专利摘要:
HUMANIZED ANTIBODY, HUMANIZED MONOCLONAL ANTIBODY OR ANTIBODY FRAGMENT, ITS USES, ISOLATED POLYNUCLEOTIDE, AND PHARMACEUTICAL COMPOSITION. The present invention relates to new humanized antagonistic anti-CD40 antibodies and to therapeutic and diagnostic methods and compositions for using them.
公开号:BR112012024713B1
申请号:R112012024713-1
申请日:2011-03-30
公开日:2021-02-17
发明作者:Rachel Barret;Scott Brodeur;Keith A. Canada;Tobias LITZENBURGER;Sanjaya Singh
申请人:Boehringer Ingelheim International Gmbh;
IPC主号:
专利说明:

[0001] [0001] This invention relates, in general, to humanized anti-CD40 antibodies for diagnostic and therapeutic use. More specifically, humanized anti-CD40 antibodies and methods of use for the treatment of various diseases or disorders characterized by cells expressing CD40 are disclosed. Pharmaceutical compositions and kits comprising the humanized anti-CD40 antibody are also disclosed. BACKGROUND OF THE INVENTION
[0002] [0002] CD40 is a 48 kDa type I integral membrane glycoprotein and a member of the tumor necrosis factor (TNF) receptor superfamily. CD40 is expressed on a variety of cell types, including normal and neoplastic B cells, interdigitation cells, carcinomas, epithelial cells (for example, keratinocytes), fibroblasts (for example, synoviocytes) and platelets. It is also present on monocytes, macrophages, some endothelial cells, and follicular dendritic cells. CD40 is initially expressed in B cell ontogeny, appearing on B cell precursors, subsequent to the appearance of CD10 and CD19, but before the expression of CD21, CD23, CD24, and the appearance of surface immunoglobulin M (sigM) (Uckun et al., 1990, Blood 15: 2449). CD40 has also been detected on tonsil and plasma cells derived from bone marrow (Pellat-Decounynck et al., 1994, Blood 84: 2597).
[0003] [0003] The CD40 ligand is CD40L (also referred to as CD154, gp39, and TRAP), a member of the TNF superfamily. CD40L is a transmembrane protein expressed predominantly on activated CD4 + T cells and a small subset of CD8 + T cells (Reviewed by Van Kooten C. and Banchereau, 2000).
[0004] [0004] The interaction of CD40 with CD40L induces both humoral and cell-mediated immune responses. CD40 regulates this ligand-receptor pair to activate B cells and other antigen presenting cells (APC), including dendritic cells (DCs) (Reviewed by Toubi and Shoenfeld, 2004); (Kiener, et al., 1995). The role of CD40 on B cells has been studied extensively. The activation of CD40 on B cells induces proliferation, differentiation into antibody-secreting cells and the exchange of isotypes in the germinal centers of secondary lymphoid organs. In vitro studies have shown direct effects of CD40 activation on cytokine production (IL-6, IL-10, TNF-α, LT-α), the expression of adhesion molecules and co-stimulating receptors (ICAM, CD23, CD80 and CD86), and increased expression of MHC class I, MHC class II, and TAP transporter by B lymphocytes (Liu, et al, 1996). For most of these processes, CD40 acts in harmony with any cytokines or other interactions between receptor-ligands.
[0005] [0005] CD40 signaling over monocytes and DCs results in increased survival as well as cytokine secretion (IL-1, IL-6, IL-8, IL-10, IL-12, TNF-α and MIP-1α ). The binding of CD40 on these APCs also results in the overloading of costimulatory molecules, such as ICAM-1, LFA-3, CD80, and CD86. The activation of CD40 receptors is one of the critical signals that allows full CD maturation in efficient APCs guiding the activation of T cells (Banchereau and Steinman, 1998) (Van Kooten C. and Banchereau, 2000).
[0006] [0006] Recent studies in mouse models have shown that CD40 signaling on dendritic cells also plays an important role in the generation of TH17 cells, which are considered to mediate autoimmunity in diseases such as arthritis and multiple sclerosis (Iezzi, et al., 2009) (Perona-Wright, et al., 2009).
[0007] [0007] The availability of knockout mice from CD40 and CD40L, as well as agonistic and antagonistic anti-mouse antibodies offered the possibility of studying the role of interactions between CD40-CD40L in several disease models. Administration of anti-CD40L blockade has been shown to be beneficial in several models of autoimmunity, including spontaneous diseases such as lupus nephritis in SNF1 mice or diabetes in NOD mice or in experimentally induced forms of disease such as collagen-induced arthritis (ASD) ) or experimental autoimmune encephalomyelitis (EAE) (Toubi and Shoenfeld, 2004). CIA in mice was inhibited by an anti-CD40L mAb that blocked the development of inflammation in the joint, the antibody titers in the serum for collagen, the infiltration of inflammatory cells in the subsinovial tissue, in addition to the wear of cartilage and bone (Durie, et al., 1993). For both lupus nephritis and EAE, it was shown that anti-CD40L could also alleviate the disease in progress, confirming the role of CD40-CD40L in the effector phase of the disease (Kalled, et al., 1998); (Howard, et al., 1999).
[0008] [0008] The role for CD40-CD40L interactions in the development of EAE has also been studied in CD40L-deficient mice that carried a specific transgenic T cell receptor for the basic myelin protein. These mice were unable to develop EAE after pre-activation with antigen, and the CD4 + T cells remained inactive and did not produce any INF-γ (Grewal, et al., 1996).
[0009] [0009] In addition, inhibitory antibodies directed against CD40 have shown beneficial effects in models of inflammatory diseases, such as EAE. Lamann et al. Demonstrated that the antagonistic mouse human anti-CD40 mAb mu5D12 and a chimeric version of this mAb effectively prevented the clinical expression of chronic demyelinating EAE in exogamous monkeys (Laman, et. 2002); (Boon, et al., 2001). A follow-up study showed that therapeutic treatment with the chimeric human anti-CD40 antibody reduces MRI-detectable inflammation and delays the increase in pre-existing brain lesions in the EAE model of mico (Hart, et al., 2005).
[0010] [00010] Anti-CD40 antibodies with agonistic activity have been tested in mouse models of arthritis with some conflicting results. As expected for an immunostimulatory agent, FGK45 agonistic mouse anti-CD40 mAb has been shown to exacerbate the disease in the CIA DBA / 1 mouse model (Tellander, et al., 2000). However, in another model of chronic ASD, both FGK45 and another agonistic mouse anti-CD40 mAb, 3/23, exhibited positive therapeutic effects (Mauri, et al., 2000). It has been postulated by this group that agonistic antibodies in this therapeutic treatment regimen have a beneficial effect by inducing immune deviation in relation to a Th2 response, with decreased levels of IFN-γ and increased levels of IL-4 and IL10 (Mauri , et al., 2000).
[0011] [00011] The prevention of transplant rejection by blocking interactions between CD40 / CD154 has also been documented. The use of ch5D12, a chimeric anti-CD40 antagonist, in studies of renal allografts in rhesus monkeys indicates that the CD40 antagonism is sufficient for the modification of the disease and the extension of the average survival times after 100 days. When ch5D12 was combined with an anti-CD86 antibody and given only at the beginning of allograft studies, followed by prolonged treatment with cyclosporine, average survival times greater than 4 years were obtained, indicating that this combination can potentially induce tolerance (Haanstra, et al., 2005).
[0012] [00012] Thus, there are extensive preclinical studies that provide evidence for the crucial role of the CD40-CD40L dyad in driving an efficient T cell-dependent immune response. Blocking CD40 signaling is therefore recognized as an appropriate and necessary therapeutic strategy to suppress a pathogenic autoimmune response in diseases such as RA, multiple sclerosis or psoriasis. However, to date, there is no antibody to CD40 that has been approved for the therapeutic intervention of such disorders, due to the findings that anti-CD40 antibodies previously under development were shown to have significant side effects. Thus, there remains a significant need for therapeutic agents that can be used to interfere with the action of CD40-CD40L and block CD40 signaling. This need could be addressed by new humanized anti-CD40 antibodies that specifically bind CD40 and that show the antigen binding specificity, affinity, and pharmacokinetic and pharmacodynamic properties that allow its use in the therapeutic intervention of CD40-based disorders. BRIEF SUMMARY OF THE INVENTION
[0013] [00013] The present invention provides a humanized monoclonal antibody, where said antibody specifically binds to human CD40 having an antagonistic activity, IC50, of less than 1 nM and has no agonism up to 100 g / ml in cell proliferation B and where said antibody is further characterized by the fact that the antibody has an in vivo half-life in non-human primates that is at least 10 days.
