C ⁇ 2a, C ⁇ 2b, C ⁇ 3, C ⁇ , C ⁇ , C ⁇ 1, C ⁇ 2, C ⁇ can be used as the H chain C region of the mouse antibody
- C ⁇ , C ⁇ can be used as the L chain C region.
- animal antibodies such as rats, rabbits, goats, sheep, camels and monkeys can be used as animal antibodies other than mouse antibodies. These sequences are known.
- the C region can be modified to improve the stability of the antibody or its production.
- an antibody when administered to a human, it can be a genetically modified antibody that has been artificially modified for the purpose of reducing the heterologous antigenicity of the human.
- the recombinant antibody includes, for example, a chimeric antibody and a humanized antibody. These modified antibodies can be produced using known methods.
- Chimeric antibody refers to an antibody in which variable regions and constant regions derived from each other are linked.
- an antibody consisting of the variable regions of the heavy and light chains of a mouse antibody and the constant regions of the heavy and light chains of a human antibody is a mouse-human-heterologous chimeric antibody.
- a DNA encoding the variable region of a mouse antibody is linked to DNA encoding the constant region of a human antibody, and this is incorporated into an expression vector, whereby a recombinant vector that expresses the chimeric antibody can be prepared.
- the chimeric antibody produced during the culture can be obtained by culturing recombinant cells transformed with the vector and expressing the incorporated DNA.
- a human antibody is used for the C region of the chimeric antibody and the humanized antibody.
- C ⁇ 1, C ⁇ 2, C ⁇ 3, C ⁇ 4, C ⁇ , C ⁇ , C ⁇ 1, C ⁇ 2, and C ⁇ can be used as the C region.
- C ⁇ and C ⁇ can be used as the C region.
- the amino acid sequences of these C regions, as well as the base sequences encoding them, are known.
- the human antibody C region can be modified to improve the stability of the antibody itself or the production of the antibody.
- a chimeric antibody is composed of a V region of an antibody derived from a non-human animal and a C region derived from a human antibody.
- humanized antibodies include complementarity determining regions (CDRs) of non-human animal-derived antibodies, framework regions derived from human antibodies (FR; framework regions), and C regions derived from human antibodies. Consists of Since humanized antibodies have reduced antigenicity in the human body, they are useful as the active ingredient of the therapeutic agent of the present invention.
- Antibody variable regions are usually composed of three complementarity-determining regions (CDRs) sandwiched between four frames (FR).
- CDRs are regions that substantially determine the binding specificity of an antibody.
- the amino acid sequence of CDR is rich in diversity.
- the amino acid sequence constituting FR often shows high homology among antibodies having different binding specificities. Therefore, it is generally said that the binding specificity of a certain antibody can be transplanted to another antibody by CDR grafting.
- Humanized antibodies are also referred to as reshaped human antibodies. Specifically, non-human animals, for example, humanized antibodies obtained by grafting mouse antibody CDRs to human antibodies are known. General genetic recombination techniques for obtaining humanized antibodies are also known.
- overlap extension PCR is known as a method for transplanting mouse antibody CDRs to human FRs.
- PCR extension the base sequence which codes CDR of the mouse antibody which should be transplanted is added to the primer for synthesize
- the base sequences to be linked are designed to be connected to each other in frame.
- Human FRs are synthesized individually by each primer.
- a product in which DNA encoding mouse CDR is added to each FR is obtained.
- the base sequences encoding mouse CDRs of each product are designed to overlap each other.
- the overlapping CDR portions of the products synthesized using the human antibody gene as a template are annealed with each other to perform a complementary chain synthesis reaction. By this reaction, human FRs are linked via the mouse CDR sequence.
- a human-type antibody expression vector can be prepared by inserting the DNA obtained as described above and a DNA encoding the human antibody C region into an expression vector so as to be fused in frame. After introducing the integration vector into a host to establish a recombinant cell, the recombinant cell is cultured, and a DNA encoding the humanized antibody is expressed, whereby the humanized antibody is cultured in the cultured cell. (See European Patent Publication EP 239,400, International Publication WO 96/02576).
- the CDR forms a favorable antigen-binding site when linked via CDR.
- a human antibody FR can be suitably selected.
- FR amino acid residues can be substituted so that the CDR of the reshaped human antibody forms an appropriate antigen-binding site.
- amino acid sequence mutations can be introduced into FRs by applying the PCR method used for transplantation of mouse CDRs into human FRs.
- partial nucleotide sequence mutations can be introduced into primers that anneal to the FR.
- a nucleotide sequence mutation is introduced into the FR synthesized by such a primer.
- a mutant FR sequence having a desired property can be selected by measuring and evaluating the antigen-binding activity of a mutant antibody substituted with an amino acid by the above method (Sato, K.et al., Cancer Res, 1993, 53 , 851-856).
- a method for obtaining a human antibody is also known.
- human lymphocytes are sensitized in vitro with the desired antigen or cells expressing the desired antigen.
- a desired human antibody having an antigen-binding activity can be obtained by fusing sensitized lymphocytes with human myeloma cells (see Japanese Patent Publication No. 1-59878).
- U266 can be used as a human myeloma cell as a fusion partner.
- a desired human antibody can be obtained by immunizing a transgenic animal having all repertoires of human antibody genes with a desired antigen (International Publication WO 93/12227, WO 92/03918, WO 94/02602, WO 94). / 25585, WO 96/34096, WO 96/33735).
- a technique for obtaining a human antibody by panning using a human antibody library is also known.
- the V region of a human antibody is expressed as a single chain antibody (scFv) on the surface of the phage by the phage display method, and a phage that binds to the antigen can be selected.
- the DNA sequence encoding the V region of the human antibody that binds to the antigen can be determined. After determining the DNA sequence of scFv that binds to the antigen, the V region sequence is fused in-frame with the sequence of the desired human antibody C region, and then inserted into an appropriate expression vector, whereby an expression vector can be prepared.
- the human antibody can be obtained by introducing the expression vector into a suitable expression cell as described above and expressing the gene encoding the human antibody. These methods are already known (International Publication WO 92/01047, WO 93/20791, WO 93/06213, WO 93/11236, WO 93/19172, WO 95/01438, WO 95/15388).
- the antibodies used in the present invention include not only bivalent antibodies typified by IgG but also monovalent antibodies or multivalent antibodies typified by IgM as long as they bind to the target protein.
- the multivalent antibodies of the present invention include multivalent antibodies that all have the same antigen-binding site, or multivalent antibodies that have some or all different antigen-binding sites.
- the antibody used in the present invention is not limited to a full-length antibody molecule, and may be a low molecular weight antibody or a modified product thereof as long as it binds to a target protein.
- the low molecular weight antibody includes an antibody fragment in which a part of a full-length antibody (whole antibody, such as whole IgG) is deleted. As long as it has the ability to bind to the antigen of interest, partial deletion of the antibody molecule is acceptable.
- the antibody fragment in the present invention preferably contains either or both of a heavy chain variable region (VH) and a light chain variable region (VL).
- VH heavy chain variable region
- VL light chain variable region
- the amino acid sequence of VH or VL can include substitutions, deletions, additions and / or insertions.
- VH and VL can be deleted.
- the variable region may be chimerized or humanized.
- antibody fragments include, for example, Fab, Fab ′, F (ab ′) 2, and Fv.
- Specific examples of the low molecular weight antibody include, for example, Fab, Fab ′, F (ab ′) 2, Fv, scFv (single chain Fv), Diabody, sc (Fv) 2 (single chain (Fv) 2) And so on. Multimers of these antibodies (eg, dimer, trimer, tetramer, polymer) are also included in the low molecular weight antibody of the present invention.
- Antibody fragments can be obtained by treating antibodies with enzymes to generate antibody fragments.
- enzymes that produce antibody fragments include, for example, papain, pepsin, and plasmin.
- genes encoding these antibody fragments can be constructed, introduced into an expression vector, and then expressed in an appropriate host cell (for example, Co, MS et al., J. Immunol. (1994) 152). , 2968-2976, Better, M. and Horwitz, A. H. Methods in Enzymology (1989) 178, 476-496, Plueckthun, A. and Skerra, A.
- a digestive enzyme cleaves an antibody at a specific position to give an antibody fragment having a specific structure as follows. Papain digestion: F (ab) 2 or Fab Pepsin digestion: F (ab ') 2 or Fab' Plasmin digestion: Facb With respect to the antibody fragment obtained enzymatically as described above, any part of the antibody can be deleted by a method using a genetic engineering technique.
- the low molecular weight antibody in the present invention can be an antibody fragment lacking any region as long as it has binding affinity for the target protein.
- the antibody may maintain its effector activity. That is, the low molecular weight antibody of the present invention may be an antibody having both a binding affinity for the target protein and an effector function. Antibody effector functions include ADCC activity and CDC activity. The therapeutic antibody in the present invention may have either or both of ADCC activity and CDC activity as an effector function.
- Diabody refers to a bivalent low molecular weight antibody constructed by gene fusion (HolligerolliP et al.,. Proc.Natl.Acad.Sci.USA 90: 6444-6448 (1993), EP404,097) , WO 93/11161 etc.).
- Diabody is a dimer composed of two polypeptide chains. Usually, in the polypeptide chain constituting the dimer, VL and VH are connected by a linker in the same chain.
- the linker that links the polypeptide chains in Diabody is generally short enough to prevent VL and VH from binding to each other.
- the amino acid residues constituting the linker are preferably 2 to 12 residues, more preferably 3 to 10 residues, and particularly about 5 residues. Therefore, VL and VH encoded on the same polypeptide chain cannot form a single chain variable region fragment but form a dimer with another single chain variable region fragment. As a result, Diabody has two antigen-binding sites.
- ScFv can be obtained by linking antibody H chain V region and L chain V region.
- the H chain V region and the L chain V region are linked via a linker, preferably a peptide linker (Huston, J. S. et al., Proc. Natl. Acad. Sci. USA, 1988, 85). , 5879-5883.).
- the H chain V region and L chain V region in scFv may be derived from any antibody described as an antibody in the present specification.
- region For example, any single chain peptide consisting of about 3 to 25 residues can be used as a linker. Specifically, for example, a peptide linker described later can be used.
- V regions can be ligated by, for example, the PCR method as described above.
- the DNA encoding the desired partial amino acid sequence is used as a template.
- DNA sequence encoding H chain or H chain V region of the antibody, and DNA sequence encoding L chain or L chain V region of the antibody is used as a template.
- the DNAs encoding the H region and L chain V regions are each amplified by PCR using a pair of primers having sequences corresponding to the sequences at both ends of the DNA to be amplified.
- DNA encoding a peptide linker portion is prepared.
- DNA encoding a peptide linker can also be synthesized using PCR.
- a base sequence that can be linked to the amplification product of each V region synthesized separately is added to the 5 ′ side of the primer to be used.
- PCR reaction is performed using each DNA of [H chain V region DNA]-[peptide linker DNA]-[L chain V region DNA] and assembly PCR primers.
- the primer for assembly PCR consists of a combination of a primer that anneals to the 5 ′ side of [H chain V region DNA] and a primer that anneals to the 3 ′ side of [L chain V region DNA]. That is, the assembly PCR primer is a primer set that can amplify DNA encoding the full-length sequence of scFv to be synthesized. On the other hand, a base sequence that can be linked to each V region DNA is added to [peptide linker DNA]. As a result, these DNAs are ligated, and the full length of scFv is finally produced as an amplification product by the primers for assembly PCR.
- an expression vector containing them and a recombinant cell transformed with the expression vector can be obtained according to a conventional method. Further, the scFv can be obtained by culturing the resulting recombinant cells and expressing the DNA encoding the scFv.
- Sc (Fv) 2 is a low molecular weight antibody in which two VHs and two VLs are combined with a linker or the like to form a single chain (Hudson et al., J Immunol. Methods Methods 1999; 231: 177-189).
- sc (Fv) 2 can be prepared, for example, by linking scFv with a linker.
- VHs and two VLs are arranged in the order of VH, VL, VH, and VL ([VH] linker [VL] linker [VH] linker [VL]) starting from the N-terminal side of the single-chain polypeptide.
- An antibody characterized in that it is preferred.
- a peptide linker is preferred.
- the length of the peptide linker is not particularly limited, and can be appropriately selected by those skilled in the art according to the purpose.
- the amino acid residues constituting the peptide linker are 1 to 100 amino acids, preferably 3 to 50 amino acids, more preferably 5 to 30 amino acids, particularly preferably 12 to 18 amino acids (for example, 15 amino acids).
- the amino acid sequence constituting the peptide linker can be any sequence as long as it does not inhibit the scFv binding action.
- the following amino acid sequence can be used.
- n for determining the length of the peptide linker is usually 1 to 5, preferably 1 to 3, more preferably 1 or 2.
- sc (Fv) 2 examples include the following sc (Fv) 2.
- V regions can be linked using a synthetic chemical linker (chemical cross-linking agent).
- a crosslinking agent usually used for crosslinking such as peptide compounds can be used in the present invention.
- the following chemical crosslinking agents are known. These crosslinking agents are commercially available.
- N-hydroxysuccinimide (NHS), Disuccinimidyl suberate (DSS), Bis (sulfosuccinimidyl) suberate (BS3), Dithiobis (succinimidyl propionate) (DSP), Dithiobis (sulfosuccinimidyl propionate) (DTSSP), Ethylene glycol bis (succinimidyl succinate) (EGS), Ethylene glycol bis (sulfosuccinimidyl succinate) (sulfo-EGS), Disuccinimidyl tartrate (DST), disulfosuccinimidyl tartrate (sulfo-DST), Bis [2- (succinimideoxycarbonyloxy) ethyl] sulfone (BSOCOES), bis [2- (sulfosuccinimideoxycarbonyloxy) ethyl] sulfone (sulfo-BSOCOES), etc
- the preferred low molecular weight antibody in the present invention is Diabody or sc (Fv) 2.
- the antibody is treated with an enzyme such as papain or pepsin to generate antibody fragments, or DNA encoding these antibody fragments is constructed and used as an expression vector. After introduction, expression in an appropriate host cell (for example, Co, M. S. et al., J. Immunol. (1994) 152, 2968-2976; Better, M. and Horwitz, A. H Methods Enzymol. (1989) 178, 476-496; Pluckthun, A.
- An antibody conjugated with various molecules such as polyethylene glycol (PEG) can also be used as a modified antibody. It is also possible to bind a chemotherapeutic agent, a toxic peptide or a radioactive chemical substance to the antibody.
- a modified antibody hereinafter referred to as antibody conjugate
- Such a modified antibody can be obtained by chemically modifying the obtained antibody.
- the modification method of an antibody has already been established in this field.
- bispecific antibodies designed using genetic recombination technology to recognize not only the target protein but also chemotherapeutic agents, toxic peptides or radioactive chemicals It can also be obtained as a molecular type. These antibodies are also included in the “antibody” in the present invention.
- a preferable chemotherapeutic agent is a low-molecular chemotherapeutic agent.
- Small molecule chemotherapeutic agents are less likely to interfere with antibody function after binding to the antibody.
- the low molecular weight chemotherapeutic agent usually has a molecular weight of 100 to 2000, preferably 200 to 1000.
- the chemotherapeutic agents exemplified here are all low-molecular chemotherapeutic agents.
- These chemotherapeutic agents in the present invention include a prodrug that is converted into an active chemotherapeutic agent in vivo. Activation of the prodrug may be enzymatic conversion or non-enzymatic conversion.
- ricin abrin, ribonuclease, onconase, DNase I, Staphylococcal enterotoxin-A, pokeweed antiviral protein, gelonin, diphtheria toxin, Pseudomonas exotoxin, Pseudomonas endotoxin, L-asparaginase, PEG L-Asparaginase You can also.
- one or more small molecule chemotherapeutic agents and toxic peptides can be used in combination to modify the antibody. Coupling between the anti-EREG antibody and the above-described small molecule chemotherapeutic agent can be covalent or non-covalent. Methods for producing antibodies bound with these chemotherapeutic agents are known.
- proteinaceous drugs and toxins can be bound to antibodies by genetic engineering techniques.
- a recombinant vector in which a DNA encoding the toxic peptide and a DNA encoding an anti-EREG antibody are fused in frame and incorporated into an expression vector can be constructed.
- a transformed cell obtained by introducing the vector into an appropriate host cell is cultured, and the incorporated DNA is expressed, whereby an anti-EREG antibody bound with a toxic peptide can be obtained as a fusion protein.
- a proteinaceous drug or toxin is generally arranged on the C-terminal side of the antibody.
- a peptide linker can be interposed between the antibody and the proteinaceous drug or toxin.
- bispecific antibodies refer to antibodies that have variable regions that recognize different epitopes within the same antibody molecule.
- bispecific antibody of the present invention include bispecific antibodies in which one antigen binding site recognizes EREG and the other antigen binding site recognizes TGF ⁇ .
- bispecific antibodies can be produced by combining two types of antibodies with different recognition antigens.
- the antibody to be bound may be a 1 ⁇ 2 molecule each having an H chain and an L chain, or may be a 1 ⁇ 4 molecule consisting only of an H chain.
- bispecific antibody-producing fused cells can be prepared by fusing hybridomas that produce different monoclonal antibodies.
- bispecific antibodies can be produced by genetic engineering techniques.
- the antibody of the present invention can be produced by methods known to those skilled in the art as described below.
- it in the case of mammalian cells, it can be expressed by functionally binding a useful promoter commonly used, an antibody gene to be expressed, and a poly A signal downstream of the 3 ′ side thereof.
- the promoter / enhancer can include human cytomegalovirus immediate early promoter / enhancer.
- a promoter / enhancer derived from a mammalian cell such as a viral promoter / enhancer or human elongation factor 1 ⁇ (HEF1 ⁇ ) can be used for antibody expression.
- viruses that can utilize promoters / enhancers include retroviruses, polyomaviruses, adenoviruses, and simian virus 40 (SV40).
- the method of Mulligan et al. (Nature (1979) 277, 108) can be used. Further, the HEF1 ⁇ promoter / enhancer can be easily used for target gene expression by the method of Mizushima et al. (Nucleic Acids Res. (1990) 18, 5322).
