![]() Blocking Immune Response to a Foreign Antigen Using an Antagonist Which Binds to CD20
专利摘要:
The present invention relates to a method of blocking an immune response to foreign antigens in a mammal using an antagonist that binds to CD20. 公开号:KR20020027490A 申请号:KR1020027000481 申请日:2000-07-10 公开日:2002-04-13 发明作者:안토니오 제이. 그릴로-로페즈;로리 에이. 쿤켈;티모시 에이. 스튜어트 申请人:제넨테크, 인크.;울콧 케네쓰 제이.;아이덱 파마슈티칼즈 인코포레이티드; IPC主号:
专利说明:
Blocking Immune Response to a Foreign Antigen Using an Antagonist Which Binds to CD20} [2] Lymphocytes are one of the many types of white blood cells produced in the bone marrow during the hematopoietic response. There are two main groups of lymphocytes: B lymphocytes (B cells) and T lymphocytes (T cells). Lymphocytes of particular interest herein are B cells. [3] B cells mature in the bone marrow and leave the bone marrow to express antigen binding antibodies on their cell surface. When an inexperienced B cell first encounters an antigen specific for its membrane-bound antibody, the cell begins to divide rapidly and its progeny differentiate into memory B cells and functional cells called "plasma cells". Memory B cells have a longer lifespan and continue to express membrane-bound antibodies with the same specificity as the original parent cell. Plasma cells do not produce membrane bound antibodies but instead produce a form of antibody that can be secreted. Secretory antibodies are the major functional molecules of humoral immunity. [4] CD20 antigen (also known as Bp35, a differentiation antigen restricted to human B-lymphocytes) is a hydrophobic transmembrane protein with a molecular weight of about 35 kD located on pre-B lymphocytes and mature B lymphocytes (Valentine et al. J. Biol). Chem. 264 (19): 11282-11287 (1989); and Einfeld et al. EMBO J. 7 (3): 711-717 (1988)). This antigen is also expressed in over 90% of B cell non-Hodgkin's lymphomas (NHL) (Anderson et al. Blood 63 (6): 1424-1433 (1984)), but hematopoietic stem cells, pre- It is not present in B cells, normal plasma cells or other normal tissues (Tedder et al. J Immunol. 135 (2): 973-979 (1985)). CD20 regulates the early stages of the activation of cell cycle initiation and differentiation (Tedder et al., Supra) and may act as a calcium ion channel (Tedder et al., J Cell. Biochem. 14D: 195 (1990) ). [5] In expression of CD20 in B cell lymphomas, this antigen may serve as a candidate for "targeting" such lymphomas. In essence, this targeting can be generalized as follows: An antibody specific for the CD20 surface antigen of B cells is administered to the patient. This anti-CD20 antibody binds specifically to the CD20 antigen of both normal and malignant B cells (surface), and the antibody bound to the CD20 surface antigen can lead to the destruction and killing of tumorous B cells. In addition, chemicals or radiolabels that have the potential to destroy tumors can be linked to anti-CD20 antibodies so that they are specifically "delivered" to neoplastic B cells. Regardless of this study, the main goal is to kill tumors and the specific method can be determined by the specific anti-CD20 antibody used, and therefore the available methods of targeting CD20 antigens can vary considerably. [6] Rituximab (RITUXAN®) antibodies are genetically engineered chimeric murine / human monoclonal antibodies directed against the CD20 antigen. Rituximab is an antibody called "C2B8" in US Pat. No. 5,736,137 to Anderson et al., Issued April 7, 1998. Rituxan (RITUXAN®) is indicated for the treatment of patients with recurrent or refractory low or vesicles of CD20 positive B cell non-Hodgkin's lymphoma. In vitro mechanism of action studies have demonstrated that Rituxan (RITUXAN®) binds to human complement to lyse lymphatic B cell lines via complement-dependent cytotoxicity (CDC) (Reff et al. Blood 83 (2). ): 435-445 (1994)). Rituxan also shows significant activity in cytotoxicity (ADCC) analysis of antibody-dependent cells. More recently, it has been found that Rituxan (RITUXAN®) has anti-proliferative effects and directly induces apoptosis in tritiated thymidine incorporation assays, while other anti-CD20 antibodies do not (Maloney et al. Blood 88). (10): 637a (1996)). In addition, synergism between Rituxan (RITUXAN®) and chemotherapy and toxins was observed experimentally. In particular, Rituxan (RITUXAN®) sensitizes human B cell lymphoma cell lines that are drug-resistant to the cytotoxic effects of doxorubicin, CDDP, VP-16, diphtheria toxin and lysine (Demidem et al. Cancer Chemotherapy & Radiopharmaceuticals 12 (3): 177-186 (1997). In vivo preclinical studies have shown that Rituxan (RITUXAN®) kills B cells, possibly through complement and cell-mediated responses, from peripheral blood flow, lymph nodes and bone marrow of cynomolgus monkeys (Reff et al. Blood 83 (2): 435-445 (1994)). [7] <Overview of invention> [8] In a first aspect, the present invention provides a method of blocking an immune response to foreign antigens in a mammal comprising administering a therapeutically effective amount of an antagonist that binds CD20 to a mammal not suffering from a malignant tumor. [9] In another aspect, the invention comprises administering to a mammal a therapeutic agent other than an antagonist that binds to CD20 and an antagonist that binds to CD20, wherein the therapeutic agent is immunogenic in the mammal and the antagonist is in the mammal. It provides a method of treating a mammal, which blocks the immune response to. [10] The present invention further provides a method of treating a graft-versus-host or host-versus-graft disease in a mammal comprising administering to the mammal a therapeutically effective amount of an antagonist that binds to CD20. [11] The present invention also provides a method of insensitizing a mammal comprising administering a therapeutically effective amount of an antagonist that binds to CD20 to a mammal to be implanted. [12] The invention also relates to a product used in the above methods. For example, the product may comprise a container, a composition comprising an antagonist that binds to CD20, contained in the container, and a package insert instructing the patient to be exposed to or exposed to a foreign antigen using the composition. have. The product optionally further comprises a second container and a second composition comprising a therapeutic agent contained in the container. [1] The present invention relates to a method of blocking an immune response to foreign antigens in a mammal using an antagonist that binds to CD20. [13] I. Definition [14] The “CD20” antigen is a non-glycosylated phosphoprotein of about 35 kDa present on the surface of more than 90% of B cells from peripheral blood flow or lymphoid organs. CD20 is expressed during the early development of pre-B cells and remains until the differentiation of plasma cells. CD20 is present in both normal B cells as well as malignant B cells. In the literature it is recorded under the different names "antigen restricted to B-lymphocytes" and "Bp35" for CD20. CD20 antigens are described, for example, in Clark et al. PNAS (USA) 82: 1766 (1985). [15] "Exogenous antigen" means a molecule or molecules that are not endogenous or natural, to which a mammal is exposed. Foreign antigens can induce an immune response, such as a humoral response and / or a T cell mediated response, in a mammal. In general, foreign antigens will promote the production of antibodies against them. Examples of foreign antigens contemplated herein include proteins such as immunotherapeutic agents such as antibodies, in particular antibodies comprising non-human amino acid residues (eg, rodent, chimeric / humanized and primate antibodies); Toxins (optionally linked to targeting molecules, which may be immunogenic, such as antibodies, etc.); Gene therapy viral vectors such as retroviruses and adenoviruses; Grafts; Infectious agents (eg, bacteria and viruses); Homologous antigens (ie antigens that are present in some but not in other members of the same species) such as differences in blood type, human lymphocyte antigens (HLA), platelet antigens, antigens expressed in transplanted organs, blood components, pregnancy (Rh), and hemophilia factors (eg, factor VIII and factor IX). [16] By “blocking an immune response” to a foreign antigen is meant reducing or preventing one or more immune mediated responses resulting from exposure to the foreign antigen. For example, in mammals, humoral responses to foreign antigens can be reduced by preventing or reducing the production of antibodies directed against the antigen. Alternatively or additionally, one may inhibit the idiotype, "mitigate" the removal of cells encoded with homologous antibodies, and / or affect the presentation of homologous antigens through the killing of antigen-presenting cells. [17] The mammal to be treated herein is generally an animal that does not suffer from "malignant tumors" and therefore malignant tumors or cancers such as B cell lymphoma, acute lymphoblastic leukemia (ALL), chronic lymphocytic leukemia (CLL), blast cells Animals that have not been diagnosed with leukemia, chronic myeloblastic leukemia, or post-transplant lymphatic proliferative disease (PTLD). [18] The term "therapeutic agent" means a compound or composition used to treat a disease or condition in a patient. The therapeutic agent may be, for example, a polypeptide such as an antibody; Toxin (optionally linked to a targeting molecule such as an antibody); Gene therapy viral vectors and / or hemophilia factors (eg, factor VIII or factor IX). A therapeutic agent is generally administered to a mammal in a therapeutically effective amount for the treatment of the subject disease or condition, wherein the therapeutically effective amount refers to an amount that results in an immune response induced by the therapeutic agent in the mammal to be treated. [19] As used herein, "polypeptide" generally refers to peptides and proteins having about 10 or more amino acids. Examples of mammalian polypeptides include renin, human growth hormone, growth hormones including bovine growth hormone, growth hormone releasing factor, parathyroid hormone, thyroid stimulating hormone; Lipoprotein; 1-antitrypsin; Insulin A-chain; Insulin B-chain; Proinsulin; Thrombopoietin; Follicle stimulating hormone; Calcitonin; Progesterone; Glucagon; Blood coagulation factors such as factor VIIIC, factor IX, tissue factor, and von Willebrands factor; Anti-coagulation factors such as protein C; Atrial natriuretic factor; Waste surfactants; Plasminogen activators such as urokinase or human urine or tissue-type plasminogen activator (t-PA); Bombesin; Thrombin; Blood cell growth factor; Tumor necrosis factor-alpha and -beta; Enkephalinase; Serum albumin, eg, human serum albumin; Gallerian-inhibiting substances; Relaxin A-chain; Relaxin B-chain; Prolysine; Mouse gonadotropin-associated peptide; Microbial proteins such as beta-lactamase; DNase; Inhibin; Activin; Vascular endothelial growth factor (VEGF); Receptors for hormones or growth factors; Integrin; Protein A or D; Rheumatoid factor; Neurotrophic factors such as brain-derived neurotrophic factor (BDNF), neurotrophin-3, -4, -5 or -6 (NT-3, NT-4, NT-5 or NT-6) Or nerve growth factors such as NGF; Cardiotropin (heart hypertrophy factor), for example cardiotropin-1 (CT-1); Platelet-derived growth factor (PDGF); Fibroblast growth factors such as aFGF and bFGF; Epidermal growth factor (EGF); Transforming growth factors (TGF), for example TGF-alpha and TGF-beta, including TGF-1, TGF-2, TGF-3, TGF-4 or TGF-5; Insulin-like growth factor-I and -II (IGF-I and IGF-II); des (1-3) -IGF-I (brain IGF-I), an insulin-like growth factor binding protein; CD proteins such as CD3, CD4, CD8 and CD20; Erythropoietin; Osteoinduction factors; Immunotoxins; Bone morphogenic protein (BMP); Interferons such as interferon-alpha, -beta and -gamma; Serum albumin, eg, human serum albumin (HSA) or bovine serum albumin (BSA); Colony stimulating factors (CSFs) such as M-CSF, GM-CSF and G-CSF; Inteukrine (IL), for example IL-1 to IL-10; Cytokines (see below); Superoxide dismutase; T-cell receptor; Surface membrane proteins; Decline facilitating factor; Viral antigens, such as part of an AIDS envelope; Carrier protein; Homing receptor; Addressin; Regulatory proteins; Antibodies; And molecules such as fragments or variants of any of the polypeptides listed above. [20] As used herein, the term "graft" refers to a biological material derived from a donor in transplantation into a recipient. Grafts include isolated cells such as, for example, islet cells; Tissues such as amniotic membranes of the newborn, bone marrow, hematopoietic progenitor cells, and eye tissues such as corneal tissue; And various substances such as organs such as skin, heart, liver, spleen, pancreas, thyroid lobe, lung, kidney, vascular organs (eg, intestine, blood vessels or esophagus) and the like. Such tubular tissue can be used to replace damaged areas of the esophagus, blood vessels or bile ducts. Skin grafts can be used to heal some injuries, such as burns as well as dressings for damaged intestines or diaphragmatic hernias. Grafts are derived from any mammalian source, including humans, whether carcasses or living donors. Preferably, the graft is an organ such as bone marrow or heart, and the donor and host of the graft match against HLA class II antigen. [21] As used herein, the term "mammal host" refers to any suitable transplant recipient. By "suitable" is meant suitable for a mammalian host that will accept the donor graft. Preferably, the host is a human. If the donor and host of the graft are both humans, they are preferably matched against HLA class II antigens to enhance histocompatibility. [22] As used herein, the term "donor" refers to a mammalian species from which the graft is derived, whether dead or alive. Preferably, the donor is a human. Since the hybridization of the blood group barrier can impair the survival of allografts, the main human donor is preferably a donor associated with the blood of volunteers belonging to the same major ABO blood group as normal with physical examination. However, for example, the kidneys of type O donors can be implanted into type A, B or AB recipients. [23] The term “transplant” and its synonyms include that transplantation is homogeneous (genetically identical in donor and recipient), allogeneic (different genetic source of donor and recipient but in the same species) or heterogeneous (different donor and recipient Belonging to the species, means that the graft is inserted into the host. Thus, in a typical scenario, the host is a human and the graft is an allograft derived from humans of the same or different genetic origins. In other scenarios, the graft may be a porcine heart valve or animal beta islet cell or nerve transplanted into a human host, such as a baboon heart transplanted into a human recipient host, including animals derived from phylogenetically broadly classified species. It is derived from a species other than the target to be