[0014] [00014] The humanized monoclonal antibody can be further characterized by the fact that the antibody has a half-life in cynomolgus monkeys of more than 8 days, at a dose of less than 30 mg / kg.
[0015] [00015] In the illustrative embodiments, the antibody of the invention comprises a heavy chain sequence selected from the group consisting of any of SEQ ID NO: 1 through SEQ ID NO: 4 and a light chain sequence selected from the group that consists of any of SEQ ID NO: 5 through SEQ ID NO: 8.
[0016] [00016] In other embodiments, the antibody is a humanized antibody or antigen-binding fragment of an antibody having the amino acid sequence of the variable region of the heavy chain of any of SEQ ID NO: 1 through 4, SEQ ID NO: 27, SEQ ID NO: 28, SEQ ID NO: 29, SEQ ID NO: 30, SEQ ID NO: 32, SEQ ID NO: 33, SEQ ID NO: 34, SEQ ID NO: 35, SEQ ID NO: 37, SEQ ID NO: 38, SEQ ID NO: 39, SEQ ID NO: 40, SEQ ID NO: 42, SEQ ID NO: 44, SEQ ID NO: 46, SEQ ID NO: 48, SEQ ID NO. 50 SEQ ID NO: 53, SEQ ID NO: 57, SEQ ID NO: 58, SEQ ID NO: 59, SEQ ID NO: 60, SEQ ID NO: 61, SEQ ID NO: 62, SEQ ID NO: 63, SEQ ID NO: 64, SEQ ID NO: 65, SEQ ID NO: 66, SEQ ID NO: 67, SEQ ID NO: 68, SEQ ID NO: 69, SEQ ID NO: 70, SEQ ID NO: 71, SEQ ID NO : 72, or SEQ ID NO: 73.
[0017] [00017] In other embodiments, the antibody is a humanized antibody or antigen-binding fragment of an antibody comprising an amino acid sequence of the light chain variable domain of SEQ ID NO: 5 through SEQ ID NO: 8, SEQ ID NO : 26, SEQ ID NO: 31, SEQ ID NO: 36, SEQ ID NO: 41, SEQ ID NO: 43, SEQ ID NO: 45, SEQ ID NO: 47, SEQ ID NO: 49, SEQ ID NO: 50 , SEQ ID NO: 51, SEQ ID NO: 52, SEQ ID NO: 54, SEQ ID NO: 55, SEQ ID NO: 56, SEQ ID NO: 74, SEQ ID NO: 75, or SEQ ID NO: 76.
[0018] [00018] In the specific embodiments, the monoclonal antibody described in this document is characterized by the fact that it comprises a heavy chain and a light chain, where the CDR1 sequence of the heavy chain selected from the group consisting of SEQ ID NO: 9 to SEQ ID NO: 11, a CDR2 sequence of the heavy chain selected from the group consisting of SEQ ID NO: 12 through SEQ ID NO: 15 and a CDR3 sequence of the heavy chain selected from the group consisting of SEQ ID NO: 16 to SEQ ID NO: 17; and where the light chain CDR1 sequence has a sequence selected from the group consisting of SEQ ID NO: 18 through SEQ ID NO: 21, a CDR2 sequence from the light chain from SEQ ID NO: 22 to SEQ ID NO: 23 and a CDR3 sequence of the light chain selected from the group consisting of SEQ ID NO: 24 through SEQ ID NO: 25.
[0019] [00019] In the specific embodiments, the monoclonal antibody described in this document is characterized by the fact that it comprises a sequence of CDR1 of the heavy chain of SEQ ID NO: 10, a sequence of CDR2 of the heavy chain of SEQ ID NO: 13 and a CDR3 sequence of the heavy chain of SEQ ID NO: 16; and where said antibody comprises a sequence of CDR1 of the light chain of SEQ ID NO: 19, a sequence of CDR2 of the light chain of SEQ ID NO: 22 and a sequence of CDR3 of the light chain of SEQ ID NO: 24.
[0020] [00020] In other specific embodiments, the monoclonal antibody described in this document is characterized by the fact that it comprises a heavy chain CDR1 sequence of SEQ ID NO: 9, a heavy chain CDR2 sequence of SEQ ID NO: 14 and a CDR3 sequence of the heavy chain of SEQ ID NO: 16; and where said antibody comprises a sequence of CDR1 of the light chain of SEQ ID NO: 20, a sequence of CDR2 of the light chain of SEQ ID NO: 22 and a sequence of CDR3 of the light chain of SEQ ID NO: 24.
[0021] [00021] In another specific embodiment, the monoclonal antibody described in this document is characterized by the fact that it comprises a sequence of CDR1 of the heavy chain of SEQ ID NO: 9, a sequence of CDR2 of the heavy chain of SEQ ID NO: 14 and a CDR3 sequence of the heavy chain of SEQ ID NO: 16; and where said antibody comprises a sequence of CDR1 of the light chain of SEQ ID NO: 20, a sequence of CDR2 of the light chain of SEQ ID NO: 22 and a sequence of CDR3 of the light chain of SEQ ID NO: 24.
[0022] [00022] In another specific embodiment, the monoclonal antibody described in this document is characterized by the fact that it comprises a heavy chain CDR1 sequence of SEQ ID NO: 11, a heavy chain CDR2 sequence of SEQ ID NO: 15 and a CDR3 sequence of the heavy chain of SEQ ID NO: 17; and where said antibody comprises a sequence of CDR1 of the light chain of SEQ ID NO: 21, a sequence of CDR2 of the light chain of SEQ ID NO: 23 and a sequence of CDR3 of the light chain of SEQ ID NO: 25.
[0023] [00023] Individual sequences for the preferred antibody heavy chains of the invention are also described in this document. The invention, for example, relates to an anti-CD40 antibody comprising a sequence of the heavy chain variable domain of any one of SEQ ID NOs: 1 to 4. The anti-CD40 antibody is further characterized as comprising a sequence of the domain light chain variable from any one of SEQ ID NO: 5 through SEQ ID NO: 8.
[0024] [00024] Also contemplated is a humanized antibody or antibody fragment having a heavy chain variable domain and a light chain variable region comprising the amino acid sequences of SEQ ID NO: 27 and SEQ ID NO: 26, respectively; SEQ ID NO: 28 and SEQ ID NO: 26, respectively; SEQ ID NO: 29 and SEQ ID NO: 26, respectively; SEQ ID NO: 30 and SEQ ID NO: 26, respectively; SEQ ID NO: 32 and SEQ ID NO: 31, respectively; SEQ ID NO: 33 and SEQ ID NO: 31, respectively; SEQ ID NO: 34 and SEQ ID NO: 31, respectively; SEQ ID NO: 35 and SEQ ID NO: 31, respectively; SEQ ID NO: 37 and SEQ ID NO: 36, respectively; SEQ ID NO: 38 and SEQ ID NO: 36, respectively; SEQ ID NO: 39 and SEQ ID NO: 36, respectively; SEQ ID NO: 40 and SEQ ID NO: 36, respectively.
[0025] [00025] In another embodiment, the invention relates to a humanized antibody or antibody fragment having a heavy chain variable domain and a light chain variable region comprising the amino acid sequences of SEQ ID NO: 27 and SEQ ID NO: 26 , respectively.
[0026] [00026] In another embodiment, the invention relates to a humanized antibody or antibody fragment having a heavy chain variable domain and a light chain variable region comprising the amino acid sequences of SEQ ID NO: 28 and SEQ ID NO: 26 , respectively.
[0027] [00027] In another embodiment, the invention relates to a humanized antibody or antibody fragment having a heavy chain variable domain and a light chain variable region comprising the amino acid sequences of SEQ ID NO: 29 and SEQ ID NO: 26 , respectively.
[0028] [00028] In another embodiment, the invention relates to a humanized antibody or antibody fragment having a heavy chain variable domain and a light chain variable region comprising the amino acid sequences of SEQ ID NO: 30 and SEQ ID NO: 26 , respectively.
[0029] [00029] In another embodiment, the invention relates to a humanized antibody or antibody fragment having a heavy chain variable domain and a light chain variable region comprising the amino acid sequences of SEQ ID NO: 32 and SEQ ID NO: 31 , respectively.