- the gene can be expressed by functionally combining a useful promoter commonly used, a signal sequence for antibody secretion, and an antibody gene to be expressed.
- the promoter include lacZ promoter and araB promoter.
- the lacZ promoter the method of Ward et al. (Nature (1989) 341, 544-546; FASEBJ. (1992) 6, 2422-2427) can be used.
- the araB promoter can be used for the expression of the target gene by the method of Better et al. (Science (1988) 240, 1041-1043).
- a pelB signal sequence (Lei, S. P. et al., J. Bacteriol. (1987) 169, 4379) may be used when the periplasm of E. coli is produced. If necessary, the antibody structure is refolded to have a desired binding activity by using a protein denaturant such as guanidine hydrochloride of urea.
- a selectable marker can be inserted into the expression vector for amplification of the gene copy number in the host cell system. Specifically, the following selection markers can be used. Aminoglycoside transferase (APH) gene, Thymidine kinase (TK) gene, E. coli xanthine guanine phosphoribosyltransferase (Ecogpt) gene, Dihydrofolate reductase (dhfr) gene, etc.
- APH Aminoglycoside transferase
- TK Thymidine kinase
- Ecogpt E. coli xanthine guanine phosphoribosyltransferase
- dhfr Dihydrofolate reductase
- a eukaryotic cell or a prokaryotic cell system can be used for the production of the antibody used in the present invention.
- eukaryotic cells include established mammalian cell lines, insect cell lines, fungal cells, and animal cells such as yeast cells.
- prokaryotic cells include bacterial cells such as E. coli cells.
- the antibodies used in the present invention are expressed using mammalian cells.
- mammalian cells that can be used include CHO, COS, myeloma, BHK, Vero, and Hela cells.
- the transformed host cell is cultured in vitro or in vivo to produce the desired antibody.
- Host cells are cultured according to a known method.
- DMEM, MEM, RPMI1640, and IMDM can be used as the culture medium
- serum supplements such as fetal calf serum (FCS) can be used in combination.
- the antibody expressed and produced as described above can be purified by using a known method used in normal protein purification alone or in combination as appropriate.
- antibodies can be separated and purified by appropriately selecting and combining affinity columns such as protein A columns, chromatography columns such as ions and gel filtration, filters, ultrafiltration, salting out, dialysis, etc. (Antibodies A Laboratory Manual. Ed Harlow, David Lane, Cold Spring Harbor Laboratory, 1988).
- ELISA enzyme-linked immunosorbent assay
- EIA enzyme immunoassay
- RIA radioimmunoassay
- fluorescent immunoassay can be used.
- the antibody used in the present invention may be an antibody having a modified sugar chain. It is known that the cytotoxic activity of an antibody can be enhanced by modifying the sugar chain of the antibody.
- the following antibodies are known as antibodies with modified sugar chains.
- Antibodies with modified glycosylation eg WO99 / 54342
- Antibodies lacking fucose added to the sugar chain WO00 / 61739, WO02 / 31140, etc.
- Antibodies with sugar chains with bisecting GlcNAc WO02 / 79255 etc.
- the antibody used in the present invention may have cytotoxic activity.
- cytotoxic activity in the present invention include antibody-dependent cell-mediated cytotoxicity (ADCC) activity and complement-dependent cytotoxicity (CDC) activity.
- ADCC activity means cytotoxic activity by the complement system.
- ADCC activity means that when a specific antibody is attached to the cell surface antigen of the target cell, Fc ⁇ receptor-bearing cells (immune cells, etc.) bind to the Fc portion via the Fc ⁇ receptor, causing damage to the target cell. Means activity.
- Whether or not the anti-EREG antibody has ADCC activity or CDC activity can be measured by a known method (for example, Current protocols in Immunology, Chapter7. Immunologic studies in humans, Editor, John E, Coligan et al., John Wiley & Sons, Inc., (1993)).
- effector cells Specifically, first, effector cells, complement solution, and target cells are prepared.
- (1) Preparation of effector cells Spleens are removed from CBA / N mice and the like, and spleen cells are isolated in RPMI1640 medium (Invitrogen). After washing with the same medium containing 10% fetal calf serum (FBS, HyClone), effector cells can be prepared by adjusting the cell concentration to 5 ⁇ 10 6 / ml.
- FBS fetal calf serum
- Target cells Cells expressing the target protein are cultured for 1 hour at 37 ° C in 10% FBS-containing DMEM medium with 0.2 mCi of 51 Cr-sodium chromate (GE Healthcare Japan).
- the target cell can be radiolabeled.
- Cells expressing the target protein include cells transformed with a gene encoding the target protein, primary colorectal cancer, metastatic colorectal cancer, lung adenocarcinoma, pancreatic cancer, gastric cancer, renal cancer cell, colorectal cancer cell, Esophageal cancer cells, gastric cancer cells, pancreatic cancer cells and the like can be used.
- the target cells can be prepared by washing the cells three times with 10% FBS-containing RPMI1640 medium and adjusting the cell concentration to 2 ⁇ 10 5 / ml.
- ADCC activity or CDC activity can be measured by the method described below.
- 50 ⁇ l each of target cells and anti-EREG antibody are added to a 96-well U-bottom plate (BectonBDickinson) and allowed to react on ice for 15 minutes. Thereafter, 100 ⁇ l of effector cells are added and cultured for 4 hours in a carbon dioxide incubator. The final antibody concentration is 0 or 10 ⁇ g / ml. After incubation, 100 ⁇ l of the supernatant is collected, and the radioactivity is measured with a gamma counter (COBRAIIAAUTO-GAMMA, MODEL D5005, Packard Instruments Company).
- COBRAIIAAUTO-GAMMA MODEL D5005, Packard Instruments Company
- Cytotoxic activity (%) can be calculated based on the formula (A-C) / (B-C) x 100 using the obtained value.
- A is the radioactivity (cpm) in each sample
- B is the radioactivity (cpm) in the sample with 1% NP-40 (manufactured by nacalai tesque)
- C is the radioactivity (cpm) of the sample containing only the target cells Show.
- antagonist activity against EREG and / or antagonist activity against TGF ⁇ can be measured by methods known to those skilled in the art.
- a dimer is formed by binding of a ligand, and tyrosine kinase that is its own domain existing in the cell is activated.
- Activated tyrosine kinases form peptides containing phosphorylated tyrosines by autophosphorylation and associate them with various signaling accessory molecules. They are mainly PLC ⁇ (phospholipase C ⁇ ), Shc, Grb2, and the like. Of these accessory molecules, the former two are further phosphorylated by tyrosine kinases of the EGF receptor.
- the main pathway in signal transduction from the EGF receptor is a pathway through which phosphorylation is transmitted in the order of Shc, Grb2, Sos, Ras, Raf / MAPK kinase / MAP kinase. Furthermore, it is considered that there is a route from PLC ⁇ to PKC, which is a sub route.
- target molecules can be appropriately set for each target cell, and are not limited to the above factors.
- a commercially available kit for measuring in vivo signal activation can be used as appropriate (for example, a protein kinase C activity measurement system (GE Healthcare Japan Co., Ltd.)).
- activation of in vivo signals can also be detected using the transcription inducing action on a target gene present downstream of the in vivo signal cascade as an index.
- Changes in transcriptional activity can be detected by reporter assay principles. Specifically, a reporter gene such as GFP (Green Fluorescence Protein) or luciferase is placed downstream of the transcription factor or promoter region of the target gene, and the change in the transcription activity is measured as the reporter activity by measuring the reporter activity. can do.
- GFP Green Fluorescence Protein
- the EGF receptor usually works in the direction of promoting cell proliferation
- the activation of in vivo signaling can be evaluated by measuring the proliferation activity of the target cells.
- the antagonist activity of the test substance is evaluated by evaluating the latter cell proliferation activity, but the present invention is not limited to this method, and the above-mentioned methods are preferably employed for each selected target cell. Can be evaluated.
- the antagonist activity of a test substance can be evaluated or measured by measuring the following cell proliferation activity.
- a method is used in which the uptake of [ 3 H] -labeled thymidine added to the medium by living cells is measured as an indicator of DNA replication ability.
- a dye exclusion method in which the ability to remove a dye such as trypan blue outside the cell is measured under a microscope, or an MTT method is used.
- the latter uses the ability of living cells to convert the tetrazolium salt MTT (3- (4,5-dimethylthiazol-2-yl) -2,5-diphenyl tetrazolium bromide) into a blue formazan product.
- the test antibody is added to the culture solution of the test cells, and after a predetermined time has elapsed, the MTT solution is added to the culture solution and allowed to stand for a fixed time, thereby allowing MTT to be taken into the cells.
- MTT which is a yellow compound
- succinate dehydrogenase in the mitochondria in the cell.
- the blue product is dissolved and colored, and the absorbance is measured to obtain an index of the number of viable cells.
- reagents such as MTS, XTT, WST-1, and WST-8 are also commercially available (eg, nacalaitesque) and can be suitably used.
- a method for evaluating cell proliferation activity using cell ATP or cell culture impedance as an index is also known.
- the activity for example, when the test substance is an antibody, an antibody having the same isotype as the target antibody as a control antibody and not having the antagonist activity (for example, binding to a cell expressing the target antigen)
- the antibody can be used in the same manner as the antibody of interest, and the activity of the antibody can be determined by showing stronger antagonist activity than the control antibody.
- the cell in which the antagonist suppresses proliferation is not particularly limited as long as it is a cell expressing EREG protein, TGF ⁇ protein, and / or EGFR.
- Preferred cells are for example cancer cells.
- cells derived from colon cancer, lung adenocarcinoma, pancreatic cancer, gastric cancer, and renal cancer are suitable as the cells in the present invention. According to the present invention, it is possible to obtain an effective cell growth inhibitory effect on both primary lesions and metastatic lesions of these cancers.
- Further preferred cancer cells are primary colorectal cancer, metastatic colorectal cancer, lung adenocarcinoma, pancreatic cancer, gastric cancer, and renal cancer.
- the antibody of the present invention can be used for the purpose of treating or preventing diseases caused by cell proliferation, such as colon cancer, lung adenocarcinoma, pancreatic cancer, gastric cancer, and renal cancer. These cancers can be treated or prevented regardless of whether they are primary or metastatic. More preferably, the antibody of the present invention can be used for the purpose of treating and / or preventing primary colorectal cancer, metastatic colorectal cancer, and pancreatic cancer. Furthermore, among these cancers, cancers that proliferate in an EREG and / or TGF ⁇ -dependent manner are preferred as targets for treatment and / or prevention in the present invention.
- the present invention also provides a polynucleotide encoding the antibody of the present invention, or a polynucleotide encoding an antibody that hybridizes with the polynucleotide under stringent conditions and has an activity equivalent to that of the antibody of the present invention. .
- the present invention also provides vectors containing these polynucleotides and transformants (including transformed cells) containing the vectors.
- the polynucleotide of the present invention is not particularly limited as long as it encodes the antibody of the present invention, and is a polymer comprising a plurality of bases or base pairs such as deoxyribonucleic acid (DNA) or ribonucleic acid (RNA).
- the polynucleotide of the present invention may contain a non-natural base.
- the polynucleotide of the present invention can be used when an antibody is expressed by a genetic engineering technique. It can also be used as a probe when screening an antibody having a function equivalent to that of the antibody of the present invention. That is, a polynucleotide encoding the antibody of the present invention, or a part thereof, is used as a probe, hybridized with the polynucleotide under stringent conditions by a technique such as hybridization, gene amplification technique (eg, PCR), and the like, DNA encoding an antibody having an activity equivalent to that of the antibody of the present invention can be obtained. Such DNA is also included in the polynucleotide of the present invention. Hybridization techniques (Sambrook, Jet et al., Molecular Cloning 2nd edition, 9.47-9.58, Cold Spring Harbor Lab. Press, 1989) are techniques well known to those skilled in the art.
- hybridization conditions include low stringency washing conditions.
- Low stringent conditions are, for example, room temperature, 2 ⁇ SSC, and 0.1% SDS, and preferably 42 ° C., 0.1 ⁇ SSC, and 0.1% SDS in washing after hybridization.
- More preferable hybridization conditions include highly stringent washing conditions.
- Highly stringent conditions are, for example, conditions of 65 ° C., 0.5 ⁇ SSC and 0.1% SDS. Under these conditions, it can be expected that a polynucleotide having high homology can be efficiently obtained as the temperature is increased.
- a plurality of factors such as temperature and salt concentration can be considered as factors affecting the stringency of hybridization, and those skilled in the art can realize the same stringency by appropriately selecting these factors. .
- Antibodies that are functionally equivalent to the antibodies of the present invention encoded by polynucleotides obtained by these hybridization techniques and gene amplification techniques usually have high homology in amino acid sequences with these antibodies.
- the antibody of the present invention includes an antibody functionally equivalent to the antibody of the present invention and having high homology with the amino acid sequence of the antibody.
- High homology generally refers to at least 50% identity, preferably 75% identity, more preferably 85% identity, more preferably 95% identity at the amino acid level. .
- Polypeptide homology was determined using the algorithm described in the literature (Wilbur, W. J. and Lipman, D. J. Proc. Natl. Acad. Sci. USA (1983) 80, 726-730). Just do it.
- a substance that reduces the expression level of EREG or TGF ⁇ can be obtained by methods known to those skilled in the art.
- SiRNA is double-stranded RNA in which RNA complementary to the transcription product of the target gene and RNA complementary to the RNA are bound.
- the length of siRNA is not specifically limited, It is preferable that it is the short chain which does not show toxicity in a cell.
- the length of siRNA is usually 15-50 base pairs, preferably 15-30 base pairs.
- the duplex part of siRNA is not limited to those completely paired, but includes mismatched parts due to mismatches (corresponding bases are not complementary), bulges (no bases corresponding to one strand), etc. It may be.
- the end structure of siRNA may be either a blunt end or a sticky (protruding) end.
- Antisense oligonucleotide includes antisense RNA and antisense DNA.
- Antisense oligonucleotides are RNA or DNA complementary to the transcription product of a target gene, and are usually single-stranded.
- the length of the antisense oligonucleotide is not particularly limited, but is usually 5 to 50 bases, preferably 9 to 30 bases.
- the antisense oligonucleotide does not need to be completely complementary to the transcription product of the target gene, and may contain an unpaired portion due to mismatch or bulge.
- One embodiment of the antisense RNA is microRNA (miR).
- a microRNA is a single-stranded RNA complementary to a transcript of a target gene. In general, microRNA is a small non-coding RNA that acts on mRNA to cause gene silencing.
- the length of the microRNA is not particularly limited, but is usually 10 to 30 bases, preferably 14 to 21 bases.
- the present invention provides a pharmaceutical composition containing the antibody of the present invention as an active ingredient.
- the present invention also relates to a cell growth inhibitor, particularly an anticancer agent, containing the antibody of the present invention as an active ingredient.
- the present invention provides a pharmaceutical composition containing an EREG antagonist and a TGF ⁇ antagonist as active ingredients.
- the present invention also relates to a cell growth inhibitor, particularly an anticancer agent, containing an EREG antagonist and a TGF ⁇ antagonist as active ingredients.
- the cell growth inhibitor and anticancer agent of the present invention are preferably administered to a subject suffering from or possibly suffering from cancer.
- the cancer type targeted by the anticancer agent of the present invention is not particularly limited.
- the anticancer agent of the present invention can be used for cancers such as colorectal cancer, lung adenocarcinoma, pancreatic cancer, gastric cancer, and renal cancer.
- the anticancer agent of the present invention can be used for both primary lesions and metastatic lesions.
- “containing a certain substance as an active ingredient” means that the substance is included as a main active ingredient, and does not limit the content of the substance.
- the pharmaceutical composition of the present invention can be administered to a patient by either oral or parenteral administration. Preferably, it is parenteral administration. Specific examples of such administration methods include injection administration, nasal administration, transpulmonary administration, and transdermal administration.
- injection administration the pharmaceutical composition of the present invention can be administered systemically or locally by, for example, intravenous injection, intramuscular injection, intraperitoneal injection, subcutaneous injection and the like.
- the administration method can be appropriately selected depending on the age and symptoms of the patient.
- the dose for example, the dose can be selected in the range of 0.0001 mg to 1000 mg per kg of body weight per administration. Alternatively, for example, the dose can be selected in the range of 0.001 to 100,000 mg / body per patient.
- the pharmaceutical composition of the present invention is not limited to these doses.
- a method for inhibiting cell proliferation or a method for treating cancer which comprises the step of administering the pharmaceutical composition of the present invention to a subject (eg, a mammal such as a human) is also included in the present invention.
- the EREG antagonist and TGF ⁇ antagonist may be administered simultaneously or at different timings. Moreover, the EREG antagonist and the TGF ⁇ antagonist may be one preparation or different preparations.
- the pharmaceutical composition of the present invention can be formulated in accordance with a conventional method (for example, Remington's Pharmaceutical, Science, Latest Edition, Mark Publishing, Company, Easton, USA) together with pharmaceutically acceptable carriers and additives. It may be.
- a conventional method for example, Remington's Pharmaceutical, Science, Latest Edition, Mark Publishing, Company, Easton, USA
- pharmaceutically acceptable carriers and additives may be.
- it is not limited to these, and other commonly used carriers can be used as appropriate.
- silicic acid lactose, crystalline cellulose, mannitol, starch, carmellose calcium, carmellose sodium, hydroxypropylcellulose, hydroxypropylmethylcellulose, polyvinylacetal diethylaminoacetate, polyvinylpyrrolidone, gelatin, medium chain fatty acid triglyceride
- the carrier include polyoxyethylene hydrogenated castor oil 60, sucrose, carboxymethylcellulose, corn starch, and inorganic salts.
- the present invention provides a method for screening a candidate compound for a therapeutic agent for cancer, comprising the following steps. (1) a step of measuring EREG antagonist activity of a test substance, (2) measuring the TGF ⁇ antagonist activity of the test substance, and (3) A step of selecting a substance having EREG antagonist activity and TGF ⁇ antagonist activity.
- the EREG antagonist activity and the TGF ⁇ antagonist activity can be measured by methods known to those skilled in the art as described above.