transplanted with the cell. [24] By "gene therapy" is meant a general method of introducing nucleic acids into a mammal treated by this therapy. The nucleic acid may encode the desired polypeptide or may be an antisense nucleic acid. One or more components of the gene therapy vector or composition may be immunogenic in a mammal treated therewith. For example, viral vectors (eg, adenoviruses, herpes simplex I virus or retroviruses); Lipids; And / or the targeting molecule in the composition can induce an immune response in a mammal to be treated therewith. [25] The expression “insensitive to a mammal to be transplanted” means to reduce or eliminate allergic sensitivity or responsiveness to the implant prior to administering the implant to the mammal. This can be accomplished by any mechanism, such as in reducing non-donor antibodies directed against human lymphocyte antigen (HLA) in an unsensitized mammal. [26] As used herein, "autoimmune disease" refers to a non-malignant disease or condition caused by and directed against an individual's own tissue. Autoimmune diseases herein refer to especially excluding malignant or cancerous diseases or symptoms, more particularly B cell lymphoma, acute lymphoblastic leukemia (ALL), chronic lymphocytic leukemia (CLL), blast cell leukemia and chronic myeloid leukemia. Examples of autoimmune diseases or disorders include inflammatory skin diseases including inflammatory reactions such as psoriasis and dermatitis (eg, atopic dermatitis); Systemic sclerosis and sclerosis; Reactions associated with inflammatory bowel disease (eg Crohn's disease and ulcerative colitis); Respiratory distress syndrome (adult refueling syndrome; including ARDS); dermatitis; Meningitis; encephalitis; Uveitis; colitis; Glomerulonephritis; Allergic symptoms such as eczema and asthma, and other symptoms including infiltration of T cells and chronic inflammatory responses; Atherosclerosis; Leukocyte adhesion deficiency; Rheumatoid arthritis; Systemic lupus erythematosus (SLE); Diabetes (eg, type I diabetes or insulin dependent diabetes); Multiple sclerosis; Reynaud's syndrome; Autoimmune thyroiditis; Allergic encephalomyelitis; Sjorgen's syndrome; Juvenile onset diabetes; And immune responses associated with acute and delayed hypersensitivity mediated by cytokines and T-lymphocytes commonly found in tuberculosis, sarcoidosis, multiple myositis, granulomatosis and vasculitis; Pernicious anemia (Addison's disease); Diseases including leukocyte outflow; Central nervous system (CNS) inflammatory diseases; Multiple organ damage syndrome; Hemolytic anemia (including but not limited to hypothermic globulinemia or Coomb positive anemia); Myasthenia gravis; Antigen-antibody complex mediated disease; Anti-glomerular basement membrane disease; Antiphospholipid syndrome; Allergic neuritis; Graves' disease; Lambert-Eaton work syndrome; Bullous whey; pemphigus; Autoimmune polyendocrine disease; Reiter's disease; Stiff-man syndrome; Behcet's disease; Giant cell arteritis; Immune complex nephritis; IgA kidney disease; IgM polyneuropathy; Immune thrombocytopenic purpura (ITP) or autoimmune thrombocytopenia, and the like. [27] An “antagonist”, when combined with CD20, destroys or kills B cells in a mammal and / or reduces one or more functions of B cells by, for example, reducing or preventing the humoral response induced by the B cells. It is a molecule that interferes. Antagonists are preferably capable of killing B cells (ie, reducing levels of circulating B cells) in mammals treated with them. Such killing may result from antibody-dependent cell-mediated cytotoxicity (ADCC) and / or complement dependent cytotoxicity (CDC), inhibition of B cell proliferation, and / or induction of B cell death (eg, via apoptosis) and This can be achieved through a variety of mechanisms. Antagonists included within the scope of the present invention include antibodies, synthetic or native sequence peptides and small molecule antagonists that bind to CD20, optionally linked or fused with a cytotoxic agent. Preferred antagonists include antibodies. [28] "Antibody-dependent cell-mediated cytotoxicity" and "ADCC" refer to the binding of nonspecific cytotoxic cells (eg, natural killer (NK) cells, neutrophils and macrophages) that express Fc receptors (FcRs) onto target cells. A cell-mediated reaction that recognizes an antibody and then lyses the target cell. The major cells that mediate ADCC, ie NK cells, express only FcγRIII, while monocytes express FcγRI, FcγRII and FcγRIII. Expression of FcR in hematopoietic cells is described by Ravetch and Kinet, Annu. Rev. Immunol 9: 457-92 (1991), Table 3 on page 464. To assess ADCC activity of the molecule of interest, an in vitro ADCC assay can be performed, for example as described in US Pat. No. 5,500,362 or 5,821,337. Useful functional cells for such assays include peripheral blood flow monocytes (PBMC) and natural killer (NK) cells. Alternatively or additionally, ADCC activity of the molecule of interest may be assessed in vivo, for example in animal models such as those described in Clynes et al., PNAS (USA) 95: 652-656 (1998). Can be. [29] "Human functional cells" are leukocytes that express one or more FcRs and perform functional functions. Preferably, these cells express at least FcyRIII and perform ADCC functional functions. Examples of human leukocytes that mediate ADCC include peripheral blood flow mononuclear cells (PBMC), natural killer (NK) cells, monocytes, cytotoxic T cells and neutrophils, of which PBMC and NK cells are preferred. [30] "Fc receptor" or "FcR" is a term used to describe a receptor that binds to the Fc region of an antibody. Preferred FcRs are native sequence human FcRs. In addition, preferred FcRs are receptors (gamma receptors) that bind IgF antibodies, examples include receptors of the FcγRI, FcγRII and FcγRIII subclasses, and allelic variants and optionally spliced forms of such receptors. FcγRII receptors include FcγRIIA (“activating receptor”) and FcγRIIB (“inhibiting receptor”) having similar amino acid sequences that differ primarily in their cytoplasmic domains. The activating receptor FcγRIIA contains an immunoreceptor tyrosine-based activation motif (ITAM) in its cytoplasmic domain. Inhibitory receptor FcγRIIB contains an immunoreceptor tyrosine-based inhibitory motif (ITIM) in its cytoplasmic domain [Daeron, Annu. Rev. Immunol. 15: 203-234 (1997). For FcRs, see Ravetch and Kinet, Annu. Rev. Immunol 9: 457-92 (1991); Capel et al., Immunomethods 4: 25-34 (1994); and de Haas et al., J. Lab. Clin. Med. 126: 330-41 (1995). Other FcRs, including those to be identified in the future, are included in the term "FcR" herein. The term also encompasses neonatal receptors, FcRn, which serve to deliver maternal IgG to the fetus (Guyer et al., J Immunol. 117: 587 (1976) and Kim etal, J Immunol. 24: 249 (1994). )). [31] "Complement dependent cytotoxicity" or "CDC" refers to the ability of a molecule to dissolve a target in the presence of complement. The complement active pathway is initiated by linking the first component (C1q) of the complement system to molecules (eg antibodies) complexed with homologous antigens. To assess complement activation, see, eg, Gazzano-Santoro et al., J. Immunol. Methods 202: 163 (1996) can be performed CDC analysis. [32] A "growth inhibitory" antagonist is one that prevents or reduces the proliferation of cells expressing the antigen to which the antagonist binds. For example, the antagonist can prevent or reduce the proliferation of B cells in vitro and / or in vivo. [33] An antagonist that "induces apoptosis" is determined by standard apoptosis assays such as binding of Annexin V, fragmentation of DNA, cell contraction, expansion of vesicles, fragmentation of cells and / or formation of membrane vesicles (called apoptosis bodies). As such, for example, those that induce planned cell death of B cells. [34] As used herein, "antibody" is used in the broadest sense and specifically includes intact monoclonal antibodies, polyclonal antibodies, multispecific antibodies (eg, bispecific antibodies) formed from two or more intact antibodies, as well as Antibody fragments are included as long as they exhibit the desired biological activity. [35] An “antibody fragment” comprises a portion of an intact antibody, preferably the antigen binding region or variable region of the intact antibody. Examples of antibody fragments include Fab, Fab ', F (ab') 2 and Fv fragments; Diabody; Linear antibodies; Single chain antibody molecules; And multispecific antibodies formed from antibody fragments and the like. [36] A “natural antibody” is a heterotetramer glycoprotein of about 150,000 daltons, which generally consists of two identical light chains (L) and two identical heavy chains (H). Each light chain is linked to the heavy chain by one disulfide covalent bond, but the number of disulfide bonds differs in the heavy chain of different immunoglobulin isoforms. In addition, each heavy and light chain has disulfide bonds within the chain at regular intervals. Each heavy chain has at one end a variable domain (V H ) and a number of constant domains following this domain. Each light chain has a variable domain (V L ) at one end and a constant domain at the other end, the constant domain of the light chain is arranged side by side with the first constant domain of the heavy chain, and the light chain variable domain is arranged side by side with the variable domain of the heavy chain. . Particular amino acid residues are believed to form an interface between the light and heavy chain variable domains. [37] The term "variable" refers to the fact that the sequences of specific regions of the variable domains differ greatly between antibodies and are used for the binding and specificity of each particular antibody to that particular antigen. However, the variability is not evenly distributed throughout the variable domains of antibodies. Variability is concentrated in three segments called hypervariable regions of both the light and heavy chain variable domains. The higher conserved portion of the variable domain is called the framework region (FR). The variable domains of natural heavy and light chains comprise four framework regions, mainly conformation of β-sheets, which are linked by three hypervariable regions that form a loop linkage and in some cases form part of the β-sheet structure. . The hypervariable regions in each chain are kept in close proximity to each other by the FR regions, and the hypervariable regions of the other chain contribute to the formation of the antigen binding site of the antibody (Kabat et al., NIH Publ. No. 91-3242, Vol. I, pages 647-669 (1991). The constant domains are not directly involved in antigen binding of the antibody, but exhibit various functional functions such as the involvement of the antibody in cytotoxicity (ADCC) of antibody dependent cells. [38] Digestion of the antibody with papain results in two identical antigen binding fragments, each "Fab" fragment with a single antigen binding site, and the remaining "Fc" fragment, which reflects its ability to readily crystallize. Treatment with pepsin results in an F (ab ') 2 fragment that has two antigen binding sites and still can cross-link antigen. [39] "Fv" is the minimum antibody fragment that contains a complete antigen recognition and antigen binding site. This region consists of a dimer consisting of one heavy chain variable domain and one light chain variable domain that are tightly covalently bonded. In this structure, three hypervariable regions of each variable domain interact to form antigen binding sites on the surface of the V H -V L dimer. In conclusion, six hypervariable regions confer antigen binding specificity to antibodies. However, even a single variable domain (or half of the Fv comprising only three hypervariable regions specific for the antigen) has affinity than the entire binding site but has the ability to recognize and bind the antigen. [40] The Fab fragment also contains the constant domain of the light chain and the first constant domain of the heavy chain (CH1). Fab 'fragments differ from Fab fragments in that several residues have been added to the carboxy terminus of the heavy chain CH1 domain comprising one or more cysteines derived from the antibody hinge region. Fab'-SH herein refers to Fab 'in which the cysteine residue (s) of the constant domains bear one or more free thiol groups. F (ab ') 2 antibody fragments originally were produced as pairs of Fab' fragments with hinge cysteines between them. Other chemical couplings of antibody fragments are also known. [41] The "light chain" of an antibody (immunoglobulin) derived from any vertebrate species can be classified into one of two distinctly different types called kappa (κ) and lambda (λ) based on the amino acid sequences of its constant domains. have. [42] Antibodies can be classified into different classes depending on the amino acid sequence of the constant domain of the heavy chain. Intact antibodies have five major classes of IgA, IgD, IgE, IgG and IgM, some of which can be further classified into subclasses (isotypes), for example IgG1, IgG2, IgG3, IgG4, IgA and IgA2. have. The heavy chain constant domains corresponding to the different classes of antibodies are called α, δ, ε, γ, and μ, respectively. Subunit structures and three-dimensional structures for different classes of immunoglobulins are well known. [43] "Single-chain Fv" or "scFv" antibody fragments comprise the V H and V L domains of an antibody present in a single polypeptide chain. Preferably, the Fv polypeptide also comprises a polypeptide linker between the V H and V L domains, which allows the scFv to form the desired structure for antigen binding (see Pluckthun in The Pharmacology of Monoclonal Antibodies, vol. 113, Rosenburg and Moore eds., Springer-Verlag, New York, pp. 269-315 (1994)). [44] The term "diabody" refers to a small, two antigen binding site comprising a heavy chain variable domain (V H ) linked to a light chain variable domain (V L ) within the same polypeptide chain (V H -V L ). Refers to an antibody fragment. By using linkers that are too short to link two domains in the same chain, the domains are forcibly linked with the complementary domains of the other chain to create two antigen binding sites. Diabodies are described, for example, in more detail in European Patent 404,097, WO 93/11611 and in Hollinger et al., Proc. Natl. Acad. Sci. USA, 90: 6444-6448 (1993). It is described. [45] As used herein, the term “monoclonal antibody” refers to an antibody obtained from a population of substantially homologous antibodies, ie, identical individual antibodies that form a population except for possible naturally occurring mutations that may be present in small amounts. Monoclonal antibodies are highly specific antibodies directed against a single antigenic site. In addition, unlike conventional (polyclonal) antibody preparations that typically include different antibodies against