[0030] [00030] In another embodiment, the invention relates to a humanized antibody or antibody fragment having a heavy chain variable domain and a light chain variable region comprising the amino acid sequences of SEQ ID NO: 33 and SEQ ID NO: 31 , respectively.
[0031] [00031] In another embodiment, the invention relates to a humanized antibody or antibody fragment having a heavy chain variable domain and a light chain variable region comprising the amino acid sequences of SEQ ID NO: 34 and SEQ ID NO: 31 , respectively.
[0032] [00032] In another embodiment, the invention relates to a humanized antibody or antibody fragment having a heavy chain variable domain and a light chain variable region comprising the amino acid sequences of SEQ ID NO: 35 and SEQ ID NO: 31 , respectively.
[0033] [00033] In another embodiment, the invention relates to a humanized antibody or antibody fragment having a heavy chain variable domain and a light chain variable region comprising the amino acid sequences of SEQ ID NO: 37 and SEQ ID NO: 36 , respectively.
[0034] [00034] In another embodiment, the invention relates to a humanized antibody or antibody fragment having a heavy chain variable domain and a light chain variable region comprising the amino acid sequences of SEQ ID NO: 38 and SEQ ID NO: 36 , respectively.
[0035] [00035] In another embodiment, the invention relates to a humanized antibody or antibody fragment having a heavy chain variable domain and a light chain variable region comprising the amino acid sequences of SEQ ID NO: 39 and SEQ ID NO: 36 , respectively.
[0036] [00036] In another embodiment, the invention relates to a humanized antibody or antibody fragment having a heavy chain variable domain and a light chain variable region comprising the amino acid sequences of SEQ ID NO: 40 and SEQ ID NO: 36 , respectively.
[0037] [00037] Another embodiment relates to an isolated antibody or antigen-binding fragment that specifically binds to human CD40, comprising a humanized heavy chain variable domain comprising a framework region having an amino acid sequence at least 90% identical to amino acid sequence of the structure region of the heavy chain amino acid sequence of the human variable domain of SEQ ID NO: 27, SEQ ID NO: 28, SEQ ID NO: 29 or SEQ ID NO: 30, and comprising an amino acid sequence of light chain at least 90% identical to a variable domain of the corresponding light chain of SEQ ID NO: 26.
[0038] [00038] Another embodiment relates to an isolated antibody or antigen-binding fragment that specifically binds to human CD40, comprising a humanized heavy chain variable domain comprising a framework region having an amino acid sequence at least 90% identical to amino acid sequence of the structure region of the heavy chain amino acid sequence of the human variable domain of SEQ ID NO: 32, SEQ ID NO: 33, SEQ ID NO: 34 or SEQ ID NO: 35, and comprising an amino acid sequence of light chain at least 90% identical to a corresponding light chain variable of SEQ ID NO: 31.
[0039] [00039] In another aspect, the invention relates to the isolated antibody or antigen-binding fragment described in the modality immediately above, where the amino acid sequence of the heavy chain is SEQ ID NO: 32; in another embodiment, the amino acid sequence of the heavy chain is SEQ ID NO: 33; in another embodiment, the amino acid sequence of the heavy chain is SEQ ID NO: 34; and in another embodiment, the amino acid sequence of the heavy chain is SEQ ID NO: 35.
[0040] [00040] An isolated antibody or antigen-binding fragment that specifically binds to human CD40 is also contemplated, comprising a humanized heavy chain variable domain comprising a framework region having an amino acid sequence at least 90% identical to the amino acid sequence the heavy chain amino acid sequence structure region of the human variable domain of SEQ ID NO: 37, SEQ ID NO: 38, SEQ ID NO: 39 or SEQ ID NO: 40, and comprising a light chain amino acid sequence by minus 90% identical to a corresponding light chain of SEQ ID NO: 36.
[0041] [00041] In another aspect, the invention relates to the isolated antibody or antigen-binding fragment described in the modality immediately above, where the amino acid sequence of the heavy chain is SEQ ID NO: 37; in another embodiment, the amino acid sequence of the heavy chain is SEQ ID NO: 38; in another embodiment, the amino acid sequence of the heavy chain is SEQ ID NO: 39; and in another embodiment, the amino acid sequence of the heavy chain is SEQ ID NO: 40.
[0042] [00042] The antibodies of the present invention can be further characterized by the fact that said antibodies fail to stimulate the production of cytokines from B cells in this absence of CD40L.
[0043] [00043] The antibodies of the present invention can be further characterized by the fact that said antibodies bind to human CD40 in the presence of 50% human serum with a reduction in the rate of less than twice.
[0044] [00044] The antibodies of the present invention can be further characterized by the fact that said antibody produces inhibition of the production of IgM and IgG in a mammal at a concentration of 1 mg / kg.
[0045] [00045] The antibodies of the present invention can be used in a variety of therapeutic, prophylactic, diagnostic and other methods. For example, the present invention describes a method of blocking the function of human CD40 in a mammal, comprising administering to said mammal a composition comprising an antibody of the invention in an amount sufficient to block a CD40-mediated immune response in said mammal.
[0046] [00046] Also included in this document is a method of treating or ameliorating graft vs. disease. host in a mammal, comprising administering to said mammal a composition comprising an antibody of the invention, in an amount sufficient to lessen one or more of the symptoms of graft vs. disease. host in said animal.
[0047] [00047] As an example, autoimmune or inflammatory disease may include, but is not limited to rheumatoid arthritis, multiple sclerosis, proliferative lupus glomerulonephritis, inflammatory bowel disease (IBD), psoriasis, idiopathic thrombocytopenic purpura (ITP), Disease Crohn's disease and systemic lupus erythematosus (SLE), Hashimoto's thyroiditis, primary myxoedema, thyrotoxicosis / Graves' disease, pernicious anemia, autoimmune atrophic gastritis, autoimmune carditis, Addison's disease, premature menopause, type 1 diabetes mellitus, Good pasture syndrome, myasthenia gravis, autoimmune hemolytic anemia, idiopathic leukopenia, primary biliary cirrhosis, chronic active hepatitis (HBs Ag negative), cryptogenic cirrhosis, Sjogren's syndrome, dermatomyositis, scleroderma, mixed connective tissue disease, discoid lupus erythematosus, and systemic vasculitis. In the illustrative modalities, the mammal has rheumatoid arthritis.
[0048] [00048] The methods of the invention may additionally comprise administering a second therapeutic agent selected from the group consisting of a TNF antagonist, a disease modifying antirheumatic drug, a CTLA4 antagonist, an IL-6 antireceptor mAb and an mAb anti-CD20.
[0049] [00049] In specific modalities, inflammatory disease or autoimmune disease is an inflammatory disease or autoimmune disease that is associated with cells that express both CD40 and CD20.
[0050] [00050] In specific methods, treatment involves administering the antibody composition via a parenteral route of administration.
[0051] [00051] In specific methods, treatment involves administering the antibody composition intravenously or subcutaneously.
[0052] [00052] Additional methods of the invention comprise inhibiting the production of antibodies by B cells in a human patient, comprising administering to said human patient an effective amount of an anti-CD40 antibody of the invention.
[0053] [00053] More specifically, the human patient has an inflammatory disease or autoimmune disease that is associated with cells that express CD40.
[0054] [00054] In the illustrative modalities, the human patient is suffering from an autoimmune disease selected from the group consisting of autoimmune or inflammatory disease, selected from the group consisting of rheumatoid arthritis, multiple sclerosis, lupus proliferative glomerulonephritis, inflammatory disease bowel disease (IBD), psoriasis, idiopathic thrombocytopenic purpura (ITP), Crohn's disease and systemic lupus erythematosus (SLE), Hashimoto's thyroiditis, primary myxoedema, thyrotoxicosis / Graves' disease, pernicious anemia, autoimmune atrophic gastritis, carditis Addison's disease, premature menopause, type 1 diabetes mellitus, Good pasture syndrome, myasthenia gravis, autoimmune hemolytic anemia, idiopathic leukopenia, primary biliary cirrhosis, chronic active hepatitis (HBs Ag negative), cryptogenic cirrhosis, Sjogren's syndrome, dermatomyositis, scleroderma, mixed connective tissue disease, discoid lupus erythematosus, and systemic vasculitis.
[0055] [00055] Another method of the invention relates to inhibiting the growth of cells expressing the human CD40 antigen, comprising administering the antibody or antigen binding fragment of the invention to the cells, antibody or antigen binding fragment which specifically binds to the CD40 antigen on the human cell surface, where the binding of the antibody or antigen-binding fragment to the CD40 antigen inhibits cell growth or differentiation.