- the candidate compound selected by the screening method of the present invention is useful as a candidate compound for a therapeutic agent for an anticancer drug having an action mechanism that suppresses the action of EREG and TGF ⁇ .
- the candidate compound selected by the screening method of the present invention is further evaluated for effects on other cancer cell lines or primary cultured cells, and toxicity on normal cells, if necessary. Through these evaluations, compounds useful as cancer therapeutics can be selected. A series of evaluation methods as cancer therapeutic agents has already been established.
- any natural or artificially synthesized compound can be used as a test compound.
- a protein library or an antibody library is preferable as the library of test compounds in the present invention.
- a phage library displaying proteins and antibodies can be used as a test compound.
- a library of artificially synthesized compounds such as a combinatorial library can be used as a test compound.
- the EREG antagonist activity and the TGF ⁇ antagonist activity can be measured by methods known to those skilled in the art, such as the method described above and the method of the present example.
- this invention provides the manufacturing method of the therapeutic agent of cancer including the following processes. (1) selecting a substance having EREG antagonist activity; (2) selecting a substance having TGF ⁇ antagonist activity; (3) A step of combining the substance selected in (1) and the substance selected in (2) into an anticancer agent.
- the substance having EREG antagonist activity is not particularly limited, and a known substance that is already known to have EREG antagonist activity may be used, or newly obtained using the above-described measurement of antagonist activity as an index It may be a substance.
- the substance having TGF ⁇ antagonist activity is not particularly limited, and a known substance that is already known to have TGF ⁇ antagonist activity may be used, or newly obtained using the above-mentioned measurement of antagonist activity as an index. It may be a substance.
- the mode is not limited as long as the substance having EREG antagonist activity and the substance having TGF ⁇ antagonist activity are administered to the same subject.
- the substance having EREG antagonist activity and the TGF ⁇ antagonist activity may be administered as one drug, or may be administered as different drugs.
- the substance having EREG antagonist activity and the substance having TGF ⁇ antagonist activity may be administered simultaneously or at different times.
- Ba / F3 cell line expressing EGFR-GCSFR chimeric receptor grows depending on Epiregulin Isolation by PCR amplification of human EGFR (GenBank: NM_005228) and mouse GCSFR (GenBank: NM_007782) cDNA did.
- a chimeric receptor gene consisting of EGFR extracellular domain and GCSFR intracellular domain was prepared.
- the chimeric receptor base sequence is shown in SEQ ID NO: 1
- the amino acid sequence is shown in SEQ ID NO: 2.
- a mammalian expression vector for the chimeric receptor was constructed. In the expression vector, the chimeric receptor is designed to be transcribed under the human EF1 ⁇ promoter.
- the chimeric receptor expression vector has a geneticin resistance gene.
- Ba / F3 cells were transformed with the linearized chimeric receptor expression vector.
- Cells into which the vector was introduced were selected in the presence of geneticin.
- the selected cells were maintained by adding EGF instead of IL3 in the medium. The purpose is to retain the properties of proliferating cells depending on the chimeric receptor signal.
- a luciferase expression vector was introduced into the chimeric receptor-expressing cells. The purpose is to allow cell proliferation to be detected as luciferase activity.
- a luciferase gene (pGL3-Control Vector, Promega) was incorporated into a mammalian cell expression vector having a zeocin resistance marker.
- luciferase is designed to be transcribed under the mouse CMV promoter. Cells were transformed with a linearized luciferase expression vector. After selecting cells in the presence of zeocin and geneticin, cells with high luciferase activity were selected.
- EGF Ba / F3 cells expressing a chimeric receptor, luciferase
- EGF was maintained in RPMI1640 medium containing 10% fetal bovine serum and EGF, geneticin and zeocin.
- EGF (R) proliferated depending on the addition of human Epiregulin (EREG).
- EGF (R) was centrifuged and resuspended in RPMI1640 + 10% FBS medium. Cells were seeded into 96-well plates at 1 ⁇ 10 4 / well. Human EREG (R & D Systems) was added to the culture solution to achieve the final concentration of interest and incubated at 37 ° C. under 5% CO 2 for 4 days. The number of cells after incubation was measured based on the following two enzyme activities. For measurement of dehydrogenase activity, WST-8 reagent Cell Count Reagent SF (Nacalai Tesque) was used. The method followed the protocol specified by the manufacturer.
- Example 2 Cells expressing EREG activate the EGFR chimeric receptor on the cell surface.
- the EREG cDNA (NM_001432) represented by SEQ ID NO: 3 was cloned into the mammalian expression vector pMCN.
- pMCN can express a foreign gene under the mouse CMV promoter (GenBank: U68299).
- the pMCN vector has a geneticin resistance gene.
- CHO cell strain DG44 Invitrogen
- a cell line EREG / DG that stably expresses EREG protein was isolated.
- EGF (R) EGF (R) proliferation activation was caused by EREG.
- an anti-EREG neutralizing antibody EP20, EP27; WO2008047723
- an anti-EGFR neutralizing antibody cetuximab Merck KGaA
- EREG / DG cells were suspended in ⁇ MEM medium (Invitrogen) containing 10% FBS and propagated to a 96-well plate at 1 ⁇ 10 4 / well. The cells were incubated overnight at 37 ° C., 5% CO 2 and adhered to the plates. After removing the culture solution, EGF (R) cells (1 ⁇ 10 4 ) suspended in RPMI1640 containing 10% FBS were added together with the antibody. Incubation for 3 days was performed.
- ⁇ MEM medium Invitrogen
- FIG. 2 shows the luciferase activity, that is, the number of EGF (R) cells under each condition. It shows that EGF (R) proliferation is promoted by co-culturing EREG / DG, and the promotion is invalidated by addition of antibody. Compared with the culture conditions without EREG / DG44 cells (EGF (R) only), the coexistence (control) of EREG / DG44 cells significantly promoted the proliferation of EGF (R) cells. Cell proliferation promoted by the coexistence of EREG / DG44 cells was suppressed by addition of anti-EREG or anti-EGFR antibody. From the above results, it was revealed that EREG-expressing cells induce signal activation of nearby EGFR-expressing cells. Invalidation by anti-EREG antibody indicates that EREG is involved in signal activation.
- EGF (R) The proliferation of EGF (R) is promoted by the coexistence of colon cancer strain DLD-1 cells. This growth promotion is abolished by adding anti-EREG neutralizing antibody and anti-TGF- ⁇ neutralizing antibody.
- Multiple EGF ligands may be involved in EGFR activation in cancer cells. When multiple ligands are involved in receptor activation, inhibiting only one ligand is not sufficient to completely inhibit receptor activation.
- DLD-1 in addition to EREG, AREG and TGF ⁇ mRNA expression is enhanced.
- EGF (R) and DLD-1 were co-cultured in the presence of neutralizing antibody, and the ligand involved in EGF (R) activation was identified by monitoring the growth of EGF (R).
- DLD-1 cells were suspended in RPMI1640 medium containing 10% FBS and propagated to a 96-well plate at 2 ⁇ 10 3 / well. After incubation overnight at 37 ° C., 5% CO 2 , the culture supernatant was removed. EGF (R) cells (1 ⁇ 10 4 ) suspended in RPMI 1640 containing 10% FBS were added together with antibodies having neutralizing activity. After 3 days of incubation, cell number was quantified by measuring luciferase activity.
- EGF (R) proliferation promotion by DLD-1 cells is mediated by EGFR stimulation.
- Anti-TGF ⁇ antibody (R & D® Systems, AB-239-NA) partially inhibited EGF (R) proliferation promoted by DLD1 co-culture.
- Anti-EREG antibody EP20 partially suppressed cell proliferation.
- Anti-AREG antibody (R & D®Systems, ®MAB262) did not inhibit proliferation at all.
- the effect obtained by simultaneously adding the anti-EREG antibody and the anti-AREG antibody was similar to that of the anti-EREG antibody alone.
- anti-TGF ⁇ antibody and anti-AREG antibody were added simultaneously, the growth of EGF (R) was not suppressed more than anti-TGF ⁇ antibody alone.
- Fig. 8 shows flow cytometry analysis for AREG protein and EREG protein on DLD-1 cells.
- DLD-1 was detached from the culture dish by treatment with PBS / 1 mM EDTA. The cells were centrifuged and the cell pellet was resuspended in PBS containing 1% FBS.
- Anti-EREG antibody (EP20) and anti-AREG antibody were added to the cell resuspension to a final concentration of 4 ⁇ g / ml. Incubated for 30 minutes on ice. After the incubation, unreacted antibodies were removed by centrifugation and resuspension.
- FITC-labeled anti-mouse IgG antibody (Beckman Coulter, IM0819) was added to the cell suspension after washing, and incubated on ice for 30 minutes. After centrifugation and resuspension, the cells were analyzed with a flow cytometer FACSCalibur. The results showed that AREG protein and EREG protein were expressed on DLD1.
- Anti-EGREG antibody and anti-TGF ⁇ antibody inhibit the proliferation of DLD1 It was analyzed whether the proliferation of DLD-1 itself was inhibited by blocking the EGFR signal. DLD-1 was suspended in RPMI 1640 containing 10% FBS, and placed in a 96-well plate to 2 ⁇ 10 3 / well. A low adhesion Costar® 3595 plate was used to weaken the anchorage-dependent growth potential. Various neutralizing antibodies (10 ⁇ g / ml) and a topoisomerase inhibitor Campto (Yakult) were added, followed by incubation at 37 ° C. under 5% CO 2 .
- Cetuximab has been reported to be enhanced when combined with topoisomerase inhibitors (Ciardiello et al. Clin Cancer Res 5: 909-916, 1999). After 3 days incubation, cell proliferation was quantified by WST-8 assay.
- cetuximab inhibited the growth of DLD-1 cells compared to the control (FIG. 4).
- EP20 or anti-TGF ⁇ antibody alone did not inhibit proliferation.
- Simultaneous addition of both antibodies suppressed DLD-1 growth to the same extent as cetuximab.
- colorectal cancer cells it has been clarified that proliferation is suppressed by neutralizing EREG and TGF ⁇ that are endogenously expressed by the addition of antibodies simultaneously.
- EREG and TGF ⁇ are members of the EGF ligand family. As shown in FIG. 5, there are few amino acid residues conserved in EREG and TGF ⁇ except for Cys which forms a disulfide bond. Because of the small number of amino acids conserved in both, it seems that there usually cannot be a specific antibody that binds to both EREG and TGF ⁇ . Based on the EREG conformational analysis, the present inventors have found that a few three amino acid residues conserved in EREG and TGF ⁇ form a common three-dimensional structural epitope (FIG. 5).
- SMART-5'-RACE cDNA library was prepared from total hybridoma RNA producing antibody EP19. Total RNA preparation was performed using RNeasy Mini columns. The cDNA library was prepared according to the manufacturer's instructions. Antibody variable region sequences (VH, VL) were PCR amplified using primers complementary to the antibody constant region sequences. The PCR amplified fragment was cloned into pCR2.1TOPO and its nucleotide sequence was determined. The H chain variable region base sequence is shown in SEQ ID NO: 5, and the translated sequence (amino acid sequence) is shown in SEQ ID NO: 6.
- the nucleotide sequence of the L chain variable region is shown in SEQ ID NO: 7, and the translated sequence (amino acid sequence) is shown in SEQ ID NO: 8.
- a chimeric antibody expression vector comprising the VH and VL sequences and a human IgG1 constant region sequence was constructed.
- the base sequence of the chimeric antibody heavy chain is shown in SEQ ID NO: 21, the amino acid sequence thereof is shown in SEQ ID NO: 22, the base sequence of the chimeric antibody light chain is shown in SEQ ID NO: 23, and the amino acid sequence thereof is shown in SEQ ID NO: 24.
- the expression vector was introduced into COS-7 to transiently express the chimeric antibody.
- the concentration of the chimeric antibody in the culture supernatant was calculated by sandwich ELISA.
- a purified chimeric antibody having another mouse variable region sequence was used.
- EREG or TGF ⁇ was immobilized on a Nunc immunoplate, and the binding of the chimeric antibody to the immobilized protein was analyzed.
- the EP19 recombinant chimeric antibody bound to EREG and TGF ⁇ in a dose-dependent manner (FIG. 6).
- the EP20 recombinant chimeric antibody bound to EREG and did not bind to TGF ⁇ at all.
- binding to the EREG protein was not observed in a commercially available anti-TGF ⁇ polyclonal antibody (R & D® System, AB-2390-NA: lot T06).
- Example 6 Expression of EREG is induced by activation of EGFR EREG expression control in the renal cancer cell line Caki1 was analyzed. It was found by chance that the number of EREG proteins on the surface of Caki1 cells changes depending on the cell culture state.
- Caki1 was maintained in MEM (Invitrogen) medium containing 10% FBS. Cells were seeded at 0.3-1.5 ⁇ 10 4 cells / cm 2 and incubated for 2-3 days. After incubation, the cells were detached with PBS / EDTA. The cells were centrifuged and the cell pellet was resuspended in PBS containing 1% FBS.
- Anti-EREG antibody (EP20) and anti-EGFR antibody (cetuximab) were added to the cell resuspension to a final concentration of 4 ⁇ g / ml. Incubated for 30 minutes on ice. After the incubation, unreacted antibodies were removed by centrifugation and resuspension. After washing, the FITC-labeled secondary antibody was added to the cell suspension and incubated on ice for 30 minutes. After the cells were centrifuged and resuspended, the expression of EREG protein and EGFR protein was analyzed with a flow cytometer FACSCalibur (FIG. 7).
- EGFR is activated and simultaneously taken into cells by addition of a soluble ligand.
- EGFR signaling is activated by stimulation with membrane ligands, but activated EGFR may not be taken into cells due to binding with membrane ligands.
- Many cancer cells express both EGFR and EGF ligands. It was speculated that the membrane-type ligand EREG might be involved in the mechanism that maintains constant EGFR signal activation in a dense state in cancer tissues.
- TGF ⁇ recombinant protein Human TGF ⁇ cDNA amplified by PCR was isolated from the human colon cancer cell line DLD-1 cDNA library.
- pMCN is a vector that allows a foreign gene to be expressed under the mouse CMV promoter (GenBank: U68299).
- the pMCN vector has a geneticin resistance gene.
- DG44 cells were transformed with the linearized expression vector by electroporation. Cells into which the vector was introduced were selected in the presence of geneticin. By quantifying the amount of Fc protein in the culture supernatant, cells that highly produced the recombinant protein were selected.
- the target protein was purified from the culture supernatant of the Fc fusion protein-producing strain by using Protein G affinity column chromatography and gel filtration chromatography. Purified protein concentration was determined by DC protein assay (BioRad) using known concentrations of IgG as a standard.
- An E. coli expression vector of thioredoxin-mature human TGF ⁇ fusion protein (SEQ ID NO: 39, polynucleotide sequence SEQ ID NO: 40) was constructed by inserting the TGF ⁇ DNA fragment into pET32a (Novagen).
- TrxTGFA the thioredoxin-mature human TGF ⁇ fusion protein
- BL21 (DE) was transformed with the expression vector.
- the expression of the fusion protein was induced by the addition of IPTG.
- the fusion protein was purified from the soluble fraction of Escherichia coli by using Q Sepharose FF and HisTrap columns (GE Healthcare). It was confirmed by SDS-PAGE that the molecular weight size of the purified protein was as expected.
- the protein concentration was determined by DC protein assay.
- Example 8 Isolation of anti-TGF ⁇ antibody having neutralizing ability
- Balb / c mice (Nippon Charles River) were immunized with TGF ⁇ -Fc. That is, at the time of the first immunization, an antigen emulsion was prepared using Freund's complete adjuvant (Becton Dickinson), and the antigen protein was subcutaneously administered to 0.1 mg human TGF ⁇ -Fc / head. After an interval of two weeks, an antigen emulsion made with Freund's incomplete adjuvant was administered subcutaneously in a volume of 0.05 mg / head once a week for a total of seven times. 0.05 mg of the antigen protein was intravenously administered to individual mice whose serum antibody titers were elevated.
- Freund's complete adjuvant Becton Dickinson
- the spleen cells were removed, mixed with mouse myeloma cells P3-X63Ag8U1 (ATCC) at a cell number ratio of about 3: 1, and then cell fusion was performed by the polyethylene glycol (PEG) method. . After removing PEG by centrifugation, the cells are suspended in RPMI1640 medium containing 1x HAT media supplemet (Sigma), 0.5x BM-Condimed H1 Hybridoma cloning supplement (Roche Diagnostics), 10% fetal calf serum, The cell concentration was adjusted. Next, cells were seeded in 96 well plates.
- the presence or absence of anti-TGF ⁇ antibody contained in the culture supernatant was analyzed by ELISA using a plate coated with human TGF ⁇ -Fc.
- Hybridoma cells contained in positive wells were cloned by limiting dilution, and a hybridoma strain producing anti-TGF ⁇ antibody was isolated.
- the isotype of the monoclonal antibody was determined using IsoStrip (Roche Diagnostics). From the established hybridoma culture supernatant, IgG monoclonal antibody was purified by Protein G affinity column chromatography and desalting. Purified antibody concentration was determined by DC protein assay.
- FIG. 9 shows that mouse hybridoma antibody # 2-20 bound to mature TGF ⁇ in a dose-dependent manner.
- EGF (R) expressing the EGFR-GCSFR chimeric receptor proliferated depending on the added concentration of mature TGF ⁇ (FIG. 10 left).
- EGF (R) cells suspended in RPMI1640 containing 10% FBS were seeded in a 96-well culture plate at 5 ⁇ 10 3 / well. After adding TGF ⁇ to the appropriate final concentration, the plates were incubated for 3 days at 37 ° C., 5% CO 2 . Cell proliferation was quantified using Cell Count Reagent SF (Nacalai Tesque).
- EGF (R) cells The proliferation of EGF (R) cells by TGF ⁇ was neutralized by the addition of antibody # 2-20.
- EGF (R) cells suspended in RPMI1640 containing 10% FBS were seeded on the culture plate so as to be 5 ⁇ 10 3 / well.