different determinants (epitopes), each monoclonal antibody is directed against a single determinant on the antigen. In addition to their specificity, monoclonal antibodies have the advantage of being synthesized by hybridoma cultures that are not contaminated by other immunoglobulins. The modifier “monoclonal” refers to antibody characteristics obtained from a substantially homogeneous antibody population and is not considered to be required for the production of antibodies in any particular manner. For example, monoclonal antibodies to be used in accordance with the present invention may first be prepared by the hybridoma method described in Kohler et al., Nature, 256: 495 [1975], or recombinant DNA methods (eg For example, US Pat. No. 4,816,576. "Monoclonal antibodies" are also described, eg, in Clackson et al., Nature, 352: 624-628 [1991] and Marks et al., J. Mol. Biol., 222: 581-597 (1991). It can also be isolated from phage antibody library using the technique described in the). [46] Monoclonal antibodies herein specifically comprise a corresponding sequence of an antibody derived from a particular species or a corresponding class of an antibody belonging to a particular antibody class or subclass as long as it exhibits the desired biological activity. The same or homologous, but the remainder of the chain (s) is a "chimeric" antibody (immunoglobulin) that is identical or homologous to an antibody from another species or an antibody belonging to another antibody class or subclass or fragment of such an antibody. (US Pat. No. 4,816,567; Morrison et al., Proc. Natl. Acad. Sci. USA, 81: 6851-6855 [1984]). Chimeric antibodies of interest herein include "primatization" comprising variable domain antigen binding sequences and human constant region sequences derived from non-human primates (e.g., Old World monkeys such as baboons, rhesus or cynomolgus monkeys). "Antibodies are included (US Pat. No. 5,693,780). [47] A “humanized” form of a non-human (eg murine) antibody is a chimeric antibody comprising a minimal sequence derived from a non-human immunoglobulin. In most cases humanized antibodies are residues from hypervariable regions of non-human species (donor antibodies) such as mice, rats, rabbits or non-human primates that have the desired specificity, affinity and ability to residues of the hypervariable region of the recipient. Human immunoglobulin (receptor antibody) substituted with a. In some cases, residues of the framework region (FR) of human immunoglobulins are replaced by corresponding non-human residues. Humanized antibodies may also include residues that are not found in either the recipient antibody or the donor antibody. Such modifications are further made to improve antibody performance. In general, humanized antibodies will comprise substantially all of one or more, typically two variable domains, where all or substantially all hypervariable loops correspond to regions of non-human immunoglobulins and all or substantially all FR corresponds to the region of human immunoglobulin sequence. In addition, the humanized antibody will optionally comprise at least a portion of an immunoglobulin constant region (Fc), typically a portion of a human immunoglobulin region. For more details, see Jones et al., Nature, 321: 522-525 (1986); Riechmann et al., Nature, 332: 323-329 (1988); And Presta, Curr. Op. Struct. Biol. 2: 593-596 (1992). [48] As used herein, “hypervariable region” refers to an amino acid residue of an antibody that is responsible for antigen binding. The hypervariable region is referred to as the "complementarity determining region" or "CDR" (residues 24 to 34 (L1), 50 to 56 (L2) and 89 to 97 (L3) of the light chain variable domain, and residues 31 to 35 ( H1), 50-65 (H2) and 95-102 (H3); Kabar et al., Sequences of Proteins of Immunological Interests, 5th Ed. Public Health Services, National Institute of Health, Bethesda, MD. [1990] Amino acid residues and / or “hypervariable loops” (residues 26 to 32 (L1), 50 to 52 (L2) and 91 to 96 (L3) of the light chain variable domain, and residues 26 to 32 (H1) of the heavy chain variable domain ), 53-55 (H2) and 96-101 (H3); amino acid residues from Clothia and Lesk, J. Mol. Biol., 196: 901-917 [1987]). “Framework” or “FR” residues are those residues of the variable domain other than the hypervariable region residues as herein defined. [49] An antagonist that "binds" with an antigen of interest, for example CD20, is one that is capable of binding an antigen with sufficient affinity and / or affinity such that the antagonist is useful as a therapeutic agent targeting cells expressing this antigen. [50] Examples of antibodies that bind CD20 include "C2B8" called "rituximab" (RITUXAN®) (US Pat. No. 5,736,137, incorporated herein by reference); Yttrium- [90] -labeled 2B8 murine antibody designated “Y2B8” (US Pat. No. 5,736,137, which is incorporated herein by reference); Murine IgG2a "B1" (BEXXAR®) optionally labeled with 131 I to generate a " 131 I-B1" antibody (US Pat. No. 5,595,721, incorporated herein by reference); Murine monoclonal antibody “1F5” (Press et al., Blood 69 (2): 584-591 (1987)); "Chimeric 2H7" antibodies (US Pat. No. 5,677,180, which is incorporated herein by reference); And monoclonal antibodies L27, G28-2, 93-1B3, B-C1 or NU-B2 (Valentine et al, In: Leukocyte Tvping III (McMichael, Ed., P. 440) available from the International Lucosite Typing Workshop. , Oxford University Press (1987)). [51] As used herein, the term rituximab or rituxan (RITUXAN®) is a genetically engineered chimeric rat designated as “C2B8” in US Pat. No. 5,736,137, which is directed to the CD20 antigen and incorporated herein by reference. Human monoclonal antibody. This antibody is an IgGl kappa immunoglobulin comprising murine light and heavy chain variable region sequences and human constant region sequences. Rituximab has a binding affinity of about 8.0 nM for the CD20 antigen. [52] An “isolated” antagonist is an antagonist identified and separated from and / or recovered from a component of its natural environment. Contaminant components of the natural environment are substances that interfere with the diagnostic or therapeutic use of antagonists and may include enzymes, hormones, and other proteinaceous or nonproteinaceous solutes. In a preferred embodiment, the antagonist is (1) greater than 95% by weight, most preferably greater than 99% by weight of the antagonist as measured by the Lowry method, and (2) using a spinning cup sequencer. Sufficient to obtain 15 or more of the N-terminal or internal amino acid sequence, or (3) purified in one band by SDS-PAGE under reducing or non-reducing conditions using Coomassie blue, preferably silver staining Will be. Isolated antagonist includes the antagonist present in its original position in the recombinant cell since at least one component will not be present in the natural environment of the antagonist. However, isolated antagonists will generally be purified by one or more purification steps. [53] "Mammal" for therapeutic purposes refers to any animal that is classified as a mammal, including humans, livestock and breeding animals, and zoos, competitions or pets such as dogs, horses, cats, cows, and the like. Preferred mammals are humans. [54] "Treatment" refers to both therapeutic treatment and preventive or preventive measures. Subjects in need of treatment include those already afflicted with the disease or condition as well as those who need to prevent the disease or condition. Thus, the mammal may be diagnosed with, or prone to, or susceptible to a disease or condition. [55] The expression “therapeutically effective amount” refers to the amount of antagonist that is effective in preventing, ameliorating or treating the disease or condition. [56] As used herein, the term "immunosuppressant" for adjuvant therapy refers to a substance that functions to inhibit or mask the immune system of the mammal to be treated herein. These substances include substances that inhibit cytokine production, substances that downregulate or inhibit self-antigen expression, or substances that mask MHC antigens. Examples of such materials include 2-amino-6-aryl-5-substituted pyrimidines (US Pat. No. 4,665,077, the disclosure of which is incorporated herein by reference); Antiproliferative substances such as azathioprine, leflunomide or sirolimus; Cyclophosphamide; Bromocriptine; Danazol; Dapson, glutaraldehyde (masking MHC antigens as described in US Pat. No. 4,120,649); Anti-idiotype antibodies against MHC antigens and MHC fragments; Cyclosporin A; Steroids such as corticosteroids, for example prednisone, methylprednisolone and dexamethasone; Mycophenolate mofetil; Calcineurin inhibitors (eg tacrolimus); Cytokines, including anti-interferon-γ, -β or -α antibodies, anti-tumor necrosis factor-α antibodies, anti-tumor necrosis factor-β antibodies, anti-interleukin-2 antibodies and anti-IL-2 receptor antibodies Cytokine receptor antagonists; Anti-LFA-1 antibodies, including anti-CD11a and anti-CD18 antibodies; Anti-L3T4 antibodies; Anti-lymphocyte antibodies, eg, polyclonal anti-lymphocyte antibodies; pan-T antibodies, preferably anti-CD3 or anti-CD4 / CD4a antibodies; Soluble peptide comprising an LFA-3 binding domain (W090 / 08187 published July 26, 1990); Streptokinase; TGF-β; Streptodonase; RNA or DNA from the host; FK506; RS-61443; Deoxyspergualin; Rapamycin; T-cell receptor (Cohen et al., US Pat. No. 5,114,721); T-cell receptor fragments (Offner et al., Science, 251: 430-432 (1991); WO 90/11294; Ianeway, Nature, 341: 482 (1989); and WO 91/01133); And T cell receptor antibodies (European Patent No. 340,109), for example T10B9. [57] As used herein, the term "cytotoxic agent" refers to a substance that inhibits or inhibits the function of a cell and / or destroys a cell. This term refers to radioisotopes (e.g., radioisotopes of At 211 I 131 , I 125 , Y 90 , Re 186 , Re 188 , Sm 153 , Bi 212 , P 32 and Lu), chemotherapeutic agents and toxins ( Small molecule toxins or enzymatically active toxins of bacterial, fungal, plant or animal origin, or fragments thereof). [58] A "chemotherapeutic agent" is a compound useful for the treatment of cancer. Examples of chemotherapeutic agents include alkylating agents such as thiotepa and cyclophosphamide (CYTOXAN®); Alkyl sulfonates such as busulfan, impprosulfan and pifosulfan; Aziridine such as benzodopa, carbocuone, methuredopa and uredopa; Ethyleneimines and methylamelamine, including altretamine, triethylenemelamine, triethylenephosphoramide, triethylenethiophosphoramide and trimethylolomelamine; Nitrogen mustards such as chlorambucil, chlornaphazine, colophosphamide, esturamustine, ifosfamide, mechloretamine, mechloretamine oxide hydrochloride, melphalan, nozambisin, fensterrin , Prednismustine, trophosphamide, uracil mustard; Nitrosureas such as carmustine, chlorozotocin, potemustine, lomustine, nimustine, rannimustine; Antibiotics, for example alaccinomycin, actinomycin, outtramycin, azaserine, bleomycin, carctinomycin, calicheamicin, carabicin, carminomycin, carcinophylline, chromomycin, dactinomycin , Daunorubicin, detorrubicin, 6-diazo-5-oxo-L-norleucine, doxorubicin, epirubicin, esorubicin, idarubicin, marcelomycin, mitomycin, mycophenolic acid, nogala Mycin, olibomycin, peplomycin, port pyromycin, puromycin, quelamycin, rhorubicin, streptonigrin, streptozosin, tubercidine, ubenimex, ginostatin, zorubicin; Anti-metabolites such as methotrexate and 5-fluorouracil (5-FU); Folic acid analogs such as denophtherine, methotrexate, putrophtherin, trimetrexate; Purine analogs such as fludarabine, 6-mercaptopurine, thiamiprine, thioguanine; Pyrimidine analogs such as ancitabine, azacytidine, 6-azauridine, carmofur, cytarabine, dideoxyuridine, doxyfluridine, enositabine, phloxuridine, 5-FU; Androgens such as calussterone, dromostanolone propionate, epithiostanol, mepitiostane, testosterone; Anti-adrenal such as aminoglutetimide, mitotan, trilostane; Folic acid sources such as proline acid; Aceglaton; Aldophosphamide glycosides; Aminolevulinic acid; Amsacrine; Vestravusyl; Bisantrene; Edatraxate; Depopamine; Demecolsin; Diajikuon; Elponnitine; Elftinium acetate; Etogluside; Gallium nitrate; Hydroxyurea; Lentinane; Rodidamine; Mitoguazone; Mitoxantrone; Fur mall; Nitracrine; Pentostatin; Penammet; Pyrarubicin; Grape filinic acid; 2-ethylhydrazide; Procarbazine; PSK®; Lakamic acid; Sizopyran; Spirogermanium; Tenuazone acid; Triazcuone; 2,2 ', 2' '-trichlorotriethylamine; urethane; Bindesin; Dacarbazine; Mannomustine; Mitobronitol; Mitolactol; Fifobroman; Pricktocin; arabinoside ("Ara-C"); Cyclophosphamide; Thiotepa; Taxoids such as paclitaxel (TAXOL®, Bristol-Myers Squibb Oncology, Princeton, NJ) and docetaxel (TAXOTERE®, Rhone-Poulenc Rorer, Antony, France); Chlorambucil; Gemcitabine; 6-thioguanine; Mercaptopurine; Methotrexate; Platinum analogs such as cisplatin and carboplatin; Vinblastine; platinum; Etoposide (VP-16); Ifosfamide; Mitomycin C; Mitoxantrone; Vincristine; Vinorelbine; Navelvin; Novantron; Teniposide; Daunomycin; Aminopterin; Xceloda; Ibandronate; CPT-11; Topoisomerase inhibitor RFS2000; Difluoromethylornithine (DMFO); Retinoic acid; Esperamycin; Capecitabine; And pharmaceutically acceptable salts, acids or derivatives of those mentioned above. This definition also includes, for example, tamoxifen, raloxifene, aromatase inhibitory 4 (5) -imidazole, 4-hydroxytamoxifen, trioxyphene, keoxyphene, LY117018, onafristone and toremiphene (Fareston). Anti-hormonal agents that act to modulate or inhibit hormonal action on the tumor, such as estrogens; And anti-androgens such as flutamide, nilutamide, bicalutamide, lutrolide and goserelin; And pharmaceutically acceptable salts, acids or derivatives of those mentioned above. [59] "Cytokine" is a generic term for a protein that acts on another cell as an intercellular mediator, released by one cell population. Examples of such cytokines are lymphokine, monocaine and typical polypeptide hormones. Cytokines include growth hormones such as human growth hormone, N-methionyl human growth hormone and bovine growth hormone; Parathyroid hormone; Thyroxine; insulin; Proinsulin; Relaxin; Prolylacin; Glycoprotein hormones such as follicle stimulating hormone (FSH), thyroid stimulating hormone (TSH) and progesterone (LH); Hepatocyte growth factor; Fibroblast growth factor; Prolactin; Placental lactogen; Tumor necrosis factor-α and -β; Gallerian inhibitors; Mouse gonadotropin-related peptide; Inhibin; Activin; Vascular endothelial growth factor; Integrin; Thrombopoietin (TPO); Nerve growth factors such as NGF-β; Platelet growth factor; Transforming growth factors (TGFs) such as TGF-α and -β; Insulin-like growth factor-I and -II; Erythropoietin (EPO); Osteoinductive factor; Interferons such as interferon-α, -β and -γ; Colony stimulating factor (CSF), eg, macrophage-CSF (M-CSF); Granulocyte-macrophage-CSF (GM-CSF); And granulocyte-CSF (G-CSF); Interleukin (IL), for example IL-1, IL-1α, IL-2, IL-3, IL-4, IL-5, IL-6, IL-7, IL-8, IL-9, IL -11, IL-12, IL-15; Tumor necrosis factors such as TNF-α or TNF-β; And other polypeptide factors including LIF and kit ligand (KL). As used herein, the term cytokine includes biologically active equivalents of native sequence cytokines and proteins from natural sources or recombinant cell culture. [60] As used herein, the term “prodrug” refers to a precursor or derivative form of a pharmaceutically active substance that is less cytotoxic to tumor cells than the parent drug and can be converted into an enzymatically activated or more active parental form. (Wilman, "Prodrugs in Cancer Chemotherapy", Biochemical Society Transactions, 14: 375-382, 615th Meeting, Belfast (1986)) and Stella et al., "Prodrugs: A Chemical Approach to Targeted Drug Delivery", Directed Drug Delivery, Borchardt et al., (Ed.), Pp. 147-276, Humana Press (1985)). Prodrugs of the present invention include phosphate-containing prodrugs, thiophosphate-containing prodrugs, sulfate-containing prodrugs, peptide-containing prodrugs, D-amino acid modified prodrugs, glycosylated which can be converted to more active cell nontoxic drugs Prodrugs, β-lactam containing prodrugs, optionally substituted phenoxyacetamide containing prodrugs or optionally substituted phenylacetamide containing prodrugs, 5-fluorocytosine and other 5-fluorouridine prodrugs, It is not limited to this. Examples of cytotoxic drugs that can be derivatized with prodrugs for use in the present invention include, but are not limited to, the chemotherapeutic agents described above. [61] A "liposome" is a vesicle consisting of several types of lipids, phospholipids, and / or surfactants useful for delivering drugs (eg, antagonists and optionally chemotherapeutic agents disclosed herein) to a mammal. Liposomal components are typically arranged in bilayer form similar to the lipid arrangement of biological membranes. [62] The term "package insert" is commonly included in commercially available therapeutic product packages that contain information about indications, usage, dosage, dosage, contraindications, and / or cautions in the use of such therapeutic products. Means things. [63] II. Production of antagonists [64] The methods and products of the present invention use or include antagonists that bind to CD20. Thus, the following describes a method for producing such antagonists. [65] The CD20 antigen used for the production or screening of the antagonist may be, for example, a soluble form of the antigen comprising the epitope of interest or a portion thereof. Alternatively or additionally, cells expressing CD20 on their cell surface can be used to generate or screen antagonists. Other forms of CD20 useful for producing antagonists are apparent to those skilled in the art. [66] Preferred antagonists are antibodies, but antagonists other than antibodies are also considered herein. For example, the antagonist may optionally include small molecule antagonists that are fused to or associated with a cytotoxic agent (such as those described herein). Small molecule libraries can be screened for CD20 to identify small molecules that bind to this antigen. These small molecules may be further screened for their antagonist properties and / or combined with cytotoxic agents. [67] The antagonist may be a peptide produced by rational design or phage display (see, eg, W098 / 35036, published August 13, 1998). In an embodiment, the selection molecule can be a “CDR mimetic” or antibody analogue designed based on the CDRs of the antibody. While these peptides may themselves be antagonistic, they may be optionally fused to cytotoxic agents to add or enhance the antagonist properties of the peptide. [68] In the following, exemplary techniques for the production of antibody antagonists for use in accordance with the present invention are described. [69] (i) polyclonal antibodies [70] Polyclonal antibodies are preferably produced by several subcutaneous (sc) or intraperitoneal (ip) injections of the relevant antigen and adjuvant in an animal. Difunctional or derivatized materials such as maleimidobenzoyl sulfosuccinimide esters (linked through cysteine residues), N-hydroxysuccinimides (linked through lysine residues), glutaraldehyde, succinic anhydride, Proteins that are immunogenic in the species to be immunized with the relevant antigen using SOCl 2 , or R 1 N═C═NR where R and R 1 are other alkyl groups, eg, keyhole limpet hemocyanin, serum albumin, Linking with bovine thyroglobulin or soybean trypsin inhibitor may be useful. [71] Antibodies are challenged by combining 100 μg or 5 μg of protein or conjugate (for rabbits or mice, respectively) with Freund's complete adjuvant, which is three times their amount and injecting the solution into the skin at various sites. , Immunogenic conjugates or derivatives. One month thereafter, animals were challenged by subcutaneous injection of 1/5 to 1/10 of the original amount of peptide or conjugate into various sites in the Freund's complete adjuvant. After 7-14 days, animals were bled and serum was analyzed for antibody titer. Animals were challenged until titer retention. Preferably, the antigens were challenged with conjugates of the same antigen linked to other proteins and / or linked by different cross-linking reagents. Conjugates can also be prepared as protein fusions in recombinant cell culture. In addition, flocculants such as alum are suitably used to boost the immune response. [72] (ii) monoclonal antibodies [73] Monoclonal antibodies are obtained substantially from a population of homologous antibodies, i.e. each antibody constituting the population is identical except for possible naturally occurring mutations that may be present in small amounts. Thus, the modifier "monoclonal" characterizes an antibody rather than as a mixture of various antibodies. [74] For example, monoclonal antibodies can be prepared using the hybridoma method first described in Kohler et al., Nature, 256: 495 (1975) or by recombinant DNA method (US Pat. No. 4,816,567). have. [75] In hybridomas, mice or other suitable host animals, such as hamsters, are immunized as mentioned above to induce lymphocytes capable of producing or producing antibodies that specifically bind to proteins used for immunization. Alternatively, lymphocytes can be immunized in vitro. Lymphocytes are then fused with myeloma cells to form hybridoma cells using a suitable fusing agent such as polyethylene glycol (Goding, Monoclonal Antibodies. Principles and Practice, pp. 59-103 (Academic Press, 1986)). [76] The hybridoma cells thus prepared are seeded and cultured in a suitable culture medium, preferably comprising one or more substances that inhibit the proliferation or survival of unfused parental myeloma cells. For example, if the parental myeloma cells lack the enzyme hypoxanthine guanine phosphoribosyl transferase (HGPRT or HPRT), the culture medium for hybridomas is typically hypo Forc, a substance that inhibits the proliferation of HGPRT-deficient cells. Will include xanthine, aminopterin and thymidine (HAT medium). [77] Preferred myeloma cells are cells that effectively fuse, support stable high levels of production of antibodies by selected antibody-producing cells, and are sensitive to media such as HAT medium. Among these, preferred myeloma cell lines are derived from mouse myeloma cell lines, eg, MOPC-21 and MPC-11 mouse tumors available from Salk Institute Cell Distribution Center (San Diego, California USA). And those derived from SP-2 or X63-Ag8-653 cells available from the American Type Culture Collection (Rockville, Maryland USA). Human myeloma cell lines and mouse human heteromyeloma cell lines have also been described for the production of human monoclonal antibodies (Kozbor, J. Immunol., 133: 3001 (1984); Brodeur et al., Monoclonal Antibody Production Techniques and Applications, pp. 51-63 (Marcel Dekker, Inc., New York, 1987)). [78] Culture medium in which hybridoma cells are grown is assayed for production of monoclonal antibodies directed against the antigen. Preferably, the binding specificity of the monoclonal antibodies produced by the hybridoma cells is determined by immunoprecipitation or in vitro binding assays such as radioimmunoassay (RIA) or enzyme-linked immunosorbent assay (ELISA). Decide [79] The binding affinity of monoclonal antibodies can be determined, for example, by the Scatchard assay of Munson et al, Anal Biochem., 107: 220 (1980). [80] Hybridoma cells producing antibodies of the desired specificity, affinity, and / or activity were identified, then the clones were subcloned by limiting dilution methods and cultured by standard methods (Goding, Monoclonal Antibodies: Principles and Practice). , pp. 59-103 (Academic Press, 1986)). Suitable culture media for this purpose include, for example, D-MEM or RPMI-1640 medium. In addition, the hybridoma cells may be cultured in vivo as ascites tumors in an animal. [81] The monoclonal antibodies secreted by the subclones are suitably, for example, conventional immunonoids such as protein A-Sepharose, hydroxylapatite chromatography, gel electrophoresis, dialysis or affinity chromatography. It is isolated from the culture medium, ascites fluid or serum by the globulin purification method. [82] DNA encoding monoclonal antibodies is readily isolated and sequenced by conventional methods (e.g., using oligonucleotide probes that can specifically bind to the heavy and light chains of a murine antibody). . Hybridoma cells serve as a preferred source of such DNA. After isolation, the DNA can be placed in an expression vector, which then does not produce immunoglobulin proteins. Monoclonal antibodies are synthesized in recombinant host cells by transfection into host cells such as E. coli cells, monkey COS cells, Chinese hamster ovary (CHO) cells or myeloma cells. For studies on recombinant expression of DNA encoding such antibodies in bacteria, see Skerra et al, Curr. Opinion in Immunol., 5: 256-262 (1993) and Pluckthun, ImmunoL. Revs., 130: 151-188 (1992). [83] In a further embodiment, the antibody or antibody fragment is described using McPafferty et al, Nature, 348: 552-554 (1990), using phage libraries to describe the isolation of each of murine and human antibodies. Clackson et al, Nature, 352: 624. -628 (1991) and Marks et al., J. Mol Biol., 222: 581-597 (1991)) can be isolated from the antibody phage library obtained. Subsequent documents include chain shuffling (Marks et al, Biol. Technology, 10: 779-783 (1992)), as well as combinatorial infection and in vivo recombination (Waterhouse et al., Nuc), a method for producing very large phage libraries. The production of high affinity (nM range) human antibodies by Acids Res., 21: 2265-2266 (1993)) is described. Thus, these techniques are available as an alternative to conventional monoclonal antibody hybridoma techniques for isolation of monoclonal antibodies. [84] Also, for example, DNA can be substituted for non-immunoglobulin polypeptides by replacing sequences encoding human heavy and light chain constant domains instead of homologous murine sequences (US Pat. No. 4,816,567; Morrison et al., Supra). All or part of the coding sequence for the mutation may be covalently linked to an immunoglobulin coding sequence. [85] Typically, the non-immunoglobulin polypeptide is substituted for a constant domain of an antibody or substituted for a variable domain of one antigen-binding site of an antibody to have specificity for one antigen-binding site and another antigen. Chimeric bivalent antibodies comprising other antigen-binding sites with may be generated. [86] (iii) humanized antibodies [87] Methods for humanizing nonhuman antibodies are well known in the art. Preferably, the humanized antibody has one or more amino acid residues introduced into the antibody derived from a non-human source. These nonhuman amino acid residues are often referred to as "import" residues and are typically obtained from an "import" variable domain. Humanization is essentially performed by Winter et al. (Jones et al., Nature, 321: 522-525 (1986); Riechmann et al., Nature, by substituting the hypervariable region sequences in place of the corresponding sequences of human antibodies). 332: 323-327 (1988); Verhoeyen et al., Science, 239: 1534-1536 (1988)). Thus, such “humanized” antibodies are chimeric antibodies (US Pat. No. 4,816,567) in which substantially less intact human variable domains are replaced by corresponding sequences from non-human species. In practice, humanized antibodies are typically human antibodies in which some hypervariable region residues and possibly some FR residues are substituted by residues from the cognate sites of rodent antibodies. [88] The choice between the human light chain variable domain and the human heavy chain variable domain to be used to prepare a humanized antibody is very important for reducing antigenicity. According to the so-called "most suitable" method, the sequences of the variable domains of rodent antibodies are screened against the entire library of known human variable-domain sequences. Next, the human sequence closest to the rodent sequence is adopted as the human framework region (FR) for humanized antibodies (Sims et al, J. Immunol, 151: 2296 (1993); Chothia et al., J. Mol Biol, 196: 901 (1987)). Another method uses a specific framework region derived from the consensus sequence of all human antibodies of a particular subgroup of light or heavy chains. The same backbone can be used for several different humanized antibodies (Carter et al., Proc. Natl. Acad. Sci. USA, 89: 4285 (1992); Presta et al, J. Immunol., 151: 2623 ( 1993). [89] It is also important to humanize the antibodies to have high affinity for antigens and other beneficial biological properties. To achieve this object, humanized antibodies are prepared by methods of analysis of the parental sequences and various theoretical humanized products, using three-dimensional models of the parental and humanized sequences, according to a preferred method. Three-dimensional immunoglobulin models are commercially available and known to those skilled in the art. Computer programs are available that illustrate and show possible three-dimensional structures of selected candidate immunoglobulin sequences. Examining the display of such computer programs allows for the analysis of possible roles of residues in the functionalization of candidate immunoglobulin sequences, ie the analysis of residues that affect the ability of a candidate immunoglobulin to bind its antigen. In this manner, FR residues can be selected and combined from the acceptor sequence and the import sequence to obtain the properties of the desired antibody, such as increased affinity for the target antigen (s). In general, the residues of the hypervariable regions are directly and most substantially involved in influencing antigen binding. [90] (iv) human antibodies [91] As an alternative to humanization, human antibodies can be generated. For example, upon immunization, it is also currently possible to make transgenic