[0056] [00056] Also contemplated is a method of treating a patient having a CD40-associated disorder, comprising administering to the patient the antibody or antigen-binding fragment of the invention, antibody or antigen-binding fragment which specifically binds to human CD40 , where the binding of the antibody or antigen-binding fragment to CD40 inhibits the growth or differentiation of cells from the disorder associated with CD40. The cells may, but are not limited to, B lymphoblastoid cells, pancreatic cells, lung cells, breast cells, ovarian cells, colon cells, prostate cells, skin cells, head and neck cells, bladder cells, cells of bone or kidney cells.
[0057] [00057] The treatment method to inhibit cell growth or differentiation may be useful in the treatment for chronic lymphocytic leukemia, Burkitt's lymphoma, multiple myeloma, a T-cell lymphoma, Non-Hodgkin's lymphoma, Hodgkin's disease, macroglobulinemia Waldenstrom's disease or Kaposi's sarcoma.
[0058] [00058] A method of inducing depletion of peripheral B cells is also contemplated, comprising administering to the cells the antibody or antigen-binding fragment of the invention, antibody or antigen-binding fragment which specifically binds to a CD40 antigen of the human cell surface, where the binding of the antibody or antigen-binding fragment to the CD40 antigen induces cell depletion.
[0059] [00059] In the specific embodiments, the antibody or antigen-binding fragment is administered to a patient having an immune disorder. For example, the immune disorder is rheumatoid arthritis or systemic lupus erythematosus.
[0060] [00060] Also contemplated is a method of treating rheumatoid arthritis in a patient, comprising administering to said patient an antibody of the invention, wherein said antibody is an antagonistic antibody that blocks the function of CD40 in said patient.
[0061] [00061] Preferably, the antibody is administered in an effective amount to inhibit B cell differentiation and antibody isotype exchange in said patient.
[0062] [00062] In other embodiments, the antibody is administered in an effective amount to inhibit the production of cytokine and chemokine and the uptake of adhesion molecules in T cells and macrophages in said patient. Preferably, the antibody is administered in an effective amount to inhibit activation of dendritic cells in said patient.
[0063] [00063] In other embodiments, the method is additionally characterized by the fact that the antibody is administered in an effective amount to inhibit the production of proinflammatory cytokines, chemokines, matrix metalloproteinases, prostaglandins, and to unregulate the adhesion molecules in non-immune cells in said patient.
[0064] [00064] In the specific embodiments, the antibody is administered in combination with a regimen comprising the administration of methotrexate and / or the administration of Enbrel / Humira.
[0065] [00065] The patient to receive therapy is one who has rheumatoid arthritis and has been unresponsive to treatment with methotrexate alone.
[0066] [00066] In the specific modalities, the method comprises treating said patient with a regimen comprising the administration of methotrexate and / or the administration of Enbrel / Humira.
[0067] [00067] The method of the invention can be further characterized, where the treatment of said patient with said antagonistic anti-CD40 antibody has a superior efficacy to treatment with methotrexate alone, Enbrel alone, a combination of Enbrel + methotrexate.
[0068] [00068] The method of the invention can be further characterized, where the treatment of said patient with said antagonistic anti-CD40 antibody is more effective than treatment with Enbrel + MTX in patients who have had an inadequate response to methotrexate.
[0069] [00069] In specific embodiments, the antibody is administered in combination with a regimen comprising an anti-TNF agent.
[0070] [00070] In the specific modalities, the patient is characterized as one who has rheumatoid arthritis and has been unresponsive to treatment with an anti-TNF agent alone. In such embodiments, the method may comprise treating said patient with a regimen comprising treatment with an anti-TNF agent in combination with said antagonistic anti-CD40 antibody.
[0071] [00071] In the specific embodiments, the treatment of said patient with said antagonistic anti-CD40 antibody is more effective than treatment with an anti-TNF agent.
[0072] [00072] In still other modalities, the method is characterized by the fact that the treatment of said patient with said antagonistic anti-CD40 antibody is more effective than treatment with Orencia or Rituxan, in patients who have had an inadequate response to a anti-TNF agent alone.
[0073] [00073] The present invention further contemplates a pharmaceutical composition comprising: (i) the antibody or antigen-binding fragment as described herein; and (ii) a pharmaceutically acceptable excipient. In such compositions, the antibody or its antigen binding fragment can advantageously be conjugated to a second agent, such as, for example, a cytotoxic agent, a PEG vehicle, an enzyme or a marker.
[0074] [00074] An isolated polynucleotide encoding an amino acid sequence of the variable region of the heavy chain of any of SEQ ID NO: 1 to 4, SEQ ID NO: 27, SEQ ID NO: 28, SEQ ID NO: 29 is also contemplated in this document. , SEQ ID NO: 30, SEQ ID NO: 32, SEQ ID NO: 33, SEQ ID NO: 34, SEQ ID NO: 35, SEQ ID NO: 37, SEQ ID NO: 38, SEQ ID NO: 39, SEQ ID NO: 40, SEQ ID NO: 42, SEQ ID NO: 44, SEQ ID NO: 46, SEQ ID NO: 48, SEQ ID. AT THE. 50, SEQ ID NO: 53, SEQ ID NO: 57, SEQ ID NO: 58, SEQ ID NO: 59, SEQ ID NO: 60, SEQ ID NO: 61, SEQ ID NO: 62, SEQ ID NO: 63, SEQ ID NO: 64, SEQ ID NO: 65, SEQ ID NO: 66, SEQ ID NO: 67, SEQ ID NO: 68, SEQ ID NO: 69, SEQ ID NO: 70, SEQ ID NO: 71, SEQ ID NO: 72, or SEQ ID NO: 73.
[0075] [00075] Also contemplated in this document is an isolated polynucleotide encoding an amino acid sequence of the variable region of the light chain of any of SEQ ID NO: 5 to SEQ ID NO: 8, SEQ ID NO: 26, SEQ ID NO: 31, SEQ ID NO: 36, SEQ ID NO: 41, SEQ ID NO: 43, SEQ ID NO: 45, SEQ ID NO: 47, SEQ ID NO: 49, SEQ ID NO: 50, SEQ ID NO: 51, SEQ ID NO : 52, SEQ ID NO: 54, SEQ ID NO: 55, SEQ ID NO: 56, SEQ ID NO: 74, SEQ ID NO: 75, or SEQ ID NO: 76.
[0076] [00076] The invention further relates to the use of the antibodies described in this document for the manufacture of a medicament for blocking human CD40 function in a mammal, where the medicament blocks a CD40-mediated immune response in said mammal.
[0077] [00077] In one embodiment, the invention relates to the manufacture of a medicament to treat or ameliorate graft vs. disease. host in a mammal.
[0078] [00078] In illustrative modalities, the drug is manufactured for the treatment of an autoimmune or inflammatory disease selected from the group consisting of rheumatoid arthritis, multiple sclerosis, lupus proliferative glomerulonephritis, inflammatory bowel disease (IBD), psoriasis, purple idiopathic thrombocytopenic (ITP), Crohn's disease and systemic lupus erythematosus (SLE), Hashimoto's thyroiditis, primary myxoedema, thyrotoxicosis / Graves' disease, pernicious anemia, atrophic autoimmune gastritis, autoimmune carditis, Addison's disease, premature menopause, diabetes mellitus, premature melanoma 1, Good pasture syndrome, myasthenia gravis, autoimmune hemolytic anemia, idiopathic leukopenia, primary biliary cirrhosis, chronic active hepatitis (HBs Ag negative), cryptogenic cirrhosis, Sjogren's syndrome, dermatomyositis, scleroderma, mixed connective tissue disease, discoid lupus erythematosus , and systemic vasculitis.
[0079] [00079] In some embodiments, the drug may additionally comprise a second therapeutic agent selected from the group consisting of a TNF antagonist, a disease-modifying antirheumatic drug, a CTLA4 antagonist, an IL-6 antireceptor mAb and an anti-CD20 mAb.
[0080] [00080] The drug can be manufactured for use in a parenteral route of administration. The drug can be manufactured for use intravenously or subcutaneously.
[0081] [00081] Another modality contemplates a use of the antibodies described in this document for the manufacture of a medicament for inhibiting the production of antibodies by B cells in a human patient.
[0082] [00082] Another modality contemplates a use of the antibodies described in this document for the manufacture of a medicine for inhibiting the growth and / or differentiation of cells that express the human CD40 antigen.