- Antibody # 2-20 is first added to the cell suspension to a final concentration of 0, 0.1, 1, 10 ⁇ g / ml, and then TGF ⁇ is added to each of the culture plates to a final concentration of 5 ng / ml. Added to the wells. Cell growth was quantified after the culture plates were incubated for 3 days at 37 ° C., 5% CO 2 . Addition of antibody # 2-20 canceled cell proliferation by TGF ⁇ (FIG. 10 right).
- a soluble EGFR-Fc protein (SEQ ID NO: 41, polynucleotide sequence SEQ ID NO: 42) was prepared by the method described in International Publication WO2008 / 047914. TrxTGFA was biotinylated using a biotin protein labeling kit (Roche Diagnostics). EGFR-Fc protein was added to the nunc immunoplate coated with anti-human antibody at a concentration of 1 ⁇ g / ml and captured on the plate. After washing the plate, biotinylated TrxTGFA was added to each well of the plate to an appropriate concentration and the plate was incubated for 1 hour.
- the amount of TGF ⁇ bound to the EGFR protein was quantified by adding alkaline phosphatase-labeled streptavidin (Zymed) and the alkaline phosphatase substrate Sigma104. Biotinylated TrxTGFA bound to EGFR protein in a dose-dependent manner (FIG. 11 left). The inhibitory activity of antibody # 2-20 on the binding of TGF ⁇ and EGFR was tested. The # 2-20 antibody or anti-TGF ⁇ goat polyclonal antibody (R & D Systems, AB-239-NA) was added to the plate that captured the EGFR protein so that the final concentration was appropriate. Subsequently, biotinylated TrxTGFA was added to a final concentration of 50 ng / ml.
- TGF ⁇ bound to EGFR was determined. Binding of TGF ⁇ to the EGFR protein was inhibited by the addition of mouse # 2-20 antibody and goat polyclonal anti-TGF ⁇ antibody (FIG. 11 right).
- a Smart-5'-RACE cDNA library (Clontech) was constructed from the hybridoma total RNA producing anti-TGF ⁇ antibody # 2-20. Total RNA was prepared using RNAeasy Mini columns (Qiagen). The cDNA library was prepared according to the manufacturer's instructions. Antibody variable region sequences (VH, VL) were amplified by PCR using primers complementary to the antibody constant region sequences. The PCR amplified fragment was cloned into pCR2.1TOPO and its nucleotide sequence was determined. Its H chain variable region base sequence is shown in SEQ ID NO: 43, and its translated sequence (amino acid sequence) is shown in SEQ ID NO: 44.
- the L chain variable region base sequence is shown in SEQ ID NO: 45, and the translated sequence (amino acid sequence) is shown in SEQ ID NO: 46.
- the sequences of CDR1, CDR2, and CDR3 of the antibody heavy chain are shown in SEQ ID NOs: 47, 48, and 49.
- the sequences of CDR1, CDR2, and CDR3 of the antibody light chain are shown in SEQ ID NOs: 50, 51, and 52.
- a chimeric antibody expression vector comprising the VH and VL sequences described above and a human IgG1 constant region sequence was constructed.
- the polynucleotide sequence and amino acid sequence of the chimeric antibody heavy chain are shown in SEQ ID NO: 53 and SEQ ID NO: 54, respectively, and the polynucleotide sequence and amino acid sequence of the chimeric antibody light chain are shown in SEQ ID NO: 55 and SEQ ID NO: 56, respectively.
- a mouse chimeric IgG2a recombinant antibody expression vector composed of VH and VL sequences and a mouse IgG2a constant region sequence was constructed.
- the polynucleotide sequence and amino acid sequence of the mouse recombinant antibody heavy chain are shown in SEQ ID NO: 57 and SEQ ID NO: 58, respectively.
- the polynucleotide sequence and amino acid sequence of the mouse recombinant antibody light chain are shown in SEQ ID NO: 59 and SEQ ID NO: 60, respectively.
- Is shown in Both light chain and heavy chain DNAs are incorporated into one mammalian cell expression vector, and both are transcribed under the mouse CMV promoter.
- the chimeric antibody expression vector was introduced into COS-7 cells using the transfection reagent FuGENE6 (Roche Diagnostics), and the recombinant antibody was transiently expressed.
- the concentration of the chimeric antibody in the COS-7 culture supernatant was calculated by the sandwich ELISA method. At this time, a known concentration of human chimeric antibody, mouse IgG2a was used as a standard for concentration calculation.
- the binding ability of the recombinant antibody to TGF ⁇ and the binding inhibitory activity between TGF ⁇ and EGFR were evaluated by the methods described above.
- the recombinant mouse chimeric IgG2a antibody inhibited the binding of biotinylated TrxTGFA to the EGFR protein (FIG. 12 right).
Landscapes
- Health & Medical Sciences (AREA)
- Chemical & Material Sciences (AREA)
- Life Sciences & Earth Sciences (AREA)
- Medicinal Chemistry (AREA)
- Engineering & Computer Science (AREA)
- General Health & Medical Sciences (AREA)
- Organic Chemistry (AREA)
- Public Health (AREA)
- Pharmacology & Pharmacy (AREA)
- Animal Behavior & Ethology (AREA)
- Veterinary Medicine (AREA)
- Immunology (AREA)
- Bioinformatics & Cheminformatics (AREA)
- Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
- General Chemical & Material Sciences (AREA)
- Chemical Kinetics & Catalysis (AREA)
- Molecular Biology (AREA)
- Biomedical Technology (AREA)
- Urology & Nephrology (AREA)
- Biochemistry (AREA)
- Hematology (AREA)
- Epidemiology (AREA)
- Microbiology (AREA)
- Genetics & Genomics (AREA)
- Proteomics, Peptides & Aminoacids (AREA)
- Biophysics (AREA)
- General Physics & Mathematics (AREA)
- Pathology (AREA)
- Toxicology (AREA)
- Tropical Medicine & Parasitology (AREA)
- Biotechnology (AREA)
- Cell Biology (AREA)
- Food Science & Technology (AREA)
- Physics & Mathematics (AREA)
- Analytical Chemistry (AREA)
- Endocrinology (AREA)
- Mycology (AREA)
- Oncology (AREA)
- Pulmonology (AREA)
- Medicines Containing Antibodies Or Antigens For Use As Internal Diagnostic Agents (AREA)
Abstract
Description
上皮増殖因子受容体(epidermal growth factor receptor: EGFR)ファミリーerbB受容体は4つのメンバーEGFR(erbB1), erbB2, erbB3, erbB4から、EGFファミリーのリガンドは13のメンバーEGF, TGFα(transforming growth factor α), HB-EGF (heparin-binding EGF-like growth factor), BTC (betacellulin), AREG(amphiregulin), EREG(epiregulin), EPGN(epigen), NRGs(neuregulin1-6)からなる(非特許文献3参照)。erbB受容体の活性化は、リガンドの存在(時間空間的な発現制御に基づく)および翻訳後プロセッシングにより制御される。EGFファミリーのリガンドはI型膜蛋白質として生成し、膜貫通型から、プロテアーゼによるプロセスを受け、可溶型リガンドとして機能する。またリガンドのうちいくつかは膜型のまま近接する細胞上の受容体を活性化する。すなわちEGFファミリーリガンドによる受容体活性化機構は、分泌ペプチドが血流を介して全身的に広がり受容体発現細胞に作用する内分泌、分泌ペプチドが発現細胞自身の受容体を刺激するオートクライン、分泌ペプチドが近傍に存在する細胞上受容体を活性化するパラクライン、膜型リガンドが隣接する細胞受容体を刺激するジャクスタクラインからなる(非特許文献6参照)。
ヒト上皮系癌におけるEGFR変異、発現上昇は以前より報告されており、EGFR自身もしくはその下流シグナルの活性亢進が癌の発生と進展に重要な役割を持つことが明らかにされている。変異したEGFRのチロシンリン酸化活性の亢進、オートクラインループを通じたリガンドによるEGFR活性化、受容体染色体遺伝子の増幅は、乳癌、前立腺癌、膵臓癌、大腸癌、肺癌、頭頸部癌などのさまざまな癌で見受けられる共通の特徴である。非腫瘍細胞株に活性化されたEGFRsを導入することで、癌化形質を誘導することが実験的に示されている。癌細胞上のEGFRはしばし、腫瘍中に存在するEGFリガンドの存在により恒常的活性化を受けている。TGFα、AREG、HB-EGFを発現する癌は予後不良や化療剤耐性と関連するとの報告も存在する。EGFRシグナル経路のK-RAS恒常的活性化は、EREGの発現を誘導し、腫瘍形成能を亢進させる(非特許文献1参照)。
〔1〕 EREGアンタゴニストおよびTGFαアンタゴニストを有効成分として含む医薬組成物、
〔2〕 医薬組成物が細胞増殖抑制剤である〔1〕に記載の医薬組成物、
〔3〕 医薬組成物が抗癌剤である〔1〕に記載の医薬組成物、
〔4〕 EREGアンタゴニストが抗EREG抗体である〔1〕~〔3〕いずれかに記載の医薬組成物、
〔5〕 抗EREG抗体が以下の(a)~(x)いずれかに記載の抗体である〔1〕~〔4〕いずれかに記載の医薬組成物;
(a)配列番号:61に記載のアミノ酸配列を有するCDR1、配列番号:62に記載のアミノ酸配列を有するCDR2、配列番号:63に記載のアミノ酸配列を有するCDR3を含む重鎖を含む抗体、
(b)配列番号:64に記載のアミノ酸配列を有するCDR1、配列番号:65に記載のアミノ酸配列を有するCDR2、配列番号:66に記載のアミノ酸配列を有するCDR3を含む軽鎖を含む抗体、
(c)(a)の重鎖と(b)の軽鎖を含む抗体、
(d)(c)の抗体が認識するエピトープと同じエピトープを認識する抗体、
(e)配列番号:67に記載のアミノ酸配列を有するCDR1、配列番号:68に記載のアミノ酸配列を有するCDR2、配列番号:69に記載のアミノ酸配列を有するCDR3を含む重鎖を含む抗体、
(f)配列番号:70に記載のアミノ酸配列を有するCDR1、配列番号:71に記載のアミノ酸配列を有するCDR2、配列番号:72に記載のアミノ酸配列を有するCDR3を含む軽鎖を含む抗体、
(g)(e)の重鎖と(f)の軽鎖を含む抗体、
(h)(g)の抗体が認識するエピトープと同じエピトープを認識する抗体、
(i)配列番号:73に記載のアミノ酸配列を有するCDR1、配列番号:74に記載のアミノ酸配列を有するCDR2、配列番号:75に記載のアミノ酸配列を有するCDR3を含む重鎖を含む抗体、
(j)配列番号:76に記載のアミノ酸配列を有するCDR1、配列番号:77に記載のアミノ酸配列を有するCDR2、配列番号:78に記載のアミノ酸配列を有するCDR3を含む軽鎖を含む抗体、
(k)(i)の重鎖と(j)の軽鎖を含む抗体、
(l)(k)の抗体が認識するエピトープと同じエピトープを認識する抗体、
(m)配列番号:79に記載のアミノ酸配列を有するCDR1、配列番号:80に記載のアミノ酸配列を有するCDR2、配列番号:81に記載のアミノ酸配列を有するCDR3を含む重鎖を含む抗体、
(n)配列番号:82に記載のアミノ酸配列を有するCDR1、配列番号:83に記載のアミノ酸配列を有するCDR2、配列番号:84に記載のアミノ酸配列を有するCDR3を含む軽鎖を含む抗体、
(o)(m)の重鎖と(n)の軽鎖を含む抗体、
(p)(o)の抗体が認識するエピトープと同じエピトープを認識する抗体、
(q)配列番号:85に記載のアミノ酸配列を有するCDR1、配列番号:86に記載のアミノ酸配列を有するCDR2、配列番号:87に記載のアミノ酸配列を有するCDR3を含む重鎖を含む抗体、
(r)配列番号:88に記載のアミノ酸配列を有するCDR1、配列番号:89に記載のアミノ酸配列を有するCDR2、配列番号:90に記載のアミノ酸配列を有するCDR3を含む軽鎖を含む抗体、
(s)(q)の重鎖と(r)の軽鎖を含む抗体、
(t)(s)の抗体が認識するエピトープと同じエピトープを認識する抗体、
(u)配列番号:91に記載のアミノ酸配列を有するCDR1、配列番号:92に記載のアミノ酸配列を有するCDR2、配列番号:93に記載のアミノ酸配列を有するCDR3を含む重鎖を含む抗体、
(v)配列番号:94に記載のアミノ酸配列を有するCDR1、配列番号:95に記載のアミノ酸配列を有するCDR2、配列番号:96に記載のアミノ酸配列を有するCDR3を含む軽鎖を含む抗体、
(w)(u)の重鎖と(v)の軽鎖を含む抗体、
(x)(w)の抗体が認識するエピトープと同じエピトープを認識する抗体、
〔6〕 TGFαアンタゴニストが抗TGFα抗体である〔1〕~〔3〕いずれかに記載の医薬組成物、
〔7〕 抗TGFα抗体が以下の(a)~(d)いずれかに記載の抗体である〔6〕に記載の医薬組成物;
(a)配列番号:47に記載のアミノ酸配列を有するCDR1、配列番号:48に記載のアミノ酸配列を有するCDR2、配列番号:49に記載のアミノ酸配列を有するCDR3を含む重鎖を含む抗体、
(b)配列番号:50に記載のアミノ酸配列を有するCDR1、配列番号:51に記載のアミノ酸配列を有するCDR2、配列番号:52に記載のアミノ酸配列を有するCDR3を含む軽鎖を含む抗体、
(c)(a)の重鎖と(b)の軽鎖を含む抗体、
(d)(c)の抗体が認識するエピトープと同じエピトープを認識する抗体、
〔8〕 EREGアンタゴニストが抗EREG抗体であり、TGFαアンタゴニストが抗TGFα抗体である〔1〕~〔3〕いずれかに記載の医薬組成物、
〔9〕 抗EREG抗体が以下の(a)~(x)いずれかに記載の抗体である〔8〕に記載の医薬組成物;
(a)配列番号:61に記載のアミノ酸配列を有するCDR1、配列番号:62に記載のアミノ酸配列を有するCDR2、配列番号:63に記載のアミノ酸配列を有するCDR3を含む重鎖を含む抗体、
(b)配列番号:64に記載のアミノ酸配列を有するCDR1、配列番号:65に記載のアミノ酸配列を有するCDR2、配列番号:66に記載のアミノ酸配列を有するCDR3を含む軽鎖を含む抗体、
(c)(a)の重鎖と(b)の軽鎖を含む抗体、
(d)(c)の抗体が認識するエピトープと同じエピトープを認識する抗体、
(e)配列番号:67に記載のアミノ酸配列を有するCDR1、配列番号:68に記載のアミノ酸配列を有するCDR2、配列番号:69に記載のアミノ酸配列を有するCDR3を含む重鎖を含む抗体、
(f)配列番号:70に記載のアミノ酸配列を有するCDR1、配列番号:71に記載のアミノ酸配列を有するCDR2、配列番号:72に記載のアミノ酸配列を有するCDR3を含む軽鎖を含む抗体、
(g)(e)の重鎖と(f)の軽鎖を含む抗体、
(h)(g)の抗体が認識するエピトープと同じエピトープを認識する抗体、
(i)配列番号:73に記載のアミノ酸配列を有するCDR1、配列番号:74に記載のアミノ酸配列を有するCDR2、配列番号:75に記載のアミノ酸配列を有するCDR3を含む重鎖を含む抗体、
(j)配列番号:76に記載のアミノ酸配列を有するCDR1、配列番号:77に記載のアミノ酸配列を有するCDR2、配列番号:78に記載のアミノ酸配列を有するCDR3を含む軽鎖を含む抗体、
(k)(i)の重鎖と(j)の軽鎖を含む抗体、
(l)(k)の抗体が認識するエピトープと同じエピトープを認識する抗体、
(m)配列番号:79に記載のアミノ酸配列を有するCDR1、配列番号:80に記載のアミノ酸配列を有するCDR2、配列番号:81に記載のアミノ酸配列を有するCDR3を含む重鎖を含む抗体、
(n)配列番号:82に記載のアミノ酸配列を有するCDR1、配列番号:83に記載のアミノ酸配列を有するCDR2、配列番号:84に記載のアミノ酸配列を有するCDR3を含む軽鎖を含む抗体、
(o)(m)の重鎖と(n)の軽鎖を含む抗体、
(p)(o)の抗体が認識するエピトープと同じエピトープを認識する抗体、
(q)配列番号:85に記載のアミノ酸配列を有するCDR1、配列番号:86に記載のアミノ酸配列を有するCDR2、配列番号:87に記載のアミノ酸配列を有するCDR3を含む重鎖を含む抗体、
(r)配列番号:88に記載のアミノ酸配列を有するCDR1、配列番号:89に記載のアミノ酸配列を有するCDR2、配列番号:90に記載のアミノ酸配列を有するCDR3を含む軽鎖を含む抗体、
(s)(q)の重鎖と(r)の軽鎖を含む抗体、
(t)(s)の抗体が認識するエピトープと同じエピトープを認識する抗体、
(u)配列番号:91に記載のアミノ酸配列を有するCDR1、配列番号:92に記載のアミノ酸配列を有するCDR2、配列番号:93に記載のアミノ酸配列を有するCDR3を含む重鎖を含む抗体、
(v)配列番号:94に記載のアミノ酸配列を有するCDR1、配列番号:95に記載のアミノ酸配列を有するCDR2、配列番号:96に記載のアミノ酸配列を有するCDR3を含む軽鎖を含む抗体、
(w)(u)の重鎖と(v)の軽鎖を含む抗体、
(x)(w)の抗体が認識するエピトープと同じエピトープを認識する抗体、
〔10〕 抗TGFα抗体が以下の(a)~(d)いずれかに記載の抗体である〔8〕又は〔9〕に記載の医薬組成物;