animals (eg mice) capable of producing the entire repertoire of human antibodies in the absence of endogenous immunoglobulin production. For example, it has been described that homozygous deletion of the antibody heavy chain binding region (J H ) gene in chimeric and germ line mutant mice completely inhibits endogenous antibody production. Delivery of the human germline immunoglobulin gene sequence into such germline mutant mice will result in the production of human antibodies upon exposure to the antigen [see, eg, Jakobovits et al, Proc. Natl. Acad. Sci. USA, 90: 2551 (1993); Jakobovits et al, Nature, 362: 255-258 (1993); Bruggermann et al, Year in Immuno., 7: 33 (1993); and US Pat. Nos. 5,591,669, 5,589,369 and 5 5,545,807). [92] Alternatively, human antibodies and antibody fragments from immunoglobulin variable (V) domain gene repertoires obtained from unimmunized donors using phage display technology (McCafferty et al., Nature 348: 552-553 (1990)) were tested. Can be produced in the building. According to this technique, antibody V domain genes are cloned in-frame into the major coating protein or minority coating protein gene of a fibrous bacteriophage such as M13 or fd and displayed as functional antibody fragments on the surface of the phage particle. Since the fibrous particles contain a single stranded DNA copy of the phage genome, selection based on the functional properties of the antibody allows the selection of genes encoding antibodies exhibiting these properties. Thus, phage mimic some of the properties of B cells. Phage display can be performed in a variety of formats (see, eg, Johnson, Kevin S. and Chiswell, David J., Current Opinion in Structural Biology 3: 564-571 (1993)). Several sources can be used for phage display: Clackson et al., Nature, 352: 624-628 (1991) is a variable array of terms from a small random combination library of V genes derived from the spleen of immunized mice. -Oxazolone antibody was isolated A repertoire of V genes obtained from non-immunized human donors was constructed and essentially described in Marks et al., J. Mol Biol. 222: 581-597 (1991), or Griffith et al. , EMBO J. 12: 725-734 (1993), also see US Pat. Nos. 5,565,332 and 5,573,905, to isolate antibodies against a wide array of antigens (including self-antigens). . [93] Human antibodies are also produced by activated B cells in vitro (see US Pat. Nos. 5,567,610 and 5,229,275). [94] (v) antibody fragments [95] Various techniques have been developed for producing antibody fragments. Conventionally, these fragments were derived through proteolytic cleavage of intact antibodies (see, eg, Morimoto et al., Journal of Biochemical and Biophysical Methods 24: 107-117 (1992) and Brennan et al., Science, 229: 81 (1985)). However, these fragments can now be produced directly by recombinant host cells. For example, antibody fragments can be isolated from the antibody phage libraries discussed above. Or in the alternative, the Fab'-SH fragment is E. coli. Direct recovery from E. coli can be chemically coupled to F (ab ') 2 fragments (Carter et al., Bio / Technology 10: 163-167 (1992)). According to another method, F (ab ') 2 fragments can be isolated directly from the host cell culture. Other techniques for producing antibody fragments will be apparent to those skilled in the art. In other embodiments, the antibody of choice is a single chain Fv fragment (scFv) (see WO 93/16185; US Pat. No. 5,571,894; and US Pat. No. 5,587,458). The antibody fragment may be, for example, a "linear antibody" as described in US Pat. No. 5,641,870. Such linear antibodies may be monospecific or bispecific. [96] (vi) bispecific antibodies [97] Bispecific antibodies are antibodies that have binding specificities for at least two different epitopes. Exemplary bispecific antibodies may bind to two different epitopes of CD20. Alternatively, the anti-CD20 binding arm may be a triggering molecule on leukocytes, such as a T-cell receptor molecule (eg CD2 or CD3), or an Fc receptor (FcγR), eg FcγRI (CD64) for IgG. ) And FcγRII (CD32) and FcγRIII (CD16) can be combined with the arms to focus on cellular defense mechanisms against B cells. Bispecific antibodies can also be used to target cytotoxic agents to B cells. These antibodies have CD20-binding arms and arms that bind cytotoxic agents (eg, saporin, anti-interferon-α, vinca alkaloids, lysine A chain, methotrexate or radioisotope hapten). Bispecific antibodies can be prepared as full length antibodies or antibody fragments (eg, F (ab ') 2 bispecific antibodies). [98] Methods of making bispecific antibodies are known in the art. Conventional methods for producing full-length bispecific antibodies are based on the expression of two immunoglobulin heavy-light chain pairs with different specificities together [Milstein et al., Nature, 305: 537-539 (1983)]. . Due to the random classification of immunoglobulin heavy and light chains, these hybridomas generate potential mixtures of ten different antibody molecules, only one of which has the correct bispecific structure. Purification of these exact molecules, usually done by affinity chromatography steps, is a rather cumbersome process and the product yield is low. Similar procedures are described in WO 93/08829; and Traunecker et al., EMBO J., 10: 3655-3659 (1991). [99] According to different methods, antibody variable domains with the desired binding specificities (antibody-antigen binding sites) are fused to immunoglobulin constant domain sequences. This fusion preferably uses an immunoglobulin heavy chain constant domain comprising at least a portion of the hinge, CH2 and CH3 regions. It is preferred to have a first heavy chain constant region (CH1) containing a site necessary for light chain binding, present in one or more of these fusions. The immunoglobulin heavy chain fusions are encoded, and if desired, the DNA encoding the immunoglobulin light chains is inserted into separate expression vectors and these are transfected together into a suitable host organism. This provides great flexibility in controlling the mutual ratios of these three polypeptide fragments in embodiments in which unequal proportions of the three polypeptide chains used in the construction provide optimal yields. However, if the expression of two or more polypeptide chains in equal proportions yields high yields, or if these proportions are of no particular significance, the coding sequences for two or all of the three polypeptide chains are inserted into one expression vector. It is possible to do [100] In a preferred embodiment of this method, the bispecific antibody comprises a hybrid immunoglobulin heavy chain with a first binding specificity on one arm and a hybrid immunoglobulin heavy chain-light chain pair on the other arm (provides a second binding specificity). It consists of. Such asymmetric structures have been found to facilitate the separation of the desired bispecific compounds from undesired immunoglobulin chain combinations, since the immunoglobulin light chain is only present in half of these bispecific molecules. One separation method is provided. This method is described in WO 94/04690. For further details on bispecific antibody production, see, eg, Suresh et al., Methods in Enzymology, 121: 210 (1986). According to another method described in US Pat. No. 5,731,168, the interface between antibody molecule pairs can be genetically engineered to maximize the percentage of heterodimer recovered from recombinant cell culture. Preferred interfaces include at least a portion of the C H 3 domain of the antibody constant domains. In this method, one or more small amino acid side chains from the interface of the first antibody molecule are replaced with larger side chains (eg tyrosine or tryptophan). Compensating large side chain (s) with "cavities" of the same or similar size occurs on the interface of the second antibody molecule by replacing large amino acid side chains with smaller amino acids (eg alanine or threonine). This provides a mechanism for increasing the yield of heterodimer relative to other undesirable end products (eg homodimer). [101] Bispecific antibodies include crosslinked or "heteroconjugate" antibodies. For example, one of the antibodies in the heteroconjugate can be coupled to avidin and the other antibody can be coupled to biotin. Such antibodies are proposed, for example, to target immune system cells with unwanted cells (US Pat. No. 4,676,980) and to treat HIV infection (WO 91/00360, WO 92/200373 and EP 03089). Heteroconjugate antibodies can be prepared using conventional crosslinking methods. Suitable crosslinkers are well known in the art with many crosslinking techniques and are described in US Pat. No. 4,676,980. [102] Techniques for generating bispecific antibodies from antibody fragments have also been reported in the literature. For example, bispecific antibodies can be prepared using chemical crosslinking. Brennan et al., Science, 229: 81 (1985) describe the process of cleaving intact antibodies with proteolytic enzymes to produce F (ab ') 2 fragments. These fragments are reduced in the presence of dithiol binder former sodium arsenite to stabilize adjacent dithiols and prevent intermolecular disulfide formation. The Fab 'fragment thus generated is then converted to thionitrobenzoate (TNB) derivatives. Subsequently, one of the Fab'-TNB derivatives is converted back to Fab'-thiol by reduction with mercaptoethylamine, which is mixed with an equimolar amount of other Fab'-TNB derivatives to form a bispecific antibody. The bispecific antibodies thus produced can be used as substances for the selective immobilization of enzymes. [103] Due to recent scientific developments, this. The process of directly recovering Fab'-SH fragments from E. coli has been facilitated, and such fragments can be chemically coupled to form bispecific antibodies. Shalaby et al., J. Exp. Med., 175: 217-225 (1992) describe the production of a fully humanized bispecific antibody F (ab ') 2 molecule. Each Fab 'fragment Secreted separately from E. coli and chemically coupled directly in vitro to form bispecific antibodies. The bispecific antibody thus formed was able to bind cells overexpressing ErbB2 receptor and normal human T cells as well as induce lytic activity of human cytotoxic lymphocytes against human breast tumor targets. [104] Various techniques for preparing and isolating bispecific antibody fragments directly from recombinant cell culture have also been described. For example, leucine zippers were used to generate bispecific antibodies (Kostelny et al., J. Immunol., 148 (5): 1547-1553 (1992)). The leucine zipper peptides from Fos and Jun proteins were linked to the Fab 'portion of two different antibodies by gene fusion. This antibody homodimer was reduced in the hinge region to form a monomer and then reoxidized to form an antibody heterodimer. This method can also be used to generate antibody homodimers. Hollinger et al., Proc. Natl. Acad. Sci. USA, 90: 6444-6448 (1993), provides another mechanism for making bispecific antibody fragments. The fragment comprises a heavy chain variable domain (V H ) linked to the light chain variable domain (V L ) by a linker that is too short to pair between two domains on the same chain. Thus, the focus is on forming two antigen binding sites by pairing the V H and V L domains of one fragment with the complementary V L and V H domains of another fragment. Another method of making bispecific antibody fragments by using single chain Fv (sFv) dimers is also reported (Grubber et al., J. Immunol., 152: 5368 (1994)). [105] Bivalent or higher antibodies are also contemplated. For example, trispecific antibodies can be prepared [Tutt et al., J. Immunol. 147: 60 (1991). [106] III. Other Variants of Conjugates and Antagonists [107] Antagonists used in the methods of the invention or included in the products herein optionally bind to cytotoxic agents. [108] Chemotherapeutic agents useful for the production of such antagonist-cytotoxic agent conjugates are described above. [109] Also included herein are conjugates with antagonists and one or more small molecule toxins such as calicheamicin, maytansine (US Pat. No. 5,208,020), tricotene and CC1065. In one preferred embodiment of the invention, the antagonist is bound to one or more maytansine molecules (eg, about 1 to about 10 maytansine molecules per antagonist molecule). Maytansine can be converted, for example, to May-SS-Me, which can be reduced to May-SH3, and can react with a modified antagonist to form a maytansinoid-antagonist conjugate [Chari et al. Cancer Research 52: 127-131 (1992). [110] Alternatively, the antagonist is bound to one or more calicheamicin molecules. Antibiotics of the calicheamicin family that can produce double stranded DNA are destroyed at concentrations below picomolar. Structural homologues of calicheamicin that may be used include, but are not limited to, γ 1 I , α 2 I , α 3 I , N-acetyl-γ 1 I , PSAG and θ 1 I [Hinman et al. Cancer Research 53: 3336-3342 (1993) and Lode et al. Cancer Research 58: 2925-2928 (1988). [111] Enzymatically active toxins and fragments thereof that can be used include diphtheria A chain, unbound active fragment of diphtheria toxin, exotoxin A chain (derived from Pseudomonas aeruginosa), lysine A chain, abrine A chain, modesin A chain , Alpha-sarsine, alleletes fordyi protein, diantine protein, phytoca americana protein (PAPI, PAPII and PAP-S), Momordica Charantia inhibitor, cursin, crotin, sapaonaria office Lys inhibitors, gelonin, mitogeline, restrictocin, phenomycin, enomycin and tricortesene [1993. 10. WO 93/21232, published 28.]. [112] The invention also includes antagonists bound to compounds having nuclease activity (eg ribonuclease or DNA endonucleases such as deoxyribonuclease; DNase). [113] Various radioisotopes can be used in the production of radioconjugate antagonists. Examples are radioisotopes of At 211 , I 131 , I 125 , Y 90 , Re 186 , Re 188 , Sm 153 , Bi 212 , P 32 and Lu. [114] Conjugates of antagonists with cytotoxic agents include various difunctional protein coupling agents, such as N-succinimidyl-3- (2-pyridyldithiol) propionate (SPDP), succinimidyl-4- (N-maleimidomethyl) cyclohexane-1-carboxylate, iminothiolane (IT), difunctional derivatives of imidoesters (e.g. dimethyl adipimidate HCL), active esters (e.g. dissuccinimidyl suve ), Aldehydes (e.g. glutaraldehyde), bis-azido compounds (e.g. bis (p-azidobenzoyl) hexanediamine), bis-diazonium derivatives (e.g. bis- (p-diazoniumbenzoyl)- Ethylenediamine), diisocyanates (eg tolyene 2,6-diisocyanate) and bis-active fluorine compounds (eg 1,5-difluoro-2,4-dinitrobenzene). For example, lysine immunotoxins can be prepared as described in Vitetta et al., Scence 238: 1098 (1987). Carbon-14-labeled 1-isothiocyanatobenzyl-3-methyldiethylene triaminepentaacetic acid (MX-DPTA) is an exemplary chelating agent for binding of radionucleotides to antagonists (see WO 94/11026). . The linker may be a “cleavable linker” that facilitates the release of cytotoxic drugs in the cell. For example, acid labile linkers, peptidase-sensitive linkers, dimethyl linkers or disulfide containing linkers can be used (see Chari et al., Cancer Research 52: 127-131). [115] Alternatively, fusion proteins comprising antagonists and cytotoxic agents can be prepared, eg, by recombinant techniques or peptide synthesis. [116] In another embodiment, the antagonist may be bound to a "receptor" (eg, streptavidin) for use in tumor pretargeting, wherein the antagonist-receptor conjugate is administered to the patient and then rinsed off from the circulatory system using a washing agent. After removal of the bound conjugate, a "ligand" (eg avidin) that is bound to a cytotoxic agent (eg a radionucleotide) is administered. [117] The antagonists of the invention can also be combined with prodrug activating enzymes that convert prodrugs (eg peptidyl chemotherapeutic agents; see WO 81/01145) into active anticancer agents [eg, WO 88/07378 and US patents. 