[0083] [00083] Another modality contemplates a use of the antibodies described in this document for the manufacture of a medicine for the treatment of a patient having a disorder associated with CD40, where the binding of the antibody or the antigen-binding fragment in said medicine to CD40 inhibits the growth or differentiation of cells from the disorder associated with CD40.
[0084] [00084] The drug can be manufactured for use in the treatment of cells of a CD40-associated disorder selected from B lymphoblastoid cells, pancreatic cells, lung cells, breast cells, ovarian cells, colon cells, prostate cells, cells skin, head and neck cells, bladder cells, bone cells or kidney cells.
[0085] [00085] The drug can be manufactured for use in the treatment of chronic lymphocytic leukemia, Burkitt's lymphoma, multiple myeloma, T-cell lymphoma, Non-Hodgkin's lymphoma, Hodgkin's disease, Waldenstrom's macroglobulinemia or Kaposi's sarcoma.
[0086] [00086] Another embodiment contemplates a use of the antibodies of the invention in the manufacture of a drug to induce depletion of peripheral B cells, where the antibody or antigen-binding fragment of the drug specifically binds to a CD40 antigen on the human cell surface, where binding of the antibody or antigen-binding fragment to the CD40 antigen induces cell depletion.
[0087] [00087] The drug can be manufactured for use in treating a patient having an immune disorder.
[0088] [00088] The drug can be manufactured for use in the treatment of rheumatoid arthritis or systemic lupus erythematosus.
[0089] [00089] Another embodiment contemplates a use of the antibodies of the invention in the manufacture of a medicament for the treatment of rheumatoid arthritis in a patient.
[0090] [00090] The drug can be manufactured for use in inhibiting B cell differentiation and antibody isotype exchange in said patient.
[0091] [00091] The drug can be manufactured for use in inhibiting the production of cytokine and chemokine and the overloading of adhesion molecules in T cells and macrophages in said patient.
[0092] [00092] The drug can be manufactured for use in inhibiting the activation of dendritic cells in said patient.
[0093] [00093] The drug can be manufactured for use in the inhibition of the production of proinflammatory cytokines, chemokines, matrix metalloproteinases, prostaglandins, and lack of regulation of adhesion molecules in non-immune cells in said patient.
[0094] [00094] In certain embodiments, the medicine is manufactured as a combination medicine to be administered in combination with a regimen comprising the administration of methotrexate and / or the administration of Enbrel / Humira.
[0095] [00095] In other embodiments, the medicament is manufactured as a combination medicament and the medicament, in addition to comprising the antibodies of the invention, additionally comprises an anti-TNF agent. BRIEF DESCRIPTION OF THE SEVERAL VIEWS OF THE DRAWINGS
[0096] [00096] Figure 1: A. Binding curves and corresponding EC50 values of humanized antibodies on HEK-293 cells transfected with CD40 measured by flow cytometry. B. Comparison of the binding of Antibody A, Antibody B and Antibody C to HEK cells transfected with CD40, measured by flow cytometry. The representative data for an experiment is shown.
[0097] [00097] Figure 2: Binding curves and corresponding EC50 values of humanized antibodies on RAMOS cells, measured by flow cytometry. The representative data for an experiment is shown.
[0098] [00098] Figure 3: Testing of mouse and humanized antibodies for antagonistic activity in a human primary B cell proliferation assay. (A) Representative antibody titration curves and the resulting IC 50 values are plotted for each mouse precursor antibody. Representative data for a donor is shown. (B) Overlapping inhibition curves representing the antagonism of several humanized anti-CD40 antibodies compared to 4D11.
[0099] [00099] Figure 4: Summary of humanized antibody test results for antagonistic (IC50) and agonistic (SI = stimulus index) activity in a human primary B cell proliferation assay. Several anti-CD40, 4D11, G28.5 and 5D12 antibodies are shown for comparison.
[0100] [000100] Figure 5: Antibody B, Antibody A and Antibody C testing for CD40-induced inhibition of CD86 overload in human whole blood assays. The antiCD40 4D11 antibody is shown for comparison. The IgG1 isotype control showed no effect in this assay. Representative data for a donor is shown.
[0101] [000101] Figure 6: Summary of Antibody B test results for CD40-induced inhibition of CD86 overload on purified human B cells and human whole blood. The data at both points (A) IC50 and (B) IC90 are represented. The IgG1 isotype control showed no effect in any assay. The data for multiple donors (n = 4-5) are summarized in the table.
[0102] [000102] Figure 7: Test of Antibody B, Antibody A and Antibody C for the inhibition of CD40 overloading induced by CD40 in whole blood tests of cynomolgus monkeys. The IgG1 isotype control showed no effect in this assay. Representative data for a donor is shown.
[0103] [000103] Figure 8: Plasma concentration curves for Antibody A (left panel) and Antibody B (right panel) in a cynomolgus monkey after administration of 1 and 10 mg / kg of each antibody. The data is the summary of administration in 3 animals for each antibody.
[0104] [000104] Figure 9: Change in the percentage of CD86 positive B cells from cynomolgus monkeys before (Antibody B) and (Antibody A) administration and at 3 time points after treatment with each antibody. Antibody B (Top panels) and Antibody A were administered to 3 animals, each with 1 mg / kg (left panels) or 10 mg / kg (right panels).
[0105] [000105] Figure 10: Levels of (A) human IgG and (B) human IgM in NSG mice at 2 weeks after the injection of 1.25 x 106 human PBMC. The mice were treated with vehicle, an isotype control and the antibodies Antibody A, Antibody B and Antibody C, at a dose of 1 mg / kg one day before the transfer of human PBMC.
[0106] [000106] Figure 11: Binding of several mouse anti-human CD40 antibodies to human platelets.
[0107] [000107] Figure 12: Summary of the results of the comparison of the binding of Antibody B with anti-CD40 mAb 4D11 on human B cells and platelets in whole blood.
[0108] [000108] Figure 13: ADCC activity with wild type and knockout IgG1 constructs. DETAILED DESCRIPTION OF THE INVENTION
[0109] [000109] CD40-mediated signaling is now recognized as being involved in a variety of target disorders. Despite the availability of a variety of preclinical data showing that intervention in these disorders would be therapeutically beneficial, there remains a need for antagonistic anti-CD40 antibodies that can be used in the treatment of autoimmune diseases. The present invention, in preferred embodiments, relates to humanized antibodies that recognize CD40. In the specific embodiments, the sequence of these humanized antibodies has been identified based on the sequences of certain antibodies from target mice.
[0110] [000110] The terms "CD40" and "CD40 surface antigen" refer to a glycoprotein of approximately 48 kD expressed on the surface of normal and neoplastic B cells, which acts as a receptor for the signals involved in proliferation and differentiation cell (Ledbetter et al., 1987, J. Immunol. 138: 788-785). A cDNA molecule encoding CD40 was isolated from a library prepared from the Burkitt Raji lymphoma cell line (Stamenkovic et al., 1989, EMBO J. 8: 1403).
[0111] [000111] As used herein, a cell that endogenously expresses CD40 is any cell characterized by CD40 surface expression, including, but not limited to, normal and neoplastic B cells, interdigitation cells, basal epithelial cells, carcinoma cells, macrophages, endothelial cells, follicular dendritic cells, tonsil cells, and plasma cells derived from bone marrow. In some embodiments, the CD40 molecule is a human CD40 molecule.
[0112] [000112] The antibodies of the invention specifically bind to recombinant and native human CD40. A humanized monoclonal antibody, where said antibody specifically binds to human CD40 having an antagonistic activity, IC50, of less than 1 nM and has no agonism up to 100 g / ml in the proliferation of B cells and where said antibody is additionally characterized by the fact that the antibody has an in vivo half-life in non-human primates that is at least 10 days.
[0113] [000113] Preferably, the antibody specifically binds CD40 in the CD40-Fc conjugate with an EC50 of less than 1 nM and CD40 in cells expressing CD40 with an EC50 of less than 2.5 nM. The antagonistic properties of the antibody are defined by the fact that it has an antagonistic activity on B cells or IC50 dendritic cell of less than 1 nM. The antibody additionally has superior pharmacokinetic properties, having an increased in vivo half-life compared to other anti-CD40 antibodies (for example, the anti-CD40 antibody 4D11).
[0114] [000114] As used in this document, a cell that expresses CD40 is any cell characterized by surface expression of CD40, including, but not limited to, normal and neoplastic B cells, interdigitation cells, basal epithelial cells, carcinoma cells, macrophages , endothelial cells, follicular dendritic cells, tonsil cells, and bone marrow-derived plasma cells. In some embodiments, the CD40 molecule is a human CD40 molecule.