(a)配列番号:47に記載のアミノ酸配列を有するCDR1、配列番号:48に記載のアミノ酸配列を有するCDR2、配列番号:49に記載のアミノ酸配列を有するCDR3を含む重鎖を含む抗体、
(b)配列番号:50に記載のアミノ酸配列を有するCDR1、配列番号:51に記載のアミノ酸配列を有するCDR2、配列番号:52に記載のアミノ酸配列を有するCDR3を含む軽鎖を含む抗体、
(c)(a)の重鎖と(b)の軽鎖を含む抗体、
(d)(c)の抗体が認識するエピトープと同じエピトープを認識する抗体、
〔11〕 EREGおよびTGFαに結合する抗体、
〔12〕 EREGおよびTGFαに対してアンタゴニスト活性を有する〔11〕に記載の抗体、
〔13〕 以下の(a)~(d)いずれかに記載の抗体である〔11〕又は〔12〕に記載の抗体;
(a)配列番号:10に記載のアミノ酸配列を有するCDR1、配列番号:12に記載のアミノ酸配列を有するCDR2、配列番号:14に記載のアミノ酸配列を有するCDR3を含む重鎖を含む抗体、
(b)配列番号:16に記載のアミノ酸配列を有するCDR1、配列番号:18に記載のアミノ酸配列を有するCDR2、配列番号:20に記載のアミノ酸配列を有するCDR3を含む軽鎖を含む抗体、
(c)(a)の重鎖と(b)の軽鎖を含む抗体、
(d)(c)の抗体が認識するエピトープと同じエピトープを認識する抗体、
〔14〕 配列番号:4(human EREG)の56番目のGlnから102番目のLeuまでの領域および/または配列番号:26(human TGFα)の41番目のValから87番目のLeuまでの領域を認識する抗体、
〔15〕 EREGおよびTGFαに結合する物質を有効成分として含む医薬組成物、
〔16〕 EREGおよびTGFαに結合する物質が〔11〕~〔14〕いずれかに記載の抗体である〔15〕に記載の医薬組成物、
〔17〕 以下の工程を含む抗癌剤のスクリーニング方法;
(a)被検物質のEREGに対するアンタゴニスト活性を測定する工程、
(b)被検物質のTGFαに対するアンタゴニスト活性を測定する工程、
(c)EREGおよびTGFαにアンタゴニスト活性を有する被検物質を選択する工程、
を、提供するものである。
〔18〕 本発明の医薬組成物(例えば、EREGアンタゴニストおよびTGFαアンタゴニスト)を、対象へ投与する工程を含む、細胞増殖を抑制する方法。
〔19〕 本発明の医薬組成物(例えば、EREGアンタゴニストおよびTGFαアンタゴニスト)を、対象へ投与する工程を含む、癌の治療方法。
〔20〕 細胞増殖阻害または癌治療における使用のための、EREGアンタゴニストおよびTGFαアンタゴニストを含む組成物。
〔21〕 細胞増殖阻害剤または抗癌剤の製造における、EREGアンタゴニストおよびTGFαアンタゴニストを含む組成物の使用。
(a)配列番号:61に記載のアミノ酸配列を有するCDR1、配列番号:62に記載のアミノ酸配列を有するCDR2、配列番号:63に記載のアミノ酸配列を有するCDR3を含む重鎖を含む抗体、
(b)配列番号:64に記載のアミノ酸配列を有するCDR1、配列番号:65に記載のアミノ酸配列を有するCDR2、配列番号:66に記載のアミノ酸配列を有するCDR3を含む軽鎖を含む抗体、
(c)(a)の重鎖と(b)の軽鎖を含む抗体、
(d)(c)の抗体が認識するエピトープと同じエピトープを認識する抗体、
(e)配列番号:67に記載のアミノ酸配列を有するCDR1、配列番号:68に記載のアミノ酸配列を有するCDR2、配列番号:69に記載のアミノ酸配列を有するCDR3を含む重鎖を含む抗体、
(f)配列番号:70に記載のアミノ酸配列を有するCDR1、配列番号:71に記載のアミノ酸配列を有するCDR2、配列番号:72に記載のアミノ酸配列を有するCDR3を含む軽鎖を含む抗体、
(g)(e)の重鎖と(f)の軽鎖を含む抗体、
(h)(g)の抗体が認識するエピトープと同じエピトープを認識する抗体、
(i)配列番号:73に記載のアミノ酸配列を有するCDR1、配列番号:74に記載のアミノ酸配列を有するCDR2、配列番号:75に記載のアミノ酸配列を有するCDR3を含む重鎖を含む抗体、
(j)配列番号:76に記載のアミノ酸配列を有するCDR1、配列番号:77に記載のアミノ酸配列を有するCDR2、配列番号:78に記載のアミノ酸配列を有するCDR3を含む軽鎖を含む抗体、
(k)(i)の重鎖と(j)の軽鎖を含む抗体、
(l)(k)の抗体が認識するエピトープと同じエピトープを認識する抗体、
(m)配列番号:79に記載のアミノ酸配列を有するCDR1、配列番号:80に記載のアミノ酸配列を有するCDR2、配列番号:81に記載のアミノ酸配列を有するCDR3を含む重鎖を含む抗体、
(n)配列番号:82に記載のアミノ酸配列を有するCDR1、配列番号:83に記載のアミノ酸配列を有するCDR2、配列番号:84に記載のアミノ酸配列を有するCDR3を含む軽鎖を含む抗体、
(o)(m)の重鎖と(n)の軽鎖を含む抗体、
(p)(o)の抗体が認識するエピトープと同じエピトープを認識する抗体、
(q)配列番号:85に記載のアミノ酸配列を有するCDR1、配列番号:86に記載のアミノ酸配列を有するCDR2、配列番号:87に記載のアミノ酸配列を有するCDR3を含む重鎖を含む抗体、
(r)配列番号:88に記載のアミノ酸配列を有するCDR1、配列番号:89に記載のアミノ酸配列を有するCDR2、配列番号:90に記載のアミノ酸配列を有するCDR3を含む軽鎖を含む抗体、
(s)(q)の重鎖と(r)の軽鎖を含む抗体、
(t)(s)の抗体が認識するエピトープと同じエピトープを認識する抗体、
(u)配列番号:91に記載のアミノ酸配列を有するCDR1、配列番号:92に記載のアミノ酸配列を有するCDR2、配列番号:93に記載のアミノ酸配列を有するCDR3を含む重鎖を含む抗体、
(v)配列番号:94に記載のアミノ酸配列を有するCDR1、配列番号:95に記載のアミノ酸配列を有するCDR2、配列番号:96に記載のアミノ酸配列を有するCDR3を含む軽鎖を含む抗体、
(w)(u)の重鎖と(v)の軽鎖を含む抗体、
(x)(w)の抗体が認識するエピトープと同じエピトープを認識する抗体。
(a)配列番号:47に記載のアミノ酸配列を有するCDR1、配列番号:48に記載のアミノ酸配列を有するCDR2、配列番号:49に記載のアミノ酸配列を有するCDR3を含む重鎖を含む抗体、
(b)配列番号:50に記載のアミノ酸配列を有するCDR1、配列番号:51に記載のアミノ酸配列を有するCDR2、配列番号:52に記載のアミノ酸配列を有するCDR3を含む軽鎖を含む抗体、
(c)(a)の重鎖と(b)の軽鎖を含む抗体、
(d)(c)の抗体が認識するエピトープと同じエピトープを認識する抗体。
さらに、EREGアンタゴニスト活性とTGFαアンタゴニスト活性の両方の活性を有する物質とEREGアンタゴニスト活性のみを有する物質、又はEREGアンタゴニスト活性とTGFαアンタゴニスト活性の両方の活性を有する物質とTGFαアンタゴニスト活性のみを有する物質を有効成分としてもよい。
EREGへの結合は、例えば、可溶型EREGへの結合、膜貫通型EREGへの結合、可溶型EREGおよび膜貫通型EREGへの結合、等を挙げることができる。
TGFαへの結合は、例えば、可溶型TGFαへの結合、膜貫通型TGFαへの結合、可溶型TGFαおよび膜貫通型TGFαへの結合、等を挙げることができる。
(a)配列番号:10に記載のアミノ酸配列を有するCDR1、配列番号:12に記載のアミノ酸配列を有するCDR2、配列番号:14に記載のアミノ酸配列を有するCDR3を含む重鎖を含む抗体、
(b)配列番号:16に記載のアミノ酸配列を有するCDR1、配列番号:18に記載のアミノ酸配列を有するCDR2、配列番号:20に記載のアミノ酸配列を有するCDR3を含む軽鎖を含む抗体、
(c)(a)の重鎖と(b)の軽鎖を含む抗体、
(d)(c)の抗体が認識するエピトープと同じエピトープを認識する抗体。
ヒトEREG又はTGFαタンパク質のアミノ酸配列より化学合成によって取得されたペプチド
EREG遺伝子又はTGFα遺伝子の一部を発現ベクターに組込んで発現させることによっても取得されたペプチド
EREGタンパク質又はTGFαタンパク質をタンパク質分解酵素により分解することによって取得されたペプチド
部分ペプチドとして用いるEREG又はTGFαの領域および大きさは限定されない。
P3(P3x63Ag8.653)(J. Immunol.(1979)123, 1548-1550)
P3x63Ag8U.1(Current Topics in Microbiology and Immunology(1978)81, 1-7)
NS-1(Kohler. G. and Milstein, C. Eur. J. Immunol.(1976)6, 511-519)
MPC-11(Margulies. D.H. et al., Cell(1976)8, 405-415)
SP2/0 (Shulman, M. et al., Nature(1978)276, 269-270)
FO(de St. Groth, S. F. etal., J. Immunol. Methods(1980)35, 1-21)
S194(Trowbridge, I. S. J. Exp. Med.(1978)148, 313-323)、
R210(Galfre, G. et al., Nature(1979)277, 131-133)等
グアニジン超遠心法(Chirgwin, J. M. et al., Biochemistry(1979)18, 5294-5299)
AGPC法(Chomczynski, P.et al., Anal. Biochem.(1987)162, 156-159)
(1)ハイブリドーマから得られたcDNAによってコードされるV領域を含む抗体をEREGに接触させる工程、
(2)EREGと抗体との結合を検出する工程、および
(3)EREGに結合する抗体を選択する工程
(1)哺乳類細胞、:CHO、COS、ミエローマ、BHK(baby hamster kidney)、Hela、Veroなど
(2)両生類細胞:アフリカツメガエル卵母細胞など
(3)昆虫細胞:sf9、sf21、Tn5など
酵母:サッカロミセス・セレビシエ(Saccharomyces serevisiae)などのサッカロミセス(Saccharomyces)属、メタノール資化酵母(Pichia pastoris)などのPichia属
糸状菌:アスペスギルス・ニガー(Aspergillus niger)などのアスペルギルス(Aspergillus)属
パパイン消化:F(ab)2またはFab
ペプシン消化:F(ab')2またはFab'
プラスミン消化:Facb
上記のような酵素的に得られた抗体断片に対して、遺伝子工学的手法を利用する方法では抗体の任意の部分を欠失させることができる。
前記抗体のH鎖またはH鎖V領域をコードするDNA配列、および
前記抗体のL鎖またはL鎖V領域をコードするDNA配列
[VL]リンカー[VH]リンカー[VH]リンカー[VL]
[VH]リンカー[VL]リンカー[VL]リンカー[VH]
[VH]リンカー[VH]リンカー[VL]リンカー[VL]
[VL]リンカー[VL]リンカー[VH]リンカー[VH]
[VL]リンカー[VH]リンカー[VL]リンカー[VH]
Ser
Gly・Ser
Gly・Gly・Ser
Ser・Gly・Gly
Gly・Gly・Gly・Ser(配列番号:27)
Ser・Gly・Gly・Gly(配列番号:28)
Gly・Gly・Gly・Gly・Ser(配列番号:29)
Ser・Gly・Gly・Gly・Gly(配列番号:30)
Gly・Gly・Gly・Gly・Gly・Ser(配列番号:31)
Ser・Gly・Gly・Gly・Gly・Gly(配列番号:32)
Gly・Gly・Gly・Gly・Gly・Gly・Ser(配列番号:33)
Ser・Gly・Gly・Gly・Gly・Gly・Gly(配列番号:34)
(Gly・Gly・Gly・Gly・Ser(配列番号:29))n
(Ser・Gly・Gly・Gly・Gly(配列番号:30))n
[nは1以上の整数である]
[VH]ペプチドリンカー(15アミノ酸)[VL]ペプチドリンカー(15アミノ酸)[VH]ペプチドリンカー(15アミノ酸)[VL]
N-ヒドロキシスクシンイミド(NHS)、
ジスクシンイミジルスベレート(DSS)、
ビス(スルホスクシンイミジル)スベレート(BS3)、
ジチオビス(スクシンイミジルプロピオネート)(DSP)、
ジチオビス(スルホスクシンイミジルプロピオネート)(DTSSP)、
エチレングリコールビス(スクシンイミジルスクシネート)(EGS)、
エチレングリコールビス(スルホスクシンイミジルスクシネート)(スルホ-EGS)、
ジスクシンイミジル酒石酸塩(DST)、ジスルホスクシンイミジル酒石酸塩(スルホ-DST)、
ビス[2-(スクシンイミドオキシカルボニルオキシ)エチル]スルホン(BSOCOES)、および
ビス[2-(スルホスクシンイミドオキシカルボニルオキシ)エチル]スルホン(スルホ-BSOCOES)など
azaribine、anastrozole、azacytidine、bleomycin、bortezomib、bryostatin-1、busulfan、camptothecin、10-hydroxycamptothecin、carmustine、celebrex、chlorambucil、cisplatin、irinotecan、carboplatin、cladribine、cyclophosphamide、cytarabine、dacarbazine、docetaxel、dactinomycin、daunomycin glucuronide、daunorubicin、dexamethasone、diethylstilbestrol、doxorubicin、doxorubicin glucuronide、epirubicin、ethinyl estradiol、estramustine、etoposide、etoposide glucuronide、floxuridine、fludarabine、flutamide、fluorouracil、fluoxymesterone、gemcitabine、hydroxyprogesterone caproate、hydroxyurea、idarubicin、ifosfamide、leucovorin、lomustine、mechlorethamine、medroxyprogesterone acetate、megestrol acetate、melphalan、mercaptopurine、methotrexate、mitoxantrone、mithramycin、mitomycin、mitotane、phenylbutyrate、prednisone、procarbazine、paclitaxel、pentostatin、semustine streptozocin、tamoxifen、taxanes、taxol、testosterone propionate、thalidomide、thioguanine、thiotepa, teniposide、topotecan、uracil mustard、vinblastine、vinorelbine、vincristine
例えば、哺乳類細胞の場合、常用される有用なプロモーター、発現させる抗体遺伝子、その3’側下流にポリAシグナルを機能的に結合させて発現させることができる。例えばプロモーター/エンハンサーとしては、ヒトサイトメガロウイルス前期プロモーター/エンハンサー(human cytomegalovirus immediate early promoter/enhancer)を挙げることができる。
アミノグリコシドトランスフェラーゼ(APH)遺伝子、
チミジンキナーゼ(TK)遺伝子、
大腸菌キサンチングアニンホスホリボシルトランスフェラーゼ(Ecogpt)遺伝子、
ジヒドロ葉酸還元酵素(dhfr)遺伝子等
グリコシル化が修飾された抗体(WO99/54342など)、
糖鎖に付加するフコースが欠損した抗体(WO00/61739、WO02/31140など)、
バイセクティングGlcNAcを有する糖鎖を有する抗体(WO02/79255など)など
本発明における細胞傷害活性としては、例えば抗体依存性細胞介在性細胞傷害(antibody-dependent cell-mediated cytotoxicity:ADCC)活性、補体依存性細胞傷害(complement-dependent cytotoxicity:CDC)活性などを挙げることができる。本発明において、CDC活性とは補体系による細胞傷害活性を意味する。一方ADCC活性とは標的細胞の細胞表面抗原に特異的抗体が付着した際、そのFc部分にFcγ受容体保有細胞(免疫細胞等)がFcγ受容体を介して結合し、標的細胞に傷害を与える活性を意味する。
(1)エフェクター細胞の調製
CBA/Nマウスなどから脾臓を摘出し、RPMI1640培地(Invitrogen社製)中で脾臓細胞が分離される。10%ウシ胎児血清(FBS、HyClone社製)を含む同培地で洗浄後、細胞濃度を5×106/mlに調製することによって、エフェクター細胞が調製できる。
Baby Rabbit Complement(CEDARLANE社製)を10% FBS含有培地(Invitrogen社製)にて10倍希釈し、補体溶液が調製できる。
目的のタンパク質を発現する細胞を0.2 mCiの51Cr-クロム酸ナトリウム(GEヘルスケアジャパン社製)とともに、10% FBS含有DMEM培地中で37℃にて1時間培養することにより該標的細胞を放射性標識できる。目的のタンパク質を発現する細胞としては、目的のタンパク質をコードする遺伝子で形質転換された細胞、原発性大腸癌、転移性大腸癌、肺腺癌、膵癌、胃癌、腎癌細胞、大腸癌細胞、食道癌細胞、胃癌細胞、膵癌細胞等を利用することができる。放射性標識後、細胞を10% FBS含有RPMI1640培地にて3回洗浄し、細胞濃度を2×105/mlに調製することによって、該標的細胞が調製できる。
別の観点においては、本発明は、本発明の抗体を有効成分として含有する医薬組成物を提供する。又、本発明は本発明の抗体を有効成分として含有する細胞増殖抑制剤、特に抗癌剤に関する。
本発明は、次の工程を含む、癌の治療剤の候補化合物のスクリーニング方法を提供する。
(1)被検物質のEREGアンタゴニスト活性を測定する工程、
(2)被検物質のTGFαアンタゴニスト活性を測定する工程、および
(3)EREGアンタゴニスト活性およびTGFαアンタゴニスト活性を有する物質を選択する工程。
(1)EREGアンタゴニスト活性を有する物質を選択する工程、
(2)TGFαアンタゴニスト活性を有する物質を選択する工程、
(3)(1)で選択された物質と(2)で選択する物質を組み合わせて抗癌剤とする工程。
ヒトEGFR(GenBank: NM_005228)およびマウスGCSFR(GenBank: NM_007782)cDNAをPCR増幅することで単離した。EGFR細胞外ドメインとGCSFR細胞内ドメインからなるキメラ受容体遺伝子を作製した。キメラ受容体塩基配列を配列番号:1、アミノ酸配列を配列番号:2に示した。キメラ受容体の哺乳動物発現ベクターを構築した。発現ベクターにおいて、キメラ受容体はヒトEF1αプロモーター下で転写されるよう設計されている。キメラ受容体発現ベクターはジェネティシン耐性遺伝子を持っている。
配列番号:3で示されるEREG cDNA(NM_001432)を哺乳動物発現ベクターpMCNにクローニングした。pMCNは、マウスCMVプロモーター(GenBank: U68299)下で外来遺伝子を発現させることが可能である。pMCNベクターはジェネティシン耐性遺伝子をもっている。EREG発現ベクターでCHO細胞DG44株(Invitrogen)を形質転換した。ジェネティシン存在下で薬剤耐性細胞を選抜した。EREG蛋白質を安定発現する細胞株EREG/DGを単離した。
癌細胞のEGFR活性化には複数のEGFリガンドが関与しているかもしれない。複数のリガンドが受容体活性化に関与する場合、ひとつのリガンドを抑制するだけでは、受容体活性化を完全に抑制するには十分でない。DLD-1では、EREGに加え、AREG、TGFαのmRNA発現が亢進している。DLD-1に発現するEGFリガンドのなかでEGFR受容体活性化に係る分子はどれかを解析した。中和抗体存在下でEGF(R)とDLD-1を共培養し、EGF(R)の増殖をモニターすることでEGF(R)活性化に関与するリガンドを同定した。
DLD-1自身の増殖はEGFRシグナルを遮断することで抑制されるかどうか、を解析した。DLD-1は10% FBSを含むRPMI1640に懸濁され、2 x 103/wellとなるよう96穴プレートにまき込まれた。足場依存的な増殖能を弱めるため低接着Costar(登録商標)3595プレートが使用された。各種中和抗体(10μg/ml)およびトポイソメラーゼ阻害剤であるCampto(ヤクルト)が添加された後、37℃、5 % CO2下インキュベートされた。Cetuximabの抗腫瘍効果は、トポイソメラーゼ阻害剤と併用すると増強されると報告されている(Ciardiello et al. Clin Cancer Res 5:909-916, 1999)。3日間インキュベーション後、WST-8アッセイ法により細胞増殖は定量化された。
EREGとTGFαはEGFリガンドファミリーメンバーである。図5に示すようにEREGとTGFαで保存されているアミノ酸残基はジスルフィド結合形成するCysを除くとわずかである。両者で保存されているアミノ酸の少なさから、通常、EREGとTGFα両方に結合する特異的な抗体は存在し得ないと考えられる。本発明者らはEREG立体構造解析に基づき、EREGとTGFαで保存されているわずかなアミノ残基で、共通する3次元的な構造エピトープが形成されることを見出した(図5)。EREGに結合する抗体のほんのごく一部は、TGFαにも結合すると考えた。発明者らは出願WO 2008/047723において抗EREG抗体が複数取得されたことを示した。出願WO 2008/047723実施例中で取得した抗体がTGFαに対して結合する否か、解析した。TGFα(R&D Systems, 239-A)を固相化したNuncイムノプレートに、抗EREG抗体を添加し結合を解析した。37個のモノクローナル抗体を解析した中で、ただひとつ、抗体EP19がTGFαに結合した。
腎臓癌細胞株Caki1におけるEREG発現制御を解析した。Caki1細胞表面上のEREG蛋白質数が、細胞の培養状態により変化することを偶然見出した。Caki1は10% FBSを含むMEM(Invitrogen)培地で維持された。0.3~1.5 x 104 cells/cm2となるように細胞をまき込み、2~3日インキュベートした。インキュベーション後、PBS/EDTAで細胞を剥離した。細胞は遠心され、細胞ペレットは1% FBSを含むPBSに再懸濁された。細胞再懸濁液には、終濃度4μg/mlとなるように抗EREG抗体(EP20)、抗EGFR抗体(cetuximab)がそれぞれ添加された。氷上にて30分間インキュベートされた。インキュベーション後、未反応の抗体は、遠心、再懸濁を行うことで除去された。洗浄後の細胞懸濁液にはFITCラベルした2次抗体が添加され、氷上にて30分間インキュベートされた。細胞を遠心、再懸濁した後、EREG蛋白質およびEGFR蛋白質の発現はフローサイトメーターFACSCaliburで解析された(図7)。
ヒト大腸癌細胞株DLD-1 cDNAライブラリよりPCRによって増幅されたヒトTGFα cDNAが単離された。ヒトTGFα細胞外領域とマウスIgG2a抗体定常領域配列からなるキメラ蛋白質ヒトTGFα-Fc(配列番号:37)をコードするDNA(配列番号:38)が作製され、哺乳動物発現ベクターpMCNにクローニングされた。pMCNは、マウスCMVプロモータ(GenBank: U68299)下で外来遺伝子を発現させることを可能とするベクターである。pMCNベクターはジェネティシン耐性遺伝子をもっている。直鎖化した発現ベクターでDG44細胞がエレクトロポレーション法にて形質転換された。ベクターが導入された細胞がジェネティシン存在下で選抜された。培養上清中のFc蛋白質量を定量することによって、組換え蛋白質を高産生する細胞が選抜された。
Balb/cマウス(日本チャールズ・リバー)にTGFα-Fcが免疫された。すなわち、初回免疫時、フロイント完全アジュバント(べクトン・ディッキンソン)を用いて抗原エマルジョンが作製され、0.1 mg ヒトTGFα-Fc/headとなるように皮下に抗原蛋白質が投与された。二週の間隔を置いたのち、週一回合計7回、フロイント不完全アジュバントで作製された抗原エマルジョンが0.05 mg/headの容量で皮下投与された。血清抗体価の上昇が認められたマウス個体に対し0.05 mgの抗原蛋白質が静脈内に投与された。3日後にその脾臓細胞が摘出され、マウスミエローマ細胞P3-X63Ag8U1(ATCC)と細胞数比で約3:1の割合で混合された後に、ポリエチレングリコール(PEG)法にて細胞融合が行われた。遠心操作によりPEGを除去したのち、細胞は1x HAT media supplemet(シグマ)、0.5x BM-Condimed H1 Hybridoma cloning supplement(ロシュダイアグノスティック)、10%牛胎児血清を含むRPMI1640培地に懸濁されて、その細胞濃度が調整された。次に、96ウェルプレートに細胞が播種された。ハイブリドーマによるコロニー形成が確認された後、培養上清中に含まれる抗TGFα抗体の有無をヒトTGFα-Fcをコートしたプレートを用いてELISA解析した。陽性ウェルに含まれるハイブリドーマ細胞が限界希釈法によりクローン化され、抗TGFα抗体を産生するハイブリドーマ株が単離された。モノクローナル抗体のアイソタイプがIsoStrip(ロシュダイアグノスティック)を用いて決定された。樹立したハイブリドーマの培養上清より、ProteinGアフィニティカラムクロマトグラフィ、および脱塩処理によりIgGモノクローナル抗体が精製された。精製抗体濃度はDCプロテインアッセイにより決定された。
Claims (17)
- EREGアンタゴニストおよびTGFαアンタゴニストを有効成分として含む医薬組成物。
- 医薬組成物が細胞増殖抑制剤である請求項1に記載の医薬組成物。
- 医薬組成物が抗癌剤である請求項1に記載の医薬組成物。
- EREGアンタゴニストが抗EREG抗体である請求項1~3いずれかに記載の医薬組成物。
- 抗EREG抗体が以下の(a)~(x);
(a)配列番号:61に記載のアミノ酸配列を有するCDR1、配列番号:62に記載のアミノ酸配列を有するCDR2、配列番号:63に記載のアミノ酸配列を有するCDR3を含む重鎖を含む抗体、
(b)配列番号:64に記載のアミノ酸配列を有するCDR1、配列番号:65に記載のアミノ酸配列を有するCDR2、配列番号:66に記載のアミノ酸配列を有するCDR3を含む軽鎖を含む抗体、
(c)(a)の重鎖と(b)の軽鎖を含む抗体、
(d)(c)の抗体が認識するエピトープと同じエピトープを認識する抗体、
(e)配列番号:67に記載のアミノ酸配列を有するCDR1、配列番号:68に記載のアミノ酸配列を有するCDR2、配列番号:69に記載のアミノ酸配列を有するCDR3を含む重鎖を含む抗体、
(f)配列番号:70に記載のアミノ酸配列を有するCDR1、配列番号:71に記載のアミノ酸配列を有するCDR2、配列番号:72に記載のアミノ酸配列を有するCDR3を含む軽鎖を含む抗体、
(g)(e)の重鎖と(f)の軽鎖を含む抗体、
(h)(g)の抗体が認識するエピトープと同じエピトープを認識する抗体、
(i)配列番号:73に記載のアミノ酸配列を有するCDR1、配列番号:74に記載のアミノ酸配列を有するCDR2、配列番号:75に記載のアミノ酸配列を有するCDR3を含む重鎖を含む抗体、
(j)配列番号:76に記載のアミノ酸配列を有するCDR1、配列番号:77に記載のアミノ酸配列を有するCDR2、配列番号:78に記載のアミノ酸配列を有するCDR3を含む軽鎖を含む抗体、
(k)(i)の重鎖と(j)の軽鎖を含む抗体、
(l)(k)の抗体が認識するエピトープと同じエピトープを認識する抗体、
(m)配列番号:79に記載のアミノ酸配列を有するCDR1、配列番号:80に記載のアミノ酸配列を有するCDR2、配列番号:81に記載のアミノ酸配列を有するCDR3を含む重鎖を含む抗体、
(n)配列番号:82に記載のアミノ酸配列を有するCDR1、配列番号:83に記載のアミノ酸配列を有するCDR2、配列番号:84に記載のアミノ酸配列を有するCDR3を含む軽鎖を含む抗体、
(o)(m)の重鎖と(n)の軽鎖を含む抗体、
(p)(o)の抗体が認識するエピトープと同じエピトープを認識する抗体、
(q)配列番号:85に記載のアミノ酸配列を有するCDR1、配列番号:86に記載のアミノ酸配列を有するCDR2、配列番号:87に記載のアミノ酸配列を有するCDR3を含む重鎖を含む抗体、
(r)配列番号:88に記載のアミノ酸配列を有するCDR1、配列番号:89に記載のアミノ酸配列を有するCDR2、配列番号:90に記載のアミノ酸配列を有するCDR3を含む軽鎖を含む抗体、
(s)(q)の重鎖と(r)の軽鎖を含む抗体、
(t)(s)の抗体が認識するエピトープと同じエピトープを認識する抗体、
(u)配列番号:91に記載のアミノ酸配列を有するCDR1、配列番号:92に記載のアミノ酸配列を有するCDR2、配列番号:93に記載のアミノ酸配列を有するCDR3を含む重鎖を含む抗体、
(v)配列番号:94に記載のアミノ酸配列を有するCDR1、配列番号:95に記載のアミノ酸配列を有するCDR2、配列番号:96に記載のアミノ酸配列を有するCDR3を含む軽鎖を含む抗体、
(w)(u)の重鎖と(v)の軽鎖を含む抗体、
(x)(w)の抗体が認識するエピトープと同じエピトープを認識する抗体、
のいずれかに記載の抗体である請求項4に記載の医薬組成物。 - TGFαアンタゴニストが抗TGFα抗体である請求項1~3いずれかに記載の医薬組成物。
- 抗TGFα抗体が以下の(a)~(d);
(a)配列番号:47に記載のアミノ酸配列を有するCDR1、配列番号:48に記載のアミノ酸配列を有するCDR2、配列番号:49に記載のアミノ酸配列を有するCDR3を含む重鎖を含む抗体、
(b)配列番号:50に記載のアミノ酸配列を有するCDR1、配列番号:51に記載のアミノ酸配列を有するCDR2、配列番号:52に記載のアミノ酸配列を有するCDR3を含む軽鎖を含む抗体、
(c)(a)の重鎖と(b)の軽鎖を含む抗体、
(d)(c)の抗体が認識するエピトープと同じエピトープを認識する抗体、
のいずれかに記載の抗体である請求項6に記載の医薬組成物。 - EREGアンタゴニストが抗EREG抗体であり、TGFαアンタゴニストが抗TGFα抗体である請求項1~3いずれかに記載の医薬組成物。
- 抗EREG抗体が以下の(a)~(x);
(a)配列番号:61に記載のアミノ酸配列を有するCDR1、配列番号:62に記載のアミノ酸配列を有するCDR2、配列番号:63に記載のアミノ酸配列を有するCDR3を含む重鎖を含む抗体、
(b)配列番号:64に記載のアミノ酸配列を有するCDR1、配列番号:65に記載のアミノ酸配列を有するCDR2、配列番号:66に記載のアミノ酸配列を有するCDR3を含む軽鎖を含む抗体、
(c)(a)の重鎖と(b)の軽鎖を含む抗体、
(d)(c)の抗体が認識するエピトープと同じエピトープを認識する抗体、
(e)配列番号:67に記載のアミノ酸配列を有するCDR1、配列番号:68に記載のアミノ酸配列を有するCDR2、配列番号:69に記載のアミノ酸配列を有するCDR3を含む重鎖を含む抗体、
(f)配列番号:70に記載のアミノ酸配列を有するCDR1、配列番号:71に記載のアミノ酸配列を有するCDR2、配列番号:72に記載のアミノ酸配列を有するCDR3を含む軽鎖を含む抗体、
(g)(e)の重鎖と(f)の軽鎖を含む抗体、
(h)(g)の抗体が認識するエピトープと同じエピトープを認識する抗体、
(i)配列番号:73に記載のアミノ酸配列を有するCDR1、配列番号:74に記載のアミノ酸配列を有するCDR2、配列番号:75に記載のアミノ酸配列を有するCDR3を含む重鎖を含む抗体、
(j)配列番号:76に記載のアミノ酸配列を有するCDR1、配列番号:77に記載のアミノ酸配列を有するCDR2、配列番号:78に記載のアミノ酸配列を有するCDR3を含む軽鎖を含む抗体、
(k)(i)の重鎖と(j)の軽鎖を含む抗体、
(l)(k)の抗体が認識するエピトープと同じエピトープを認識する抗体、
(m)配列番号:79に記載のアミノ酸配列を有するCDR1、配列番号:80に記載のアミノ酸配列を有するCDR2、配列番号:81に記載のアミノ酸配列を有するCDR3を含む重鎖を含む抗体、
(n)配列番号:82に記載のアミノ酸配列を有するCDR1、配列番号:83に記載のアミノ酸配列を有するCDR2、配列番号:84に記載のアミノ酸配列を有するCDR3を含む軽鎖を含む抗体、
(o)(m)の重鎖と(n)の軽鎖を含む抗体、
(p)(o)の抗体が認識するエピトープと同じエピトープを認識する抗体、
(q)配列番号:85に記載のアミノ酸配列を有するCDR1、配列番号:86に記載のアミノ酸配列を有するCDR2、配列番号:87に記載のアミノ酸配列を有するCDR3を含む重鎖を含む抗体、
(r)配列番号:88に記載のアミノ酸配列を有するCDR1、配列番号:89に記載のアミノ酸配列を有するCDR2、配列番号:90に記載のアミノ酸配列を有するCDR3を含む軽鎖を含む抗体、
(s)(q)の重鎖と(r)の軽鎖を含む抗体、
(t)(s)の抗体が認識するエピトープと同じエピトープを認識する抗体、
(u)配列番号:91に記載のアミノ酸配列を有するCDR1、配列番号:92に記載のアミノ酸配列を有するCDR2、配列番号:93に記載のアミノ酸配列を有するCDR3を含む重鎖を含む抗体、
(v)配列番号:94に記載のアミノ酸配列を有するCDR1、配列番号:95に記載のアミノ酸配列を有するCDR2、配列番号:96に記載のアミノ酸配列を有するCDR3を含む軽鎖を含む抗体、
(w)(u)の重鎖と(v)の軽鎖を含む抗体、
(x)(w)の抗体が認識するエピトープと同じエピトープを認識する抗体、
のいずれかに記載の抗体である請求項8に記載の医薬組成物。 - 抗TGFα抗体が以下の(a)~(d)いずれか;
(a)配列番号:47に記載のアミノ酸配列を有するCDR1、配列番号:48に記載のアミノ酸配列を有するCDR2、配列番号:49に記載のアミノ酸配列を有するCDR3を含む重鎖を含む抗体、
(b)配列番号:50に記載のアミノ酸配列を有するCDR1、配列番号:51に記載のアミノ酸配列を有するCDR2、配列番号:52に記載のアミノ酸配列を有するCDR3を含む軽鎖を含む抗体、
(c)(a)の重鎖と(b)の軽鎖を含む抗体、
(d)(c)の抗体が認識するエピトープと同じエピトープを認識する抗体、
に記載の抗体である請求項8又は9に記載の医薬組成物。 - EREGおよびTGFαに結合する抗体。
- EREGおよびTGFαに対してアンタゴニスト活性を有する請求項11に記載の抗体。
- 以下の(a)~(d)いずれかに記載の抗体である請求項11又は12に記載の抗体;
(a)配列番号:10に記載のアミノ酸配列を有するCDR1、配列番号:12に記載のアミノ酸配列を有するCDR2、配列番号:14に記載のアミノ酸配列を有するCDR3を含む重鎖を含む抗体、
(b)配列番号:16に記載のアミノ酸配列を有するCDR1、配列番号:18に記載のアミノ酸配列を有するCDR2、配列番号:20に記載のアミノ酸配列を有するCDR3を含む軽鎖を含む抗体、
(c)(a)の重鎖と(b)の軽鎖を含む抗体、
(d)(c)の抗体が認識するエピトープと同じエピトープを認識する抗体。 - 配列番号:4(human EREG)の56番目のGlnから102番目のLeuまでの領域および/または配列番号:26(human TGFα)の41番目のValから87番目のLeuまでの領域を認識する抗体。
- EREGおよびTGFαに結合する物質を有効成分として含む医薬組成物。
- EREGおよびTGFαに結合する物質が請求項11~14いずれかに記載の抗体である請求項15に記載の医薬組成物。
- 以下の工程を含む抗癌剤のスクリーニング方法;
(a)被検物質のEREGに対するアンタゴニスト活性を測定する工程、
(b)被検物質のTGFαに対するアンタゴニスト活性を測定する工程、
(c)EREGおよびTGFαにアンタゴニスト活性を有する被検物質を選択する工程。
Priority Applications (4)
Application Number | Priority Date | Filing Date | Title |
---|---|---|---|
US13/375,050 US20120141501A1 (en) | 2009-05-29 | 2010-05-27 | Pharmaceutical Composition Containing Antagonist of EGF Family Ligand as Component |
EP10780608.5A EP2436397B1 (en) | 2009-05-29 | 2010-05-27 | Pharmaceutical composition containing antagonist of egf family ligand as component |
JP2011516054A JP5808052B2 (ja) | 2009-05-29 | 2010-05-27 | Egfファミリーリガンドのアンタゴニストを成分とする医薬組成物 |
US14/873,861 US10005832B2 (en) | 2009-05-29 | 2015-10-02 | Method for treating a disease originated from receptor activation by EREG and TGFα |
Applications Claiming Priority (2)
Application Number | Priority Date | Filing Date | Title |
---|---|---|---|
JP2009-131331 | 2009-05-29 | ||
JP2009131331 | 2009-05-29 |
Related Child Applications (3)
Application Number | Title | Priority Date | Filing Date |
---|---|---|---|
US13/375,050 A-371-Of-International US20120141501A1 (en) | 2009-05-29 | 2010-05-27 | Pharmaceutical Composition Containing Antagonist of EGF Family Ligand as Component |
US14/873,861 Division US10005832B2 (en) | 2009-05-29 | 2015-10-02 | Method for treating a disease originated from receptor activation by EREG and TGFα |
US14/873,861 Continuation US10005832B2 (en) | 2009-05-29 | 2015-10-02 | Method for treating a disease originated from receptor activation by EREG and TGFα |
Publications (1)
Publication Number | Publication Date |
---|---|
WO2010137654A1 true WO2010137654A1 (ja) | 2010-12-02 |
Family
ID=43222759
Family Applications (1)
Application Number | Title | Priority Date | Filing Date |
---|---|---|---|
PCT/JP2010/059008 WO2010137654A1 (ja) | 2009-05-29 | 2010-05-27 | Egfファミリーリガンドのアンタゴニストを成分とする医薬組成物 |
Country Status (4)
Country | Link |
---|---|
US (2) | US20120141501A1 (ja) |
EP (1) | EP2436397B1 (ja) |
JP (2) | JP5808052B2 (ja) |
WO (1) | WO2010137654A1 (ja) |
Cited By (6)
Publication number | Priority date | Publication date | Assignee | Title |
---|---|---|---|---|
WO2012138510A1 (en) * | 2011-04-06 | 2012-10-11 | Eli Lilly And Company | Antibodies that bind tgf-alpha and epiregulin |
US9556264B2 (en) | 2011-12-28 | 2017-01-31 | Chugai Seiyaku Kabushiki Kaisha | Humanized anti-epiregulin antibody, and cancer therapeutic agent comprising said antibody as active ingredient |
WO2018034301A1 (ja) * | 2016-08-17 | 2018-02-22 | 中外製薬株式会社 | 抗Epiregulin抗体と抗EGFR抗体との併用医薬 |
US10005832B2 (en) | 2009-05-29 | 2018-06-26 | Chugai Seiyaku Kabushiki Kaisha | Method for treating a disease originated from receptor activation by EREG and TGFα |
WO2022032003A3 (en) * | 2020-08-06 | 2022-04-07 | Abpro Corporation | Anti-claudin 18.2 antibodies and uses thereof |
US11692039B2 (en) | 2020-12-31 | 2023-07-04 | Innate Pharma | Multifunctional natural killer (NK) cell engagers binding to NKp46 and CD123 |
Families Citing this family (16)
Publication number | Priority date | Publication date | Assignee | Title |
---|---|---|---|---|
RU2537245C2 (ru) * | 2006-10-12 | 2014-12-27 | Чугаи Сейяку Кабусики Кайся | Диагностика и лечение злокачественной опухоли с использованием антитела против ereg |
JP5808054B2 (ja) | 2009-12-25 | 2015-11-10 | 中外製薬株式会社 | Nog樹立癌細胞株が移植された非ヒト動物モデルを用いた抗癌剤ターゲット探索及びスクリーニング法 |
WO2012046797A1 (ja) | 2010-10-06 | 2012-04-12 | ファーマロジカルズ・リサーチ プライベート リミテッド | 癌幹細胞集団及びその作製方法 |
EP2749641B1 (en) | 2011-09-07 | 2021-06-02 | Chugai Seiyaku Kabushiki Kaisha | Cancer stem cell isolation |
WO2013062083A1 (ja) | 2011-10-28 | 2013-05-02 | ファーマロジカルズ・リサーチ プライベート リミテッド | 癌幹細胞特異的分子 |
EP3049442A4 (en) | 2013-09-26 | 2017-06-28 | Costim Pharmaceuticals Inc. | Methods for treating hematologic cancers |
JOP20200094A1 (ar) | 2014-01-24 | 2017-06-16 | Dana Farber Cancer Inst Inc | جزيئات جسم مضاد لـ pd-1 واستخداماتها |
JOP20200096A1 (ar) | 2014-01-31 | 2017-06-16 | Children’S Medical Center Corp | جزيئات جسم مضاد لـ tim-3 واستخداماتها |
EP3148579B1 (en) | 2014-05-28 | 2020-12-16 | Agenus Inc. | Anti-gitr antibodies and methods of use thereof |
ES2771926T3 (es) | 2014-09-13 | 2020-07-07 | Novartis Ag | Terapias de combinación |
US10676723B2 (en) | 2015-05-11 | 2020-06-09 | David Gordon Bermudes | Chimeric protein toxins for expression by therapeutic bacteria |
CN106349389B (zh) * | 2015-07-21 | 2019-11-15 | 科济生物医药(上海)有限公司 | 肿瘤特异性抗egfr抗体及其应用 |
MX2018006477A (es) | 2015-12-02 | 2018-09-03 | Agenus Inc | Anticuerpos y metodos de uso de estos. |
WO2022104127A1 (en) * | 2020-11-13 | 2022-05-19 | SAB Biotherapeutics, Inc. | Ungulate-derived polyclonal immunoglobulin specific for egfr and uses thereof |
CN116333116B (zh) * | 2021-12-16 | 2024-03-05 | 徕特康(苏州)生物制药有限公司 | 抗表皮生长因子受体抗体及其制备方法和用途 |
EP4324846A1 (en) * | 2022-08-16 | 2024-02-21 | Eberhard Karls Universität Tübingen, Medizinische Fakultät | Inhibitor protein of ligands of epidermal growth factor receptor (egfr) |
Citations (29)
Publication number | Priority date | Publication date | Assignee | Title |
---|---|---|---|---|
EP0239400A2 (en) | 1986-03-27 | 1987-09-30 | Medical Research Council | Recombinant antibodies and methods for their production |
JPH0159878B2 (ja) | 1982-05-21 | 1989-12-20 | Yunibaashitei Obu Karifuorunia | |
EP0404097A2 (de) | 1989-06-22 | 1990-12-27 | BEHRINGWERKE Aktiengesellschaft | Bispezifische und oligospezifische, mono- und oligovalente Rezeptoren, ihre Herstellung und Verwendung |
WO1992001047A1 (en) | 1990-07-10 | 1992-01-23 | Cambridge Antibody Technology Limited | Methods for producing members of specific binding pairs |
WO1992003918A1 (en) | 1990-08-29 | 1992-03-19 | Genpharm International, Inc. | Transgenic non-human animals capable of producing heterologous antibodies |
WO1992020791A1 (en) | 1990-07-10 | 1992-11-26 | Cambridge Antibody Technology Limited | Methods for producing members of specific binding pairs |
WO1993006213A1 (en) | 1991-09-23 | 1993-04-01 | Medical Research Council | Production of chimeric antibodies - a combinatorial approach |
WO1993011236A1 (en) | 1991-12-02 | 1993-06-10 | Medical Research Council | Production of anti-self antibodies from antibody segment repertoires and displayed on phage |
WO1993011161A1 (en) | 1991-11-25 | 1993-06-10 | Enzon, Inc. | Multivalent antigen-binding proteins |
WO1993012227A1 (en) | 1991-12-17 | 1993-06-24 | Genpharm International, Inc. | Transgenic non-human animals capable of producing heterologous antibodies |
WO1993019172A1 (en) | 1992-03-24 | 1993-09-30 | Cambridge Antibody Technology Limited | Methods for producing members of specific binding pairs |
WO1994002602A1 (en) | 1992-07-24 | 1994-02-03 | Cell Genesys, Inc. | Generation of xenogeneic antibodies |
WO1994011523A2 (en) | 1992-11-13 | 1994-05-26 | Idec Pharmaceuticals Corporation | Fully impaired consensus kozac sequences for mammalian expression |
WO1994025585A1 (en) | 1993-04-26 | 1994-11-10 | Genpharm International, Inc. | Transgenic non-human animals capable of producing heterologous antibodies |
WO1995001438A1 (en) | 1993-06-30 | 1995-01-12 | Medical Research Council | Sbp members with a chemical moiety covalently bound within the binding site; production and selection thereof |
WO1995015388A1 (en) | 1993-12-03 | 1995-06-08 | Medical Research Council | Recombinant binding proteins and peptides |
WO1996002576A1 (fr) | 1994-07-13 | 1996-02-01 | Chugai Seiyaku Kabushiki Kaisha | Anticorps humain reconstitue contre l'interleukine-8 humaine |
WO1996033735A1 (en) | 1995-04-27 | 1996-10-31 | Abgenix, Inc. | Human antibodies derived from immunized xenomice |
WO1996034096A1 (en) | 1995-04-28 | 1996-10-31 | Abgenix, Inc. | Human antibodies derived from immunized xenomice |
WO1999054342A1 (en) | 1998-04-20 | 1999-10-28 | Pablo Umana | Glycosylation engineering of antibodies for improving antibody-dependent cellular cytotoxicity |
WO2000061739A1 (en) | 1999-04-09 | 2000-10-19 | Kyowa Hakko Kogyo Co., Ltd. | Method for controlling the activity of immunologically functional molecule |
WO2002031140A1 (fr) | 2000-10-06 | 2002-04-18 | Kyowa Hakko Kogyo Co., Ltd. | Cellules produisant des compositions d'anticorps |
WO2002079255A1 (en) | 2001-04-02 | 2002-10-10 | Idec Pharmaceuticals Corporation | RECOMBINANT ANTIBODIES COEXPRESSED WITH GnTIII |
WO2003104453A1 (ja) | 2002-06-05 | 2003-12-18 | 中外製薬株式会社 | 抗体作製方法 |
WO2004076622A2 (en) | 2003-02-10 | 2004-09-10 | National Institute Of Advanced Industrial Science And Technology | Regulation of gene expression by dna interference |
WO2007015578A1 (ja) * | 2005-08-02 | 2007-02-08 | Eisai R & D Management Co., Ltd. | 血管新生阻害物質の効果を検定する方法 |
WO2008047723A1 (en) | 2006-10-12 | 2008-04-24 | Forerunner Pharma Research Co., Ltd. | Diagnosis and treatment of cancer using anti-ereg antibody |
WO2008047914A1 (fr) | 2006-10-20 | 2008-04-24 | Forerunner Pharma Research Co., Ltd. | Agent anticancéreux comprenant un anticorps anti-hb-egf en tant qu'ingrédient actif |
JP2008527978A (ja) * | 2005-01-11 | 2008-07-31 | モレキュラー ロジックス,インコーポレイテッド | Pan−HERアンタゴニストおよび使用方法 |
Family Cites Families (30)
Publication number | Priority date | Publication date | Assignee | Title |
---|---|---|---|---|
US5190858A (en) * | 1989-02-01 | 1993-03-02 | Oncogene Science, Inc. | Monoclonal antibodies directed to epitopes of human transforming growth factor--α and uses thereof |
US6800738B1 (en) | 1991-06-14 | 2004-10-05 | Genentech, Inc. | Method for making humanized antibodies |
WO1994029340A1 (fr) | 1993-06-04 | 1994-12-22 | Taisho Pharmaceutical Co., Ltd. | Facteur d'inhibition de la proliferation de cellules tumorales d'origine humaine |
US5994511A (en) | 1997-07-02 | 1999-11-30 | Genentech, Inc. | Anti-IgE antibodies and methods of improving polypeptides |
US6172213B1 (en) | 1997-07-02 | 2001-01-09 | Genentech, Inc. | Anti-IgE antibodies and method of improving polypeptides |
TR200002885T2 (tr) | 1998-04-03 | 2000-12-21 | Chugai Seiyaku Kabushiki Kaisha | İnsan doku faktörüne (TF) karşı insanlaştırılmış antikor |
US6852318B1 (en) | 1998-05-08 | 2005-02-08 | The Regents Of The University Of California | Methods for detecting and inhibiting angiogenesis |
US6949245B1 (en) | 1999-06-25 | 2005-09-27 | Genentech, Inc. | Humanized anti-ErbB2 antibodies and treatment with anti-ErbB2 antibodies |
MXPA01013458A (es) | 1999-06-25 | 2002-07-30 | Genentech Inc | Anticuerpos humanizados anti-erbb2 y tratamiento con anticuerpos anti-erbb2. |
US6946292B2 (en) | 2000-10-06 | 2005-09-20 | Kyowa Hakko Kogyo Co., Ltd. | Cells producing antibody compositions with increased antibody dependent cytotoxic activity |
CA2427858A1 (en) | 2000-11-03 | 2002-05-10 | University Of Vermont And State Agricultural College | Compositions for inhibiting grb7 |
CU22979A1 (es) | 2000-12-08 | 2004-09-09 | Centro Inmunologia Molecular | Combinación inmunoterapéutica para el tratamiento de tumores que sobre-expresan receptores con actividad quinasa en residuos de tirosina |
AU2002362197A1 (en) | 2001-12-28 | 2003-07-24 | Chugai Seiyaku Kabushiki Kaisha | Method of stabilizing protein |
ES2263984T3 (es) | 2002-06-28 | 2006-12-16 | Domantis Limited | Ligandos doble-especificos con una vida media serica aumentada. |
US7393531B2 (en) | 2003-01-21 | 2008-07-01 | Arius Research Inc. | Cytotoxicity mediation of cells evidencing surface expression of MCSP |
EP2264151B1 (en) | 2003-01-22 | 2016-04-20 | Roche Glycart AG | Fusion constructs and use of same to produce antibodies with increased FC receptor binding affinity and effector function |
CN1761682A (zh) | 2003-03-14 | 2006-04-19 | 大正制药株式会社 | 单克隆抗体与产生该单克隆抗体的杂交瘤 |
US7195764B2 (en) | 2003-04-14 | 2007-03-27 | Arius Research Inc. | Cancerous disease modifying antibodies |
DK1704166T3 (en) | 2004-01-07 | 2015-06-01 | Novartis Vaccines & Diagnostic | M-CSF-SPECIFIC MONOCLONAL ANTIBODY AND APPLICATIONS THEREOF |
CA2555145A1 (en) | 2004-02-06 | 2005-08-25 | Wyeth | Diagnosis and therapeutics for cancer |
EP2208783A1 (en) | 2004-12-22 | 2010-07-21 | Chugai Seiyaku Kabushiki Kaisha | Method of producing an antibody using a cell in which the function of fucose transporter is inhibited |
US20090318346A1 (en) * | 2006-02-08 | 2009-12-24 | Targeted Molecular Diagnostics | BIVALENT ErbB LIGAND BINDING MOLECULES AND METHODS FOR THEIR PREPARATION AND USE |
US20120141501A1 (en) | 2009-05-29 | 2012-06-07 | Forerunner Pharma Research Co. Ltd | Pharmaceutical Composition Containing Antagonist of EGF Family Ligand as Component |
GB0909904D0 (en) | 2009-06-09 | 2009-07-22 | Affitech As | Product |
GB0909906D0 (en) | 2009-06-09 | 2009-07-22 | Affitech As | Antibodies |
CN103827300A (zh) | 2011-06-30 | 2014-05-28 | 中外制药株式会社 | 异源二聚化多肽 |
WO2013062083A1 (ja) | 2011-10-28 | 2013-05-02 | ファーマロジカルズ・リサーチ プライベート リミテッド | 癌幹細胞特異的分子 |
TWI593705B (zh) | 2011-12-28 | 2017-08-01 | Chugai Pharmaceutical Co Ltd | Humanized anti-epiregulin antibody and cancer therapeutic agent containing the antibody as an active ingredient |
JP6433297B2 (ja) | 2012-12-27 | 2018-12-05 | 中外製薬株式会社 | ヘテロ二量化ポリペプチド |
RU2016101716A (ru) | 2013-06-24 | 2017-07-27 | Чугаи Сейяку Кабусики Кайся | Терапевтический агент, содержащий гуманизированное антиэпирегулиновое антитело в качестве действующего ингредиента для немелкоклеточной карциномы легкого, за исключением аденокарциномы |
-
2010
- 2010-05-27 US US13/375,050 patent/US20120141501A1/en not_active Abandoned
- 2010-05-27 EP EP10780608.5A patent/EP2436397B1/en not_active Not-in-force
- 2010-05-27 WO PCT/JP2010/059008 patent/WO2010137654A1/ja active Application Filing
- 2010-05-27 JP JP2011516054A patent/JP5808052B2/ja not_active Expired - Fee Related
-
2015
- 2015-04-06 JP JP2015077677A patent/JP6025904B2/ja not_active Expired - Fee Related
- 2015-10-02 US US14/873,861 patent/US10005832B2/en not_active Expired - Fee Related
Patent Citations (29)
Publication number | Priority date | Publication date | Assignee | Title |
---|---|---|---|---|
JPH0159878B2 (ja) | 1982-05-21 | 1989-12-20 | Yunibaashitei Obu Karifuorunia | |
EP0239400A2 (en) | 1986-03-27 | 1987-09-30 | Medical Research Council | Recombinant antibodies and methods for their production |
EP0404097A2 (de) | 1989-06-22 | 1990-12-27 | BEHRINGWERKE Aktiengesellschaft | Bispezifische und oligospezifische, mono- und oligovalente Rezeptoren, ihre Herstellung und Verwendung |
WO1992001047A1 (en) | 1990-07-10 | 1992-01-23 | Cambridge Antibody Technology Limited | Methods for producing members of specific binding pairs |
WO1992020791A1 (en) | 1990-07-10 | 1992-11-26 | Cambridge Antibody Technology Limited | Methods for producing members of specific binding pairs |
WO1992003918A1 (en) | 1990-08-29 | 1992-03-19 | Genpharm International, Inc. | Transgenic non-human animals capable of producing heterologous antibodies |
WO1993006213A1 (en) | 1991-09-23 | 1993-04-01 | Medical Research Council | Production of chimeric antibodies - a combinatorial approach |
WO1993011161A1 (en) | 1991-11-25 | 1993-06-10 | Enzon, Inc. | Multivalent antigen-binding proteins |
WO1993011236A1 (en) | 1991-12-02 | 1993-06-10 | Medical Research Council | Production of anti-self antibodies from antibody segment repertoires and displayed on phage |
WO1993012227A1 (en) | 1991-12-17 | 1993-06-24 | Genpharm International, Inc. | Transgenic non-human animals capable of producing heterologous antibodies |
WO1993019172A1 (en) | 1992-03-24 | 1993-09-30 | Cambridge Antibody Technology Limited | Methods for producing members of specific binding pairs |
WO1994002602A1 (en) | 1992-07-24 | 1994-02-03 | Cell Genesys, Inc. | Generation of xenogeneic antibodies |
WO1994011523A2 (en) | 1992-11-13 | 1994-05-26 | Idec Pharmaceuticals Corporation | Fully impaired consensus kozac sequences for mammalian expression |
WO1994025585A1 (en) | 1993-04-26 | 1994-11-10 | Genpharm International, Inc. | Transgenic non-human animals capable of producing heterologous antibodies |
WO1995001438A1 (en) | 1993-06-30 | 1995-01-12 | Medical Research Council | Sbp members with a chemical moiety covalently bound within the binding site; production and selection thereof |
WO1995015388A1 (en) | 1993-12-03 | 1995-06-08 | Medical Research Council | Recombinant binding proteins and peptides |
WO1996002576A1 (fr) | 1994-07-13 | 1996-02-01 | Chugai Seiyaku Kabushiki Kaisha | Anticorps humain reconstitue contre l'interleukine-8 humaine |
WO1996033735A1 (en) | 1995-04-27 | 1996-10-31 | Abgenix, Inc. | Human antibodies derived from immunized xenomice |
WO1996034096A1 (en) | 1995-04-28 | 1996-10-31 | Abgenix, Inc. | Human antibodies derived from immunized xenomice |
WO1999054342A1 (en) | 1998-04-20 | 1999-10-28 | Pablo Umana | Glycosylation engineering of antibodies for improving antibody-dependent cellular cytotoxicity |
WO2000061739A1 (en) | 1999-04-09 | 2000-10-19 | Kyowa Hakko Kogyo Co., Ltd. | Method for controlling the activity of immunologically functional molecule |
WO2002031140A1 (fr) | 2000-10-06 | 2002-04-18 | Kyowa Hakko Kogyo Co., Ltd. | Cellules produisant des compositions d'anticorps |
WO2002079255A1 (en) | 2001-04-02 | 2002-10-10 | Idec Pharmaceuticals Corporation | RECOMBINANT ANTIBODIES COEXPRESSED WITH GnTIII |
WO2003104453A1 (ja) | 2002-06-05 | 2003-12-18 | 中外製薬株式会社 | 抗体作製方法 |
WO2004076622A2 (en) | 2003-02-10 | 2004-09-10 | National Institute Of Advanced Industrial Science And Technology | Regulation of gene expression by dna interference |
JP2008527978A (ja) * | 2005-01-11 | 2008-07-31 | モレキュラー ロジックス,インコーポレイテッド | Pan−HERアンタゴニストおよび使用方法 |
WO2007015578A1 (ja) * | 2005-08-02 | 2007-02-08 | Eisai R & D Management Co., Ltd. | 血管新生阻害物質の効果を検定する方法 |
WO2008047723A1 (en) | 2006-10-12 | 2008-04-24 | Forerunner Pharma Research Co., Ltd. | Diagnosis and treatment of cancer using anti-ereg antibody |
WO2008047914A1 (fr) | 2006-10-20 | 2008-04-24 | Forerunner Pharma Research Co., Ltd. | Agent anticancéreux comprenant un anticorps anti-hb-egf en tant qu'ingrédient actif |
Non-Patent Citations (61)
Title |
---|
"Antibodies A Laboratory Manual.", 1988, COLD SPRING HARBOR LABORATORY |
"Immunologic studies in humans", 1993, JOHN WILEY & SONS, INC., article "Current Protocols in Immunology" |
"Remington's Pharmaceutical Science", MARK PUBLISHING COMPANY |
BABA, I. ET AL., CANCER RES., vol. 60, 2000, pages 6886 - 6889 |
BELYAVSKY, A., NUCLEIC ACIDS RES., vol. 17, 1989, pages 2919 - 2932 |
BETTER ET AL., SCIENCE, vol. 240, 1988, pages 1041 - 1043 |
BETTER, M.; HORWITZ, A. H., METHODS ENZYMOL., vol. 178, 1989, pages 476 - 496 |
BETTER, M.; HORWITZ, A. H., METHODS IN ENZYMOLOGY, vol. 178, 1989, pages 476 - 496 |
BIRD, R. E. ET AL., TIBTECH, vol. 9, 1991, pages 132 - 137 |
BIRD, R. E.; WALKER, B. W., TRENDS BIOTECHNOL., vol. 9, 1991, pages 132 - 137 |
CHIRGWIN, J. M. ET AL., BIOCHEMISTRY, vol. 18, 1979, pages 5294 - 5299 |
CHOMCZYNSKI, P. ET AL., ANAL. BIOCHEM., vol. 162, 1987, pages 156 - 159 |
CIARDIELLO ET AL., CLIN CANCER RES, vol. 5, 1999, pages 909 - 916 |
CO, M. S. ET AL., J. IMMUNOL., vol. 152, 1994, pages 2968 - 2976 |
CO, M. S., J. IMMUNOL., vol. 152, 1994, pages 2968 - 2976 |
CURRENT TOPICS IN MICROBIOLOGY AND IMMUNOLOGY, vol. 81, 1978, pages 1 - 7 |
DE ST. GROTH, S. F., J. IMMUNOL. METHODS, vol. 35, 1980, pages 1 - 21 |
DEAN C ET AL.: "Immunotherapy with antibodies to the EGF receptor", INTERNATIONAL JOURNAL OF CANCER, vol. 57, no. SUPPL., 1994, pages 103 - 107, XP008150875 * |
EBERT, K. M. ET AL., BIO/TECHNOLOGY, vol. 12, 1994, pages 699 - 702 |
FASEB J., vol. 6, 1992, pages 2422 - 2427 |
FORCE, T. ET AL., NAT. REV. CANCER, vol. 7, 2007, pages 332 - 344 |
FROHMAN, M. A. ET AL., PROC. NATL. ACAD. SCI. USA, vol. 85, 1988, pages 8998 - 9002 |
GALFRE, G., NATURE, vol. 277, 1979, pages 131 - 133 |
GROENESTEGE, W. M. ET AL., J. CLIN. INVEST., vol. 117, 2007, pages 2260 - 2267 |
HIGASHIYAMA, S. ET AL., CANCER SCI., vol. 99, 2008, pages 214 - 220 |
HOLLINGER P. ET AL., PROC. NATL. ACAD. SCI. USA, vol. 90, 1993, pages 6444 - 6448 |
HUDSON ET AL., J. HNMUNOL. METHODS, vol. 231, 1999, pages 177 - 189 |
HUSTON, J. S. ET AL., PROC. NATL. ACAD. SCI. U.S.A., vol. 85, 1988, pages 5879 - 5883 |
ITO M ET AL.: "Expression of several growth factors and their receptor genes in human colon carcinomas", VIRCHOWS ARCHIV. B, CELL PATHOLOGY INCLUDING MOLECULAR PATHOLOGY, vol. 59, no. 3, 1990, pages 173 - 178, XP009015191 * |
J. IMMUNOL., vol. 123, 1979, pages 1548 - 1550 |
JOHNSON, G. R. ET AL., J. CELL BIOL., vol. 118, 1992, pages 741 - 751 |
KOHLER. G.; MILSTEIN, C., EUR. J. IMMUNOL., vol. 6, 1976, pages 511 - 519 |
KOHLER. G.; MILSTEIN, C., METHODS ENZYMOL., vol. 73, 1981, pages 3 - 46 |
LACOUTURE, M. E., NAT. REV. CANCER, vol. 6, 2006, pages 803 - 812 |
LAMOYI, E., METHODS ENZYMOL., vol. 121, 1986, pages 652 - 663 |
LAMOYI, E., METHODS IN ENZYMOLOGY, vol. 121, 1989, pages 652 - 663 |
LEI, S. P. ET AL., J. BACTERIOL., vol. 169, 1987, pages 4379 |
LU Y ET AL.: "Immunogene Therapy of Tumors with Vaccine Based on Xenogeneic Epidermal Growth Factor Receptor", THE JOURNAL OF IMMUNOLOGY, vol. 170, no. 6, 2003, pages 3162 - 3170, XP002384263 * |
MARGULIES. D.H., CELL, vol. 8, 1976, pages 405 - 415 |
MIZUSHIMA ET AL., NUCLEIC ACIDS RES., vol. 18, 1990, pages 5322 |
MODJTAHEDI H ET AL.: "Anti-EGFR monoclonal antibodies which act as EGF, TGFa, HB-EGF and BTC antagonists block the binding of epiregulin to EGFR-expressing tumours", INTERNATIONAL JOURNAL OF CANCER, vol. 75, no. 2, 1998, pages 310 - 316, XP002492891 * |
MULLIGAN ET AL., NATURE, vol. 277, 1979, pages 108 |
PLUCKTHUN, A.; SKERRA, A., METHODS ENZYMOL., vol. 178, 1989, pages 497 - 515 |
PLUECKTHUN, A.; SKERRA, A., METHODS IN ENZYMOLOGY, vol. 178, 1989, pages 476 - 496 |
PROTEIN ENGINEERING, vol. 9, no. 3, 1996, pages 299 - 305 |
QIAN JF ET AL.: "Human transforming growth factor alpha: sequence analysis of the 4.5-kb and 1.6-kb mRNA species", GENE, vol. 132, no. 2, 1993, pages 291 - 296, XP023541751 * |
ROUSSEAUX, J. ET AL., METHODS ENZYMOL., vol. 121, 1986, pages 663 - 669 |
ROUSSEAUX, J. ET AL., METHODS IN ENZYMOLOGY, vol. 121, 1989, pages 663 - 669 |
SAMBROOK, J. ET AL.: "Molecular Cloning", 1989, COLD SPRING HARBOR LAB. PRESS, pages: 9.47 - 9.58 |
SATO, K. ET AL., CANCER RES., vol. 53, 1993, pages 851 - 856 |
SCHNEIDER, M. R.; WOLF, E., J. CELL. PHYSIOL., vol. 218, 2009, pages 460 - 466 |
See also references of EP2436397A4 * |
SETH, D. ET AL., BR. J. CANCER, vol. 80, 1999, pages 657 - 669 |
SHULMAN, M. ET AL., NATURE, vol. 276, 1978, pages 269 - 270 |
TEJPAR, S. ET AL., LANCET ONCOL., vol. 8, 2007, pages 387 - 394 |
TROWBRIDGE, 1. S., J. EXP. MED., vol. 148, 1978, pages 313 - 323 |
VANDAMME, A. M. ET AL., EUR. J. BIOCHEM., vol. 192, 1990, pages 767 - 775 |
WARD, NATURE, vol. 341, 1989, pages 544 - 546 |
WILBUR, W. J.; LIPMAN, D. J., PROC. NATL. ACAD. SCI. USA, vol. 80, 1983, pages 726 - 730 |
WILLMARTH, N. E. ET AL., J. BIOL. CHEM., vol. 281, 2006, pages 37728 - 37737 |
WILSON, K.J. ET AL., PHARMACOL. THER., vol. 122, 2009, pages 1 - 8 |
Cited By (14)
Publication number | Priority date | Publication date | Assignee | Title |
---|---|---|---|---|
US10005832B2 (en) | 2009-05-29 | 2018-06-26 | Chugai Seiyaku Kabushiki Kaisha | Method for treating a disease originated from receptor activation by EREG and TGFα |
KR101581517B1 (ko) | 2011-04-06 | 2015-12-30 | 일라이 릴리 앤드 캄파니 | Tgf-알파 및 에피레굴린에 결합하는 항체 |
CN103459423A (zh) * | 2011-04-06 | 2013-12-18 | 伊莱利利公司 | 结合TGF-α和EPIREGULIN的抗体 |
US8613921B2 (en) | 2011-04-06 | 2013-12-24 | Eli Lilly And Company | Antibodies that bind TGF-alpha and epiregulin |
JP2014515743A (ja) * | 2011-04-06 | 2014-07-03 | イーライ リリー アンド カンパニー | Tgf−アルファおよびエピレグリンに結合する抗体 |
TWI454481B (zh) * | 2011-04-06 | 2014-10-01 | Lilly Co Eli | 結合TGF-α及表皮調節素(EPIREGULIN)之抗體 |
WO2012138510A1 (en) * | 2011-04-06 | 2012-10-11 | Eli Lilly And Company | Antibodies that bind tgf-alpha and epiregulin |
CN103459423B (zh) * | 2011-04-06 | 2016-03-02 | 伊莱利利公司 | 结合TGF-α和EPIREGULIN的抗体 |
EA024921B1 (ru) * | 2011-04-06 | 2016-11-30 | Эли Лилли Энд Компани | АНТИТЕЛА, КОТОРЫЕ СВЯЗЫВАЮТ ФАКТОР РОСТА ОПУХОЛИ-α И ЭПИРЕГУЛИН |
KR20130133274A (ko) * | 2011-04-06 | 2013-12-06 | 일라이 릴리 앤드 캄파니 | Tgf-알파 및 에피레굴린에 결합하는 항체 |
US9556264B2 (en) | 2011-12-28 | 2017-01-31 | Chugai Seiyaku Kabushiki Kaisha | Humanized anti-epiregulin antibody, and cancer therapeutic agent comprising said antibody as active ingredient |
WO2018034301A1 (ja) * | 2016-08-17 | 2018-02-22 | 中外製薬株式会社 | 抗Epiregulin抗体と抗EGFR抗体との併用医薬 |
WO2022032003A3 (en) * | 2020-08-06 | 2022-04-07 | Abpro Corporation | Anti-claudin 18.2 antibodies and uses thereof |
US11692039B2 (en) | 2020-12-31 | 2023-07-04 | Innate Pharma | Multifunctional natural killer (NK) cell engagers binding to NKp46 and CD123 |
Also Published As
Publication number | Publication date |
---|---|
US20120141501A1 (en) | 2012-06-07 |
JP6025904B2 (ja) | 2016-11-16 |
JP2015155431A (ja) | 2015-08-27 |
EP2436397B1 (en) | 2017-05-10 |
JPWO2010137654A1 (ja) | 2012-11-15 |
EP2436397A1 (en) | 2012-04-04 |
US20160017028A1 (en) | 2016-01-21 |
US10005832B2 (en) | 2018-06-26 |
JP5808052B2 (ja) | 2015-11-10 |
EP2436397A4 (en) | 2014-02-19 |
Similar Documents
Publication | Publication Date | Title |
---|---|---|
JP6025904B2 (ja) | Egfファミリーリガンドのアンタゴニストを成分とする医薬組成物 | |
CA2855699C (en) | Anti-human trop-2 antibody having an antitumor activity in vivo | |
KR101568051B1 (ko) | Anexelekto에 결합하는 단일클론항체, 및 그의 이용 | |
JP5918540B2 (ja) | 抗dll3抗体 | |
CA2798778C (en) | Anti-human trop-2 antibody having anti-tumor activity in vivo | |
JP5378795B2 (ja) | 抗hb−egf抗体を有効成分として含む医薬組成物 | |
JP5788384B2 (ja) | 形質転換増殖因子アルファに結合し、Ras遺伝子変異癌に対して増殖抑制活性を有する抗体 | |
JPWO2008114733A1 (ja) | 抗Claudin−4抗体 | |
WO2011021381A1 (ja) | 抗hb-egf抗体を有効成分として含む医薬組成物 | |
WO2011105573A1 (ja) | 抗icam3抗体およびその用途 | |
JP5746018B2 (ja) | 抗tmprss11e抗体を用いた癌の診断と治療 | |
EP2990418B1 (en) | Functional monoclonal antibody against heparin-binding epidermal growth factor-like growth factor | |
WO2011052753A1 (ja) | Mansc1蛋白質に結合し、抗癌活性を有する抗体 | |
JP5838427B2 (ja) | ヘパリン結合上皮増殖因子様増殖因子に対する機能性モノクローナル抗体 | |
JP5806935B2 (ja) | 抗hb−egf抗体を有効成分として含む医薬組成物 |
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: 10780608 Country of ref document: EP Kind code of ref document: A1 |
|
ENP | Entry into the national phase |
Ref document number: 2011516054 Country of ref document: JP Kind code of ref document: A |
|
NENP | Non-entry into the national phase |
Ref country code: DE |
|
REEP | Request for entry into the european phase |
Ref document number: 2010780608 Country of ref document: EP |
|
WWE | Wipo information: entry into national phase |
Ref document number: 2010780608 Country of ref document: EP |
|
WWE | Wipo information: entry into national phase |
Ref document number: 13375050 Country of ref document: US |