4,975,278]. [118] Enzyme components of such conjugates include enzymes that can act on prodrugs in a manner that converts them into more active and cytotoxic forms. [119] Enzymes useful in the methods of the invention include alkaline phosphatase useful for converting phosphate containing prodrugs into free drugs; Arylsulfatase useful for converting sulfate-containing prodrugs into free drugs; Cytosine deaminase useful for converting non-toxic 5-fluorocytosine into the anticancer agent, 5-fluorouracil; Proteases useful for converting peptide-containing prodrugs into free drugs, such as seratia proteases, thermolysine, subtilisin, carboxypeptidase and cathepsins (eg cathepsin B and L); D-alanylcarboxypeptidase useful for converting prodrugs containing D-amino acid substituents; Carbohydrate cleavage enzymes useful for converting glycosylated prodrugs into free drugs, such as β-galactosidase and neuraminidase; β-lactamase useful for converting drugs derivatized with β-lactams into free drugs; And penicillin amidase, such as penicillin V amidase or penicillin G amidase, useful for converting drugs derivatized with phenoxyacetyl or phenylacetyl groups in amine nitrogen to free drugs, respectively. . Alternatively, antibodies with enzymatic activity, also known in the art as "abzyme", can be used to convert prodrugs of the invention to free active drugs [Massey, Nature 328: 457-458 ( 1987). Antagonist-Abzyme conjugates for delivering azyme to tumor cell populations can be prepared as described herein. [120] Enzymes of the invention can be covalently linked to the antagonist by techniques well known in the art, such as using the heterobifunctional crosslinking reagents discussed above. Alternatively, a fusion protein comprising at least the antigen binding region of the antagonist of the invention linked to at least the functionally active portion of the enzyme of the invention can be prepared using recombinant DNA techniques well known in the art [Neuberger et al. , Nature 312: 604-608 (1984). [121] Other variations of the antagonist are also included herein. For example, the antagonist can be linked to one of a variety of nonproteinaceous polymers, such as polyethylene glycol, polypropylene glycol, polyoxyalkylene, or copolymers of polyethylene glycol and polypropylene glycol. [122] Antagonists disclosed herein may also be formulated as liposomes. Liposomes containing antagonists are known in the art, such as, for example, Epstein et al., Proc. Natl. Acad. Sci. USA, 82: 3688 (1985); Hwang et al., Proc. Natl. Acad. Sci. USA, 77: 4030 (1980); U.S. Patents 4,485,045 and 4,544,545; And WO 97/38731 (published Oct. 23, 1997). Liposomes with enhanced circulation time are disclosed in US Pat. No. 5,013,556. [123] Particularly useful liposomes can be produced by reverse phase evaporation methods using lipid compositions comprising phosphatidylcholine, cholesterol and PEG-derivatized phosphatidylethanolamine (PEG-PE). Liposomes are extruded through filters of constant pore size to produce liposomes with the desired diameter. Fab ′ fragments of the antibodies of the invention are described in Martin et al., J. Biol. Chem. 257: 286-288 (1982). Optionally, chemotherapeutic agents are included in these liposomes [Gabizon et al., J. National Cancer Inst. 81 (19) 1484 (1989). [124] Amino acid sequence modification (s) of the protein or peptide antagonists described herein are contemplated. For example, it may be desirable to improve the binding affinity and / or other biological properties of the antagonist. Amino acid sequence variants of the antagonist are prepared by introducing appropriate nucleotide changes into the antagonist nucleic acid, or by peptide synthesis. Such modifications include, for example, deletions from residues within the amino acid sequence of the antagonist, and / or insertions into and / or substitutions of residues. Any combination of deletions, insertions, and substitutions to reach the final construct is possible, provided that the final construct possesses the desired characteristics. Such amino acid changes may also alter the post-translational process of the antagonist, such as changing the number or location of glycosylation sites. [125] Methods useful for identifying specific residues or regions of antagonists, which are preferred positions for mutagenesis, are called "alanine scanning mutagenesis" described in Cunningham and Wells, Science, 244: 1081-1085 (1989). Here, the target residues or groups of residues are identified (eg charged residues such as arg, asp, his, lys and glu) and replaced with neutral or negatively charged amino acids (most preferably alanine or polyalanine) This affects the interaction of the amino acid with the antigen. Subsequently, amino acid moieties that exhibit functional sensitivity to such substitutions are defined by introducing other variants or other variants into or in place of the substitution sites. Thus, while sites for introducing amino acid sequence variations are intended, the nature of the mutations need not be intended. For example, to analyze the performance of mutations at a given site, alanine scanning or random mutagenesis is performed at the target codon or region and the expressed antagonist variants are screened for the desired activity. [126] Amino acid sequence inserts include amino- and / or carboxyl-terminal fusions of polypeptides of length from one residue to several hundred or more residues, as well as intrasequence inserts of single or multiple amino acid residues. Examples of terminal inserts include antagonists with N-terminal methionyl residues or antagonists fused to a cytotoxic polypeptide. Other insertional variants of the antagonist molecule include fusions with the N- or C-terminus of the antagonist to a polypeptide or enzyme (eg, an enzyme for ADEPT) that increases the serum half-life of the antagonist. [127] Another type of variant is an amino acid substitution variant. These variants have one or more amino acid residues in the antagonist molecule, replaced with different residues. Sites of greatest interest for substitutional mutagenesis include hypervariable regions, but FR variations are also contemplated. Conservative substitutions are shown in Table 1 under the heading of "preferred substitutions." If such substitutions result in a change in biological activity, more substantial changes, as referred to in Table 1 as “exemplary substitutions” or further described below with reference to the amino acid class, can be introduced and the product can be screened. [128] Original residues Exemplary Substituents Preferred substituent Ala (A) val; leu; ile val Arg (R) lys; gln; asn lys Asn (N) gln; his; asp; lys; arg gln Asp (D) glu; asn glu Cys (C) ser; ala ser Gln (Q) asn; glu asn Glu (E) asp; gln asp Gly (G) ala ala His (H) asn; gln; lys; arg arg Ile (I) leu; val; met; ala; phe; Norleucine leu Leu (L) Norleucine; ile; val; met; ala; phe ile Lys (K) arg; gln; asn arg Met (M) leu; phe; ile leu Phe (F) leu; val; ile; ala; tyr tyr Pro (P) ala ala Ser (S) Thr thr Thr (T) Ser ser Trp (W) tyr; phe tyr Tyr (Y) trp; phe; thr; ser phe Val (V) ile; leu; met; phe; ala; Norleucine leu [129] Substantial changes in biological properties include (a) their effect on maintaining the structure of the polypeptide backbone, eg, sheet or helical conformation, within the substitution region, (b) the charge or hydrophobicity of the molecule at the target site. Their effect in retaining or (c) their effect in retaining the bulk of the side chain is carried out by selecting significantly different substitutions. Naturally occurring residues are divided into the following groups based on common side chain properties: [130] (1) hydrophobic: norleucine, met, ala, val, leu, ile; [131] (2) neutral hydrophilic: cys, ser, thr; [132] (3) acidic: asp, glu; [133] (4) basic: asn, gln, his, lys, arg; [134] (5) residues affecting chain orientation: gly, pro; And [135] (6) aromatic: trp, tyr, phe. [136] Non-conservative substitutions will be made by replacing members of one of these classes with another class. [137] Any cysteine residue that is not involved in maintaining the proper conformation of the antagonist can generally be substituted with serine to improve the oxidative stability of the molecule and to prevent abnormal crosslinking. Conversely, cysteine bond (s) can be added to the antagonist to improve its stability (especially when the antagonist is an antibody fragment such as an Fv fragment). [138] Particularly preferred types of substitutional variants involve replacing one or more hypervariable region residues of a parent antibody (eg, a humanized or human antibody). In general, the variant (s) thus produced, selected for further development, will have improved biological properties compared to the parent antibody that produced them. A convenient way to generate such substitutional variants is the affinity maturation process using phage display. Briefly, several hypervariable region sites (eg 6-7 sites) are mutated to produce all possible amino acid substitutions at each site. The resulting antibody variants are displayed in a monovalent fashion from filamentary phage particles as a fusion with the gene III product of M13 packaged in each particle. Such phage displayed variants are then screened for their biological activity (eg binding affinity) as described herein. To identify candidate hypervariable region sites suitable for modification, alanine scanning mutagenesis can be performed to identify hypervariable region residues that substantially contribute to antigen binding. Alternatively or additionally, it may be advantageous to identify the contact point between the antibody and antigen by analyzing the crystal structure of the antigen-antibody complex. Such contact residues and neighboring residues are candidates for substitution according to the techniques reviewed herein. Once such variants are generated, a panel of variants can be screened as described herein, and antibodies exhibiting excellent properties in one or more related assays can be selected for further development. [139] Other types of amino acid variants of the antagonist alter the glycosylation pattern inherent in the antagonist. By change is meant the deletion of one or more carbohydrate residues found in the antagonist and / or the addition of one or more glycosylation sites that are not present in the antagonist. [140] Glycosylation of polypeptides is typically either N-linked or O-linked. N-linked refers to a carbohydrate moiety attached to the side chain of an asparagine moiety. Tripeptide sequences asparagine-X-serine and asparagine-X-threonine, where X is any amino acid except proline, are recognition sequences for enzymatic attachment of carbohydrate moieties to the asparagine side chains. Thus, the presence of one of these tripeptide sequences in a polypeptide creates a potential glycosylation site. O-linked glycosylation means attaching one of the sugars N-acetylgalactosamine, galactose or xylose to hydroxyamino acids, most commonly serine or threonine, but 5-hydroxyproline or 5-hydroxylysine You can also use [141] The addition of glycosylation sites to the antagonist is conveniently accomplished by changing the amino acid sequence to contain one or more of the above mentioned tripeptide sequences (for N-linked glycosylation sites). Such changes may also be made by adding or replacing one or more serine or threonine residues with the sequence of the original antibody (for O-linked glycosylation sites). [142] Nucleic acid molecules encoding amino acid sequence variants of the antagonist are prepared by a variety of methods known in the art. These methods include isolation methods from natural sources (for naturally occurring amino acid sequence variants), or oligonucleotide mediated (or site specific) mutagenesis, PCR mutagenesis and cassette mutations of variants or non-variant variants of previously prepared antagonists. Production methods by induction are included, but are not limited thereto. [143] For example, it may be desirable to alter the function of the antagonist so that the antigen-dependent cell mediated cytotoxicity (ADCC) and / or complement dependent cytotoxicity (CDC) of the antagonist of the invention is enhanced. This can be accomplished by introducing one or more amino acid substitutions into the Fc region of the antibody antagonist. Alternatively or additionally, the introduction of cysteine residue (s) into the Fc region can form interchain disulfide bonds in this region. The resulting homodimeric antibodies may have improved introgression capacity and / or increased complement mediated cell death and antibody-dependent cellular cytotoxicity (ADCC) [Caron et al., J. Exp Med. 176: 1191-1195 (1992) and Shopes, B. J. Immunol. 148: 2918-2922 (1992). Homodimeric antibodies with improved antitumor activity can also be prepared using heterobifunctional crosslinkers, as described in Wolff et al., Cancer Research 53: 2560-2565 (1993). Alternatively, by genetically engineering an antibody with a double Fc region, it may have improved complement lysis and ADCC ability (Stevenson et al., Anti-Cancer Drug Design 3: 219-230 (1989)). [144] To increase the serum half-life of antagonists, salvage receptor binding epitopes can be incorporated into antagonists (especially antibody fragments), as described, for example, in US Pat. No. 5,739,277. As used herein, the term “rescue receptor binding epitope” refers to the Fc region of an IgG molecule (eg IgG 1 , IgG 2 , IgG 3 , or IgG 4 ) that is involved in increasing the serum half-life of an IgG molecule in vivo. Say epitope. [145] IV. Pharmaceutical formulation [146] Therapeutic formulations of antagonists used in accordance with the present invention may be prepared by any of the following pharmaceutically acceptable carriers, excipients or stabilizers: Remington's Pharmaceutical Sciences 16th edition, Osol, A. Ed. (1980), for storage in the form of a lyophilized formulation or aqueous solution. Acceptable carriers, excipients, or stabilizers are nontoxic to recipients at the dosages and concentrations employed, and include buffers such as phosphate, citrate and other organic acids; Antioxidants including ascorbic acid and methionine; Preservatives (e.g., octadecyldimethylbenzyl ammonium chloride; hexamethonium chloride; benzalkonium chloride; benzethonium chloride; phenol, butyl or benzyl alcohol; alkyl parabens such as methyl or propyl paraben; catechol; resorcinol Cyclohexanol; 3-pentanol; and m-cresol); Low molecular weight (less than about 10 residues) polypeptides; Proteins such as serum albumin, gelatin or immunoglobulins; Hydrophilic polymers such as polyvinylpyrrolidone; Amino acids such as glycine, glutamine, asparagine, histidine, arginine or lysine; Monosaccharides, disaccharides and other carbohydrates, including glucose, mannose, or dextrins; Chelating agents such as EDTA; Sugars such as sucrose, mannitol, trehalose or sorbitol; Salt-forming counter-ions such as sodium; Metal complexes such as Zn-protein complexes; And / or nonionic surfactants such as TWEEN ™, PLURONICS ™ or polyethylene glycol (PEG). [147] Exemplary anti-CD20 antibody formulations are described in WO98 / 56418, the content of which is expressly incorporated herein. This publication discloses 40 mg / mL rituximab, 25 mM acetate, 150 mM trehalose, 0.9% benzyl alcohol, 0.02% polysorbate 20 (pH 5.0) with a minimum half-life of 2 years at 2-8 ° C. A liquid formulation for several administrations is described. Other anti-CD20 formulations of interest include 10 mg / mL rituximab in 9.0 mg / mL sodium chloride, 7.35 mg / mL sodium citrate dihydrate, 0.7 mg / mL polysorbate 80 and sterile water for injection (pH 6.5). [148] Lyophilized formulations suitable for subcutaneous administration are described in WO 97/04801. [149] The formulations herein may also contain one or more active compounds required for the particular condition to be treated, preferably compounds with complementary activity that do not adversely affect each other. For example, cytotoxic agents, chemotherapeutic agents, cytokines or immunosuppressive agents (e.g., agents that act on T cells, such as cyclosporin or antibodies that bind to T cells, e.g. agents that bind to LFA-1) It may be desirable to further include it in the formulation. The effective amount of such an agent depends on the amount of antagonist present in the formulation, the type of disease or disorder or the type of treatment, and other factors discussed above. They are generally used at the same dosage and route of administration as previously used or at about 1-99% of the previously used dosage. [150] The active ingredient can also be used in a colloidal drug delivery system (eg liposomes, albumin microspheres, microemulsions, nano-particles and nanocapsules) or macroemulsion by coacervation techniques or by interfacial polymerization, eg For example, it may be entrapped in microcapsules prepared from hydroxymethylcellulose or gelatin-microcapsules and poly (methylmethacrylate) microcapsules, respectively. Such techniques are described in Remington's Pharmaceutical Sciences 16th edition, Osol, A. Ed. (1980). [151] Sustained release formulations may be prepared. Suitable examples of sustained release formulations include semipermeable matrices of solid hydrophobic polymers containing the antagonist, which matrices are in the form of shaped articles, eg films, or microcapsules. Examples of sustained release matrices include polyesters, hydrogels (eg, poly (2-hydroxyethyl-methacrylate) or poly (vinyl alcohol)), polylactide (US Pat. No. 3,773,919), L- Copolymers of glutamic acid with γethyl-L-glutamate, non-degradable ethylene-vinyl acetate, degradable lactic acid-glycolic acid copolymers, such as LUPRON DEPOT ™ (lactic acid-glycolic acid copolymer and leuprolide acetate Injectable microspheres), and poly-D-(-)-3-hydroxybutyric acid. [152] The formulations to be used for in vivo administration must be sterile. This is readily accomplished by filtration through sterile filtration membranes. [153] V. Treatment with antagonists [154] Antagonists that bind CD20 can be used to block the immune response to foreign antigens in a mammal, where the mammal does not suffer from malignant tumors. Preferably the antagonist comprises an anti-CD20 antibody. In one embodiment the antibody does not bind to a cytotoxic agent, and in other embodiments the antibody binds to a cytotoxic agent (eg, Y2B8 or 131 I-B1). [155] The mammal to be treated herein can be exposed to both antagonists that bind CD20, and to other therapeutic agents, such as therapeutic agents that are immunogenic in mammals. In this embodiment, the antagonist may block the immune response to the therapeutic agent in the mammal being treated with it. The therapeutic advantages may include the blocking of antibody coated cell removal by the spleen. The therapeutic agent treats a disease or disorder that can be administered to a mammal in a therapeutically effective amount to benefit from administration of the therapeutic agent. In this embodiment, the therapeutic and antagonist may be administered to the mammal at the same time or separately in any order. Thus, the antagonist may be administered to a mammal and then the therapeutic agent may be administered, or the antagonist may be administered after the therapeutic agent is administered to the mammal. [156] Thus, antagonists that bind CD20 can be used to treat graft-versus-host or host-versus-graft disease in mammals and / or reduce the mammal's susceptibility to receiving tissue. [157] For the various instructions disclosed herein, a composition comprising an antagonist that binds to CD20 is formulated in a manner consistent with good medical practice and determined and administered at a dosage. Factors contemplated herein include, but are not limited to, the particular disease or condition to be treated, the particular mammal to be treated, the clinical symptoms of the individual patient, the cause of the disease or condition, the site to which the agent is delivered, the method of administration, the schedule of administration, and the known to those skilled in the medical arts. Other factors are included. The therapeutically effective amount of the antagonist to be administered will be determined in consideration of these factors. [158] The general suggestion is that the therapeutically effective amount of an antagonist administered orally per dose is about 0.1-20 mg / kg (body weight) / day and the typical initial amount of antagonist used is about 2-10 mg / kg. [159] Preferred antagonists are antibodies which are not bound to cytotoxic agents, for example antibodies such as RITUXAN®. Appropriate doses of the unbound antibody are, for example, from about 20 mg / m 2 to about 1000 mg / m 2 . In one embodiment, the dosage of the antibody is different from the dosage currently recommended for RITUXAN®. For example, substantially less than 375 mg / m 2 antibody, eg, about 20 mg / m 2 to about 250 mg / m 2 , eg, about 50 mg / m 2 to about 200 mg / m 2 Is administered to the patient at least once. [160] Also, there the antibodies of the initial dose can be administered once or more subsequent dose after administration of once or more, mg / m 2 doses of the antibody in the subsequent dose herein is the antibody in the initial dose mg / m 2 Dose exceeded. For example, the initial dose can be about 20 mg / m 2 to about 250 mg / m 2 (eg, about 50 mg / m 2 to about 200 mg / m 2 ) and the subsequent dose is about 250 mg / m 2 to about 1000 mg / m 2 . [161] However, as noted above, this suggested amount of antagonist requires great care in terms of treatment. The key factor in selecting the appropriate dosage and in planning the administration is the result obtained as described above. For example, relatively high dosages may be required for early treatment of advanced and acute diseases. To obtain the most effective results depending on the disease or condition, the antagonist is administered as close as possible to the first sign, diagnosis, aspect or occurrence of the disease or condition, or while the disease or condition is alleviating. [162] Antagonists are administered by any suitable method, including parenteral, subcutaneous, intraperitoneal, pulmonary and intranasal, and intralesional administration if desired for local immunosuppression. , Intraperitoneal or subcutaneous administration. In addition, the antagonist may be appropriately administered in a pulse irrigation, for example, with a reduced dose of the antagonist. Preferably, administration is by injection, most preferably intravenous or subcutaneous injection, the choice between intravenous injection and subcutaneous injection depends in part on whether the administration period is short or long term. [163] Other compounds such as cytotoxic agents, chemotherapeutic agents, immunosuppressants, and / or cytokines can be administered with the antagonists herein. Such co-administration methods include administration together using separate or single pharmaceutical formulations, and successive administrations in sequence, in which both active agents (or all active agents) are simultaneously active in their biological activities. It is desirable to have a period of time that appears. [164] In addition to administering a protein antagonist to a patient, the present application also contemplates administering the antagonist by gene therapy. Such administration of nucleic acid encoding an antagonist is included in the expression "administering a therapeutically effective amount of an antagonist." For example, see WO96 / 07321, published March 14, 1996, regarding the use of gene therapy to generate intracellular antibodies. [165] There are two main ways of introducing nucleic acids (optionally included in a vector) into a patient's cells in vivo and ex vivo. For in vivo delivery, nucleic acids are usually injected directly into the site of a patient in need of an antagonist. For ex vivo treatments, cells of a patient are removed and nucleic acids are introduced into these isolated cells, and the mutated cells are administered directly to the patient or encapsulated in a porous membrane implanted, for example, into the patient (eg See, for example, US Pat. Nos. 4,892,538 and 5,283,187. There are a variety of techniques that can be used to introduce nucleic acids into viable cells. This technique depends on whether the nucleic acid is delivered to cells cultured in vitro or to cells in vivo in the host of interest. Techniques suitable for delivering nucleic acids into mammalian cells in vitro include the use of liposomes, electroporation, microinjection, cell fusion, DEAE-textlan, calcium phosphate precipitation, and the like. A vector commonly used for ex vivo delivery of genes is a retrovirus. [166] Currently preferred in vivo nucleic acid delivery techniques include transfections using viral vectors (eg, adenoviruses, herpes simplex I virus or adeno-associated viruses) and lipid based systems (lipids useful for lipid-mediated delivery of genes, for example). DOTMA, DOPE and DC-Chol). In some cases, it is desirable to provide a nucleic acid source with a substance that targets the target cell, such as cell membrane proteins or antibodies specific for the target cell, ligands for receptors on the target cell, and the like. When liposomes are used, proteins that bind to cell surface membrane proteins associated with endocytosis are used for targeting and / or, for example, capsid proteins or fragments thereof that are friendly to particular cell types, endocytosis cycles. It can be used to facilitate the influx of antibodies to proteins entering the cell in the cell, and proteins that enhance intracellular half-life by targeting intracellular localization. Techniques for receptor-mediated endocytosis are described, for example, in Wu et al., J. Biol. Chem. 262: 4429-4432 (1987); and Wagner et al., Proc. Natl. Acad. Sci. USA 87: 3410-3414 (1990). See Anderson et al., Science 256: 808-813 (1992) for details on currently known gene marking and gene therapy protocols. See also WO 93/25673 and references cited therein. [167] VI. product [168] In another aspect of the present invention there is provided a product containing a substance useful for the treatment of a disease or condition described above. Such products include containers and labels or package inserts on or associated with such containers. Suitable containers include, for example, bottles, vials, syringes and the like. Such containers can be formed from various materials such as glass or plastic. The container holds or contains a composition effective to treat the disease or condition in question and may have a sterile doorway (eg, such a container has an intravenous solution bag with a stopper that can be penetrated with a hypodermic needle). Or vials). At least one active agent in such a composition is an antagonist that binds to CD20. The label or package insert indicates that the composition is used for blocking the immune response to foreign antigens and / or for treating a disease or condition described herein. In addition, the product may further comprise a second container comprising a pharmaceutically acceptable dilution buffer such as sterile water for injection (BWFI), phosphate buffered saline, Ringer's solution and dextrose solution. In one embodiment, the second container holds or contains a composition wherein the active agent in the composition is a therapeutic agent. The package insert may indicate that the patient should be treated with the two compositions of this embodiment of the invention. The product may further include other materials desirable from a commercial and user standpoint, including other buffers, diluents, filters, needles, and syringes. [169] The detailed description of the invention is illustrated by the following non-limiting examples. The disclosures of all documents mentioned in the specification are to be included in this specification. [170] <Example 1> [171] Blocking immune response to therapeutic proteins [172] In this example, anti-CD20 antibodies are used to block the immune response to certain therapeutic proteins, ie megakaryocyte growth and development factors (also known as MGDF, thrombopoietin or Mpl ligand). In particular, the pegylated form of recombinant human MGDF (PEG-rHuMGDF) has been reported to generate neutralizing antibodies in cancer patients and platelet donors. Administration of the anti-CD20 antibodies disclosed herein will mitigate the immune response to PEG-rHuMGDF, in particular humoral immunity. [173] PEG-rHuMGDF is prepared as described in US Pat. No. 5,795,569, published August 18, 1998, the content of which is expressly incorporated herein. PEG-rHuMGDF is a human E. coli with a single polyethylene glycol (PEG) attached to an N-terminal α-amino group. Consists of amino acids 1-163 (numbered from the front of the mature protein) of E. coli derived MGDF. [174] For example, a dose suitable for increasing platelet count in a patient suffering from thrombocytopenia due to chemotherapy or radiotherapy, for example, 0.1 to 1000 micrograms of MGDF per kg body weight is administered to the patient. MGDF therapy is optionally combined with the administration of one or more other cytokines such as erythropoietin (EPO), interleukin-3 (IL-3) and granulocyte megakaryocyte colony stimulating factor (GM-CSF). [175] Anti-MGDF antibody titers in these treated patients are monitored by appropriate assays such as antibody titer enzyme-linked immunosorbent assay (ELISA). Thereafter, patients showing a low titer immune response to MGDF are candidates for treatment with an anti-CD20 antibody, such as RITUXAN®. The anti-CD20 antibody may be administered after treatment with MGDF, concurrently with treatment with MGDF or prior to treatment with MGDF. Suitable dosages of anti-CD20 antibodies are 375 mg / m 2 (administered by four irrigations per week). However, smaller dosages may also be administered, for example from about 50 to about 250 mg / m 2 . Administration of an anti-CD20 antibody to a patient will inhibit or reduce the formation of anti-MGDF antibodies to acceptable levels in patients treated with both MGDF and anti-CD20 as described above. Therefore, for protein drugs with very high therapeutic values and known immunogenicity, the co-administration of the anti-CD20 antibodies described herein will treat the immunogenic side effects that occur when the drug is administered to the patient. [176] <Example 2> [177] Blocking Immune Responses to Gene Therapy Virus Vectors [178] The ability of El, E-deletion, replication-immune recombinant adenoviruses to deliver therapeutic genes in vivo was investigated. New vectors with additional deletions in the E2a or E4 region have been developed (Christ et al., Immunol. Let. 57: 19-25 (1997)). Despite the deletion of these viral regions, early and late viral genes are expressed at low levels in vivo. Production of anti-adenovirus antibodies, cellular immune responses, as well as early nonspecific removal of the vector, is a barrier to successful gene therapy. To inhibit or reduce the production of neutralizing antibodies against adenoviruses, anti-CD20 antibodies (eg, RITUXAN®) are administered to the gene therapy patients described herein. [179] For example, cystic fibrosis patients are treated with a replication-impaired adenovirus expressing human cystic fibrosis transmembrane translocation modulator (CFTR) (Bellon et al., Human Gene Therapy 8: 15-25 (1997)). Appropriate doses of the CFTR gene therapy vector (viral plaque forming unit, defined as the term pfu) are administered by aerosol to achieve expression of CFTR in the lungs (eg, about 10 7 to about 10 9 pfu). Anti-adenovirus antibodies in a patient can be detected by ELISA, immunofluorescence, and / or complement immobilization. In patients in which anti-adenovirus antibodies are detected, anti-CD20 antibodies (eg chimeric 2H7; US Pat. No. 5,677,180) may optionally be replaced with other immunosuppressive drugs (eg cyclophosphamide, FK506, or T cells). Monoclonal antibodies that block the receptor or co-stimulatory pathway), prior to, concurrent with, or after re-administration of the gene therapy vector. Suitable dosages of anti-CD20 antibodies are 375 mg / m 2 (four irrigations per week). Administration of anti-CD20 antibodies reduces or eliminates the patient's immune response (reduces anti-adenovirus antibody production) to facilitate successful retreatment of successful gene therapy. [180] <Example 3> [181] Blocking immune response to grafts [182] Anti-CD20 antibodies are used as part of a combination immunosuppressive regimen for the prevention of acute rejection. In such cases, an anti-CD20 antibody, for example RITUXAN®, is an anti-CD as part of a series of combination dosing regimens comprising agents acting on T cells, such as cyclosporin, corticosteroids, mycophenolate mofetil Administration is performed during tissue transplantation with or without an IL2 receptor antibody. Thus, anti-CD20 antibodies are considered as part of the induction dosing regimen used with chronic immunosuppression. Anti-CD20 antibodies may contribute to the prevention of allogeneic rejection by inhibiting allogeneic antigen production and / or affecting presentation of alloantigens through depletion of antigen-presenting cells. [183] The therapy may require irrigation 4 times per week (375 mg / m 2 ) prior to or during tissue transplantation. Appropriate doses of other immunosuppressive agents are as follows: cyclosporin (5 mg / kg / day); Corticosteroids (1 mg / kg, gradually reduced); Mycophenolate mofetil (administration of 1 gram twice a day); And anti-IL2 receptor antibody (1 mg / kg, 5 reciprocations per week). In addition, anti-CD20 antibodies include other induced immunosuppressive drugs such as polyclonal anti-lymphocyte antibodies or monoclonal anti-CD3 antibodies; Maintenance immunosuppressive drugs such as calcineurin inhibitors (eg tacrolimus) and anti-proliferative agents (eg azathioprine, leflunomide or sirolimus); Or in combination therapy including disruption of T cell costimulation, interference of T cell adhesion molecules, and interference of T cell accessory molecules. [184] Except for the prevention of acute rejection, anti-CD20 antibodies can be used to treat acute rejection. Appropriate doses of anti-CD20 are as described above. Anti-CD20 antibodies are optionally used in the treatment of acute rejection with anti-CD3 monoclonal antibodies and / or corticosteroids. [185] Anti-CD20 antibodies may be used for (a) treating or preventing “chronic” allograft rejection, alone or in combination with other immunosuppressive agents and / or costimulatory blockers; (b) may be used as part of resistance induction therapy; Or (c) in the case of xenografts. [186] <Example 4> [187] Blocking Immune Responses to Blood-Friendly Factors [188] Patients genetically deficient in Factor VIII are transfused several times with Factor VIII preparations to generate high titer anti-Factor VIII antibodies. An anti-CD20 antibody, such as RITUXAN®, is administered to the patient in combination with an anti-Factor VIII antibody, eg, at a dosage as described above. Anti-CD20 antibodies may affect the production of antibodies to factor VIII or block the immune response to factor VIII through other mechanisms such as idiotype inhibition. [189] Example 5 [190] Blocking the immune response to platelets [191] Patients who have undergone platelet transfusion develop allogeneic antibodies to platelets. Other therapeutic agents (eg, cyclosporine, staff, Protein A columns, etc.) may be administered to patients who have failed treatment with steroid therapy. An anti-CD20 antibody (eg RITUXAN®) is administered to a patient, eg, at a dosage as described above. Anti-CD20 antibodies may block or mitigate the immune response through other mechanisms, such as affecting the production of antibodies or inhibiting idiotype inhibition or removal of coated platelets by the spleen.
权利要求:
Claims (30) [1" claim-type="Currently amended] A method of blocking an immune response to a foreign antigen in a mammal comprising administering a therapeutically effective amount of an antagonist that binds to CD20 to a mammal not suffering from a malignant tumor. [2" claim-type="Currently amended] The method of claim 1, wherein the antagonist is an antibody. [3" claim-type="Currently amended] The method of claim 1, wherein the foreign antigens comprise a therapeutic agent. [4" claim-type="Currently amended] The method of claim 1, wherein the foreign antigen is selected from the group consisting of antibodies, toxins, gene therapy viral vectors, grafts, infectious agents and homologous antigens. [5" claim-type="Currently amended] The method of claim 1, wherein the mammal is a human. [6" claim-type="Currently amended] The method of claim 2, wherein the antibody is not associated with a cytotoxic agent. [7" claim-type="Currently amended] The method of claim 2, wherein the antibody comprises rituximab (RITUXAN®). [8" claim-type="Currently amended] The method of claim 2, wherein the antibody is associated with a cytotoxic agent. [9" claim-type="Currently amended] The method of claim 8, wherein the cytotoxic agent is a radioactive compound. [10" claim-type="Currently amended] The method of claim 9, wherein the antibody comprises Y2B8 or 131 I-B1 (BEXXAR®). [11" claim-type="Currently amended] The method of claim 1, comprising administering the antagonist intravenously. [12" claim-type="Currently amended] The method of claim 1 comprising administering an antagonist subcutaneously. [13" claim-type="Currently amended] The method of claim 2, comprising administering the antibody to the mammal at a dosage of substantially less than 375 mg / m 2 . [14" claim-type="Currently amended] The method of claim 13, wherein the dosage range is approximately 20 mg / m 2 to 250 mg / m 2 . [15" claim-type="Currently amended] The method of claim 14, wherein the dosage range is approximately 50 mg / m 2 to 200 mg / m 2 . [16" claim-type="Currently amended] The method of claim 2 comprising administering an initial dose (mg / m 2 ) of the antibody followed by administration of an antibody in excess of this dose. [17" claim-type="Currently amended] The method of claim 4, wherein the foreign antigen is an antibody. [18" claim-type="Currently amended] The method of claim 17, wherein the antibody is a murine antibody. [19" claim-type="Currently amended] The method of claim 4, wherein the foreign antigen is a gene therapy virus vector. [20" claim-type="Currently amended] The method of claim 4, wherein the foreign antigen is a graft. [21" claim-type="Currently amended] The method of claim 4, wherein the foreign antigen is a homologous antigen. [22" claim-type="Currently amended] The method of claim 1, comprising administering the antagonist to the mammal prior to exposure to the foreign antigen. [23" claim-type="Currently amended] The method of claim 22, wherein the foreign antigens comprise a graft. [24" claim-type="Currently amended] Administering to the mammal a therapeutic agent other than an antagonist that binds to CD20 and an antagonist that binds to CD20, wherein the therapeutic agent is immunogenic in the mammal and the antagonist blocks an immune response to the therapeutic agent in the mammal. The method of treating a mammal. [25" claim-type="Currently amended] The method of claim 24 comprising administering the therapeutic and antagonist to the mammal essentially simultaneously. [26" claim-type="Currently amended] The method of claim 24, comprising administering the antagonist to the mammal prior to the therapeutic agent. [27" claim-type="Currently amended] The method of claim 24, comprising administering the therapeutic agent to the mammal prior to the antagonist. [28" claim-type="Currently amended] A method of treating a graft-versus-host or host-versus-graft disease in a mammal comprising administering to the mammal a antagonist that binds CD20. [29" claim-type="Currently amended] A product comprising a container, a composition comprising an antagonist that binds to CD20 contained in the container, and a package insert instructing the patient to be exposed to or exposed to a foreign antigen using the composition. [30" claim-type="Currently amended] The article of claim 29, further comprising a second container, and a second composition comprising a therapeutic agent contained in the container.
类似技术:
公开号 | 公开日 | 专利标题 US20190071511A1|2019-03-07|Immunoglobulin variants and uses thereof JP2020023507A|2020-02-13|Therapeutic method for advanced multiple sclerosis US9493579B2|2016-11-15|Multivalent antibodies and uses therefor CN102209556B|2018-02-23|The anti-CD 19 antibodies of improvement KR100365632B1|2003-08-02|Therapeutic application for the treatment of B cell lymphoma of chimeric and radiolabeled antibodies against human B lymphocyte-restricted differentiation antigens DE69729209T2|2005-05-19|Humanized anti-koerper against cd11a US20170233482A1|2017-08-17|Combination therapy US7462352B2|2008-12-09|Methods of treatng B-cell diseases using humanized anti-CD19 antibodies DK1631313T3|2015-06-15|Combination therapy for b cell disorders JP5128935B2|2013-01-23|Humanized anti-TGF-β antibody ES2309012T3|2008-12-16|Compositions of the anti-psca antibody and its procedures against cancerigene cells that express psca. JP5091476B2|2012-12-05|Use of hydrophobic interaction chromatography or hinge region modification to generate homogeneous antibody solutions JP4091235B2|2008-05-28|Therapeutic use of chimeric antibody and radiolabeled antibody against human B lymphocyte restricted differentiation antigen for the treatment of B cell lymphoma CA2478012C|2012-06-19|Internalizing anti-cd74 antibodies and methods of use US6252050B1|2001-06-26|Method for making monoclonal antibodies and cross-reactive antibodies obtainable by the method KR101033196B1|2011-05-09|Anti-CD20 antibodies and fusion proteins thereof and methods of use KR101348472B1|2014-01-07|Human monoclonal antibodies against cd20 AU2005249566B2|2010-11-11|Method for treating multiple sclerosis JP2008507555A|2008-03-13|Treatment for Sjogren's syndrome US6676927B1|2004-01-13|Animal model and methods for its use in the selection of cytotoxic antibodies US20170334989A1|2017-11-23|Antibodies to ntb-a CN1860367B|2010-05-26|Assay for human anti cd20 antibodies and uses therefor CN102958534B|2014-11-05|Notch1 binding agents and methods of use thereof JP2008526205A|2008-07-24|Polypeptides that bind to BR3 and uses thereof JP2008529499A|2008-08-07|Antibody variants and uses thereof
同族专利:
公开号 | 公开日 EP1216056A1|2002-06-26| CN1373672A|2002-10-09| HK1047702A1|2003-03-07| AU2005201189B2|2008-04-03| NZ516491A|2004-11-26| AU778863B2|2004-12-23| WO2001003734A1|2001-01-18| PL352758A1|2003-09-08| HU0202238A3|2004-05-28| PL201086B1|2009-03-31| BR0013201A|2002-04-30| IL147547D0|2002-08-14| MXPA02000419A|2004-09-10| HU0202238A2|2002-10-28| NO20020128D0|2002-01-11| CN101264324A|2008-09-17| AU6082500A|2001-01-30| ZA200200272B|2003-03-26| JP2003528805A|2003-09-30| US20100003252A1|2010-01-07| CA2379274A1|2001-01-18| AU2005201189A1|2005-04-14| NO20020128L|2002-02-28| KR20080075044A|2008-08-13|
引用文献:
公开号 | 申请日 | 公开日 | 申请人 | 专利标题
法律状态:
1999-07-12|Priority to US14440599P 1999-07-12|Priority to US60/144,405 2000-07-10|Application filed by 제넨테크, 인크., 울콧 케네쓰 제이., 아이덱 파마슈티칼즈 인코포레이티드 2000-07-10|Priority to PCT/US2000/018776 2002-04-13|Publication of KR20020027490A
优先权:
[返回顶部]
申请号 | 申请日 | 专利标题 US14440599P| true| 1999-07-12|1999-07-12| US60/144,405|1999-07-12| PCT/US2000/018776|WO2001003734A1|1999-07-12|2000-07-10|Blocking immune response to a foreign antigen using an antagonist which binds to cd20| 相关专利
Sulfonates, polymers, resist compositions and patterning process
Washing machine
Washing machine
Device for fixture finishing and tension adjusting of membrane
Structure for Equipping Band in a Plane Cathode Ray Tube
Process for preparation of 7 alpha-carboxyl 9, 11-epoxy steroids and intermediates useful therein an
国家/地区
|