[0115] [000115] The antibodies of the present invention recognize the specific "CD40 antigen epitope" and the "CD40 epitope". As used herein, these terms refer to a molecule (for example, a peptide) or a fragment of a molecule capable of immunoreactivity with an anti-CD40 antibody and, for example, include a CD40 antigenic determinant recognized by any of the antibodies having a heavy chain / light chain sequence of light chain SEQ ID NO: 26 with any of the heavy chain SEQ ID NOs: 27, 28, 29 or 30; or SEQ ID NO: 31 light chain with any of SEQ ID NOs 32, 33, 34 or 35 heavy chain; or light chain SEQ ID NO 36 with any of the heavy chain SEQ ID NOs 37, 38, 39 or 40. CD40 antigen epitopes can be included in proteins, protein fragments, peptides or the like. Epitopes are most commonly proteins, short oligopeptides, imitators of oligopeptides (ie, organic compounds that mimic the antibody-binding properties of the CD40 antigen), or combinations thereof.
[0116] [000116] The generalized structure of antibodies or immunoglobulin is well known to those of skill in the art, these molecules are heterotetrameric glycoproteins, typically about 150,000 daltons, composed of two identical light (L) chains and two identical heavy (H) chains . Each light chain is covalently linked to a heavy chain by a disulfide bond to form a heterodimer, and the heterotrameric molecule is formed through a covalent disulfide bond between the two identical heavy chains of the heterodimers. Although the light and heavy chains are linked together by a disulfide bond, the number of disulfide bonds between the two heavy chains varies by the immunoglobulin isotype. Each heavy and light chain also has regularly spaced disulfide bridges within the chain. Each heavy chain has a variable domain (VH) at the amino end, followed by three or four constant domains (CH1, CH2, CH3, and CH4), as well as an articulation region between CH1 and CH2. Each light chain has two domains, an amino terminal variable domain (VL) and a carboxy terminal constant domain (CL). The VL domain associates non-covalently with the VH domain, while the CL domain is commonly covalently linked to the CH1 domain through a disulfide bond. Particular amino acid residues are believed to form an interface between the variable domains of the light and heavy chains (Chothia et al., 1985, J. Mol. Biol. 186: 651-663).
[0117] [000117] Certain domains within the variable domains differ extensively between the different antibodies, that is, they are "hypervariable". These hypervariable domains contain residues that are directly involved in the binding and specificity of each particular antibody due to its specific antigenic determinant. Hypervariability, both in the light chain and in the variable domains of the heavy chain, is concentrated in three segments known as complementarity determining regions (CDRs) or hypervariable loops (HVLs). CDRs are defined by comparing the sequences in Kabat et al., 1991, In: Sequences of Proteins of Immunological Interest, 5th Ed. Public Health Service, National Institutes of Health, Bethesda, Md., While HVLs are structurally defined according with the three-dimensional structure of the variable domain, as described by Chothia and Lesk, 1987, J. Mol. Biol. 196: 901-917. Where these two methods result in slightly different identifications of a CDR, the structural definition is preferred. As defined by Kabat, CDR-L1 is positioned around residues 24-34, CDR-L2, around residues 50-56, and CDR-L3, around residues 89-97 in the variable domain of the chain Light; CDR-H1 is positioned around residues 31-35, CDR-H2 around residues 50-65, and CDR-H3 around residues 95-102 in the variable domain of the heavy chain. CDR1, CDR2, CDR3 of heavy and light chains, therefore, define the unique and functional properties, specific to a given antibody.
[0118] [000118] The three CDRs within each of the heavy and light chains are separated by structure regions (FR), which contain sequences that tend to be less variable. From the amino end to the carboxy end of the variable domains of the heavy and light chains, the FRs and CDRs are arranged in the order: FR1, CDR1, FR2, CDR2, FR3, CDR3, and FR4. The predominantly  sheet configuration of the FRs places the CDRs within each of the strands in close proximity to each other, as well as with the CDRs of the other strand. The resulting conformation contributes to the antigen-binding site (see Kabat et al, 1991, NIH Publ. No. 91-3242, Vol. I, pages 647-669), although not all CDR residues are necessarily involved directly in the binding to the antigen.
[0119] [000119] RF residues and Ig constant domains are not directly involved in binding to the antigen, but they do contribute to binding to the antigen and / or mediate the effector function of the antibody. Some RF residues are believed to have a significant effect on antigen binding in at least three ways: non-covalently binding to an epitope, interacting with one or more CDR residues, and affecting the interface between the chains heavy and light. The constant domains are not directly involved in antigen binding, but they mediate several Ig effector functions, such as the participation of the antibody in antibody-dependent cell cytotoxicity (ADCC), complement-dependent cytotoxicity (CDC) and cell-dependent phagocytosis antibody (ADCP).
[0120] [000120] Light chains of vertebrate immunoglobulins are specified for one of two clearly distinct classes, kappa (k) and lambda (λ), based on the amino acid sequence of the constant domain. By comparison, mammalian immunoglobulin heavy chains are specified for one of five main classes, according to the sequence of the constant domains: IgA, IgD, IgE, IgG, and IgM. IgG and IgA are further divided into subclasses (isotypes), for example, IgG1, IgG2, IgG3, IgG4, IgA1, and IgA2. The heavy chain constant domains that correspond to the different classes of immunoglobulins are called α, δ, ε, γ, and µ, respectively. Subunit structures and three-dimensional configurations of native immunoglobulin classes are well known.
[0121] [000121] The terms "antibody", "anti-CD40 antibody", "humanized anti-CD40 antibody" and "variant humanized anti-CD40 antibody" are used in this document in the broadest sense and specifically include monoclonal antibodies (including antibodies monoclonal antibodies), polyclonal antibodies, multispecific antibodies (for example, bispecific antibodies), and antibody fragments, such as variable domains and other parts of antibodies that exhibit a desired biological activity, for example, binding to CD40.
[0122] [000122] The term "monoclonal antibody" (mAb) refers to an antibody from a population of substantially homogeneous antibodies; that is, the individual antibodies in that population are identical, except for naturally occurring mutations that may be present in modest amounts. Monoclonal antibodies are highly specific, being directed against a single antigenic determinant, an "epitope". Therefore, the "monoclonal" modifier is indicative of a substantially homogeneous antibody population directed towards the identical epitope and is not to be interpreted as requiring production of the antibody by any particular method. It should be understood that monoclonal antibodies can be prepared by any practice or methodology known in the art, including, for example, the hybridoma method (Kohler et al., 1975, Nature 256: 495), or the recombinant DNA methods known in the art. technique (see, for example, US Pat. No. 4,816,567), or methods of isolating recombinantly produced monoclonal using phage antibody libraries, using the techniques described in Clackson et al., 1991, Nature 352: 624-628, and Marks et al., 1991, J. Mol. Biol. 222: 581-597.
[0123] [000123] Chimeric antibodies consist of the variable regions of the heavy and light chains of an antibody of one species (for example, a non-human mammal, such as a mouse) and the constant regions of the heavy and light chains of an antibody of another species (for example, human) and can be obtained by linking the DNA sequences encoding the variable regions of the antibody of the first species (for example, mouse) to the DNA sequences for the constant regions of the antibody of the second species (for example, human) and transforming a host with an expression vector containing the linked sequences to allow it to produce a chimeric antibody. Alternatively, the chimeric antibody could also be one in which one or more regions or domains of the heavy and / or light chain are identical with, homologous to, or a variant of, the corresponding sequence in a monoclonal antibody from another class or isotype immunoglobulin, or from a consensus or germline sequence. Chimeric antibodies can include fragments of such antibodies, as long as the antibody fragment exhibits the desired biological activity of its parent antibody, for example, binding to the same epitope (see, for example, US Pat. No. 4,816. 567; and Morrison et al., 1984, Proc. Natl. Acad. Sci. USA 81: 6851-6855).
[0124] [000124] The terms "antibody fragment", "anti-CD40 antibody fragment", "humanized anti-CD40 antibody fragment", "variant humanized anti-CD40 antibody fragment" refer to a part of an anti antibody - natural size CD40, in which a variable region or functional capacity is preserved, for example, the specific binding to the CD40 epitope. Examples of antibody fragments include, but are not limited to, a Fab, Fab ', F (ab') 2, Fd, Fv, scFv and scFv-Fc fragment, a diabody, a linear antibody, a single chain antibody , a minibody, a diabody formed from antibody fragments, and multispecific antibodies formed from antibody fragments.
[0125] [000125] Life-size antibodies can be treated with enzymes, such as papain or pepsin, to generate useful antibody fragments. Papain digestion is used to produce two identical antibody fragments that bind to the antigen, called "Fab" fragments, each with a single antigen binding site, and a residual "Fc" fragment. The Fab fragment also contains the constant domain of the light chain and the CH1 domain of the heavy chain. Treatment with pepsin produces an F (ab ') 2 fragment that has two antigen-binding sites and is still capable of cross-linking the antigen.
[0126] [000126] Fab 'fragments differ from Fab fragments in the presence of additional residues, including one or more cysteines from the antibody hinge region at the C-terminus of the CH1 domain. F (ab ') 2 antibody fragments are pairs of Fab' fragments linked by cysteine residues in the hinge region. Other chemical couplings of antibody fragments are also known.
[0127] [000127] The "Fv" fragment contains a complete antigen-recognition and binding site consisting of a dimer of a variable domain of the heavy chain and one of the light chain in tight, non-covalent association. In this configuration, the three CDRs from each variable domain interact to define an antigen-binding site on the surface of the VH-VL dimer. Collectively, the six CDRs confer antigen-binding specificity to the antibody.
[0128] [000128] An "Fv single chain" or "scFv" antibody fragment is a single chain Fv variant comprising the VH and VL domains of an antibody, where the domains are present on a single polypeptide chain. The Fv of a single chain is able to recognize and bind the antigen. The scFv polypeptide may optionally also contain a polypeptide linker positioned between the VH and VL domains, to facilitate the formation of a desired three-dimensional structure for scFv binding to the antigen (see, for example, Pluckthun, 1994, In The Pharmacology of monoclonal Antibodies, Vol. 113, Rosenburg and Moore eds., Springer-Verlag, New York, pp. 269-315).
[0129] [000129] The other recognized antibody fragments include those that comprise a pair of Fd segments in series (VH-CH1-VH-CH1) to form a pair of antigen-binding regions. These "linear antibodies" can be bispecific or monospecific, as described, for example, in Zapata et al. 1995, Protein Eng. 8 (10): 1057-1062.
[0130] [000130] A humanized antibody or a humanized antibody fragment is a specific type of chimeric antibody that includes a variant of the immunoglobulin amino acid sequence, or its fragment, which is capable of binding to a predetermined antigen and which comprises a or more FRs having substantially the amino acid sequence of a human immunoglobulin and one or more CDRs having substantially the amino acid sequence of a non-human immunoglobulin. This non-human amino acid sequence, often referred to as an "import" sequence, is typically obtained from an "import" antibody domain, particularly a variable domain. In general, a humanized antibody includes at least the CDRs or HVLs of a non-human antibody, inserted between the FRs of a human heavy or light chain variable domain. The present invention describes specific humanized anti-CD40 antibodies that contain CDRs derived from the murine monoclonal antibodies shown in Tables 3 and 4, inserted between the FRs of the variable domains of the heavy and light chains of human germline sequences. It will be understood that certain murine RF residues may be important for the function of humanized antibodies and, therefore, certain residues of the variable domains of the heavy and light chains of human germline sequences are modified to be the same as those of the corresponding mouse sequence.
[0131] [000131] In another aspect, a humanized anti-CD40 antibody comprises substantially all, at least one, and typically two, variable domains (such as contained, for example, in Fab, Fab ', F (ab') 2 fragments, Fabc, and Fv), where all, or substantially all, CDRs correspond to those of a non-human immunoglobulin, and specifically in this document, all CDRs are rat sequences as detailed in Tables 1 through 4 here below, and all, or substantially all, FRs are those from a consensus or germline sequence of human immunoglobulin. In another aspect, a humanized anti-CD40 antibody also includes at least part of an immunoglobulin Fc region, typically that of a human immunoglobulin. Normally, the antibody will contain both the light chain and at least the variable domain of a heavy chain. The antibody can also include one or more of the CH1, hinge, CH2, CH3, and / or CH4 regions of the heavy chain, as appropriate.
[0132] [000132] A humanized anti-CD40 antibody can be selected from any class of immunoglobulins, including IgM, IgG, IgD, IgA and IgE, and any isotype, including IgG1, IgG2, IgG3, IgG4, IgA1 and IgA2. For example, the constant domain can be a complement complement fixing domain where the humanized antibody is desired to exhibit cytotoxic activity, and the isotype is typically IgG1. Where such cytotoxic activity is not desirable, the constant domain may be of another isotype, for example, IgG2. An alternative humanized anti-CD40 antibody may comprise sequences from more than one immunoglobulin class or isotype, and the selection of particular constant domains to optimize the desired effector functions is within the skill of the art. In specific embodiments, the present invention provides antibodies that are IgG1 antibodies and, more particularly, are IgG1 antibodies in which there is a knockout of effector functions.
[0133] [000133] The FRs and CDRs, or HVLs, of a humanized anti-CD40 antibody do not need to match parental sequences exactly. For example, one or more residues in the import CDR, or HVL, or following the FR consensus or germline may be altered (eg, mutagenized) by substitution, insertion or removal, such that the resulting amino acid residue it is no longer identical to the original residue in the corresponding position, in any parental sequence, but the antibody, nevertheless, retains the function of binding to CD40. Such a change will typically not be extensive and will be conservative changes. Typically, at least 75% of the humanized antibody residues will correspond to those of the consensus FR or germline sequences and the parental import CDR, more often at least 90%, and more often still more than 95%, or more than 98% or more than 99%.
[0134] [000134] The immunoglobulin residues that affect the interface between the variable regions of the heavy and light chains ("the VL-VH interface") are those that affect the proximity or the orientation of the two chains in relation to each other. Certain residues that may be involved in interactions between the chains include VL residues 34, 36, 38, 44, 46, 87, 89, 91, 96, and 98 and VH residues 35, 37, 39, 45, 47 , 91, 93, 95, 100, and 103 (using the numbering system presented in Kabat et al., Sequences of Proteins of Immunological Interest (National Institutes of Health, Bethesda, Md., 1987)). U.S. Pat. No. 6,407,213 also argues that residues, such as VL 43 and 85 residues, and VH 43 and 60 residues, may also be involved in this interaction. Although these residues are indicated for human IgG only, they are applicable across species. Important antibody residues, which are reasonably expected to be involved in interactions between the chains, are selected for substitution in the consensus sequence.
[0135] [000135] The terms "consensus sequence" and "antibody consensus" refer to an amino acid sequence comprising the amino acid residue that most frequently occurs at each position in all immunoglobulins of any particular class, isotype, or subunit structure , for example, a variable domain of human immunoglobulin. The consensus sequence can be based on immunoglobulins from a particular species or from many species. A "consensus" sequence, structure, or antibody is understood to include a human consensus sequence, as described in certain embodiments, and to refer to an amino acid sequence that comprises the amino acid residues that most frequently occur at each position in all immunoglobulins of any class, isotype, or structure of the particular subunit. Thus, the consensus sequence contains an amino acid sequence having at each position an amino acid that is present in one or more known immunoglobulins, but which may not exactly duplicate the entire amino acid sequence of any individual immunoglobulin. The consensus sequence of the variable region is not obtained from any naturally produced antibody or immunoglobulin. Kabat et al., 1991, Sequences of Proteins of Immunological Interest, 5th Ed. Public Health Service, National Institutes of Health, Bethesda, Md., And their variants. The FRs of the heavy and light chain consensus sequences, and their variants, provide useful sequences for the preparation of humanized anti-CD40 antibodies. See, for example, Pats. U.S. Nos. 6,037,454 and 6,054,297.
[0136] [000136] Human germline sequences are found naturally in the human population. A combination of those germline genes generates antibody diversity. The germline antibody sequences for the antibody light chain originate from the conserved human germline v and genes kappa and lambda genes. Similarly, the sequences for the heavy chains originate from the germline genes v, d and j (LeFranc, M-P, and LeFranc, G, "The Immunoglobulin Facts Book" Academic Press, 2001).
[0137] [000137] As used herein, "variant", "anti-CD40 variant", "humanized anti-CD40 variant" or "humanized anti-CD40 variant" each refer to a humanized anti-CD40 antibody having at least at least one heavy chain variable mouse CDR from any of the sequences of SEQ ID NO: 1 to 4 or a mouse chain sequence of the light chain derived from the mouse monoclonal antibody, as shown in any of SEQ ID NO: 5 up to SEQ ID NO: 8, and FR sequences derived from human consensus sequences. Variants include those having one or more amino acid changes in one or both of the variable domains of the light chain or heavy chain, provided that the change in the amino acid does not substantially impair the binding of the antibody to CD40. Illustrative humanized antibodies produced in this document include those designated as Antibody A, Antibody B and Antibody C, and the various sequences of the heavy and light chains thereof are shown in SEQ ID NOs 26 through SEQ ID NO: 40.
[0138] [000138] An "isolated" antibody is one that has been identified and separated and / or recovered from a component of its natural environment. The contaminating components of the antibody's natural environment are those materials that may interfere with the diagnostic or therapeutic uses of the antibody, and may be enzymes, hormones, or other proteinaceous or non-proteinaceous solutes. In one aspect, the antibody will be purified to at least more than 95% isolation by weight of the antibody.
[0139] [000139] An isolated antibody includes an antibody in situ within the recombinant cells in which it is produced, since at least one component of the antibody's natural environment will not be present. Typically, however, an isolated antibody will be prepared by at least one purification step in which the recombinant cell material is removed.
[0140] [000140] The term "antibody performance" refers to factors that contribute to the antibody's recognition of the antigen or the effectiveness of an antibody in vivo. Changes in the amino acid sequence of an antibody can affect the properties of the antibody, such as folding, and can influence physical factors, such as the initial rate of antibody binding to the antigen (ka), the antibody's dissociation constant. antigen (kd), the antibody affinity constant for the antigen (Kd), the conformation of the antibody, the stability of the protein, and the half-life of the antibody.
[0141] [000141] The term "epitope tagged", when used in this document, refers to an anti-CD40 antibody fused to an "epitope tag". An "epitope tag" is a polypeptide having a sufficient number of amino acids to provide an epitope for the production of the antibody, however it is designed in such a way that it does not interfere with the desired activity of the humanized anti-CD40 antibody. The epitope tag is usually sufficiently unique, such that an antibody raised against the epitope tag does not react substantially with other epitopes. Suitable tag polypeptides generally contain at least 6 amino acid residues and typically contain about 8 to 50 amino acid residues, or about 9 to 30 residues. Examples of epitope tags and the antibody that binds the epitope include the flu HA tag polypeptide and its 12CA5 antibody (Field et al., 1988 Mol. Cell. Biol. 8: 2159-2165); the c-myc tag and antibodies 8F9, 3C7, 6E10, G4, B7 and 9E10 to it (Evan et al., 1985, Mol. Cell. Biol. 5 (12): 3610-3616); and the glycoprotein D (gD) tag of the Herpes simplex virus and its antibody (Paborsky et al. 1990, Protein Engineering 3 (6): 547-553). In certain embodiments, the epitope tag is a "recovery receptor binding epitope". As used in this document, the term "recovery receptor binding epitope" refers to an epitope from the Fc region of an IgG molecule (such as IgG1, IgG2, IgG3, or IgG4) that is responsible for increasing the half- in vivo serum life of the IgG molecule.
[0142] [000142] In some embodiments, the antibodies of the present invention can be conjugated to a cytotoxic agent. This is any substance that inhibits or prevents the function of cells and / or causes the destruction of cells. The term is intended to include radioactive isotopes (such as I131, I125, Y90, and Re186), chemotherapeutic agents, and toxins, such as enzymatically active toxins of bacterial, fungal, plant, or animal origin, and fragments thereof . Such cytotoxic agents can be coupled to the humanized antibodies of the present invention using standard procedures, and used, for example, to treat a patient indicated for antibody therapy.
[0143] [000143] A "chemotherapeutic agent" is a chemical compound useful in the treatment of cancer. There are several examples of chemotherapeutic agents that could be conjugated to the therapeutic antibodies of the present invention. Examples of such chemotherapeutic agents include alkylating agents, such as a thiotepa and cyclophosphamide; alkyl sulfonates, such as busulfan, improsulfan, and piposulfan; aziridines, such as benzodopa, carboquone, meturedopa, and uredopa; ethylenimines and methylamelamines, including altretamine, triethylenomelamine, triethylenophosphoramide, triethylenethiophosphoramide, and trimethylolomelamine; acetogenins (especially bulatacin and bulatacinone); camptothecin (including the synthetic analogue topotecan); briostatin; calistatin; CC-1065 (including its synthetic analogues adozelesin, carzelesin, and bizelesin); cryptoficina (particularly cryptoficina 1 and criptoficina 8); dolastatin, auristatin, (including monomethyl auristatin E and monomethyl auristatin F analogs); duocarmycin (including synthetic analogs, KW-2189 and CBI-TMI); eleuterobin; pancratistatin; sarcodictiina; spongistatin; nitrogen mustards, such as chlorambucil, clomafazine, colophosphamide, estramustine, ifosfamide, meclorethamine, meclorethamine oxide hydrochloride, melphalan, novembiquine, phenesterine, prednimustine; trophosphamide, uracil mustard; nitrosureas, such as carmustine, chlorozotocin, photemustine, lomustine, nimustine, ranimustine; antibiotics, such as enediin antibiotics (for example, calicheamicin, especially calcamicin gamma1I and calicheamicin phiI1, see, for example, Agnew, Chem. Intl. Ed. Engl., 33: 183-186; including dinemicin A; bisphosphonates such as clodronate; speramycin; as well as the neocarzinoestatin chromophore and related chromophores of the enedinine chromoprotein), aclacinomysins, actinomycin, autaminicin, azaserine, cactomycin, bleomycin, carabicin, truckomycin, carzinophylline, chromomycins, dactinomycin, daunorubicin, detorubicin, 6-diazo-5-oxo-Lnorleucine, doxorubicin (Adriamycin®) (including morpholinodoxorubicin, cyanomorpholine-2 -pyrrolinodoxorubicin, and deoxidoxorubicin), epirrubucin, esorubicin, idarubicin, marcelomycin, mitomycins, such as mitomycin C, mycophenolic acid, nogalamycin, olivomycins, peplomycin, potfi romicin, puromycin, chelamycin, rhodorubicin, streptonigrin, streptozocin, tubercidin, ubenimex, zinostatin, zorubicin; antimetabolites, such as methotrexate and 5-fluorouracil (5-FU); folic acid analogs, such as denopterin, methotrexate, pteropterin, trimetrexate; purine analogs, such as fludarabine, 6-mercaptopurine, tiamiprine, thioguanine; pyrimidine analogs, such as ancitabine, azacytidine, 6-azauridine, carmofur, cytarabine, didesoxyuridine, doxifluridine, enocitabine, floxuridine; androgens, such as calusterone, dromostanolone propionate, epithiostanol, mepitiostane, testolactone; anti-adrenal drugs, such as aminoglutetimide, mitotane, trilostane; the folic acid supplier, such as frolinic acid; aceglatone; the aldophosphamide glycoside; aminolevulinic acid; eniluracil; ansacrine; bestrabucil; bisanthrene; edatraxate; defofamine; democolcin; diaziquone; elfomitine; ellipinium acetate; an epothilone; etoglucid; gallium nitrate; hydroxyurea; the lentinan; lonidamine; maytansinoids, such as maytansin and ansamitocins; mitoguazone, mitoxantrone; mopidamol; nitracrine; pentostatin; fenamet; pyrarrubicin; the losoxantrone; podophyllinic acid; 2-ethylhydrazide; procarbazine; the PSK®; razoxane; rhizoxin; sizofuran; spirogermanium; tenuazonic acid; triaziquone; 2.2 ', 2' '- trichlorotriethylamine; trichothecenes (especially T-2 toxin, verracurin A, roridin A and anguidine); urethane; vindesina; dacarbazine; manomustine; mitabronitol; mitolactol; the pipobroman; gacytosine; the arabinoside ("Ara-C"); cyclophosphamide; the thiotepa; taxoids, for example, paclitaxel (TAXOL®, Bristol-Myers Squibb Oncology, Princeton, N.J.) and doxetaxel (TAXOTERE®, Rhone-Poulenc Rorer, Antony, France); chlorambucil; gemcitabine (Gemzar®); 6-thioguanine; mercaptopurine; methotrexate; platinum analogues, such as cisplatin and carboplatin; vinblastine; platinum; etoposide (VP16); ifosfamide; the mitoxantrone; vincristine; vinorelbine Navelbine®); the new chair; the teniposide; edatrexate; daunomycin; aminopterin; the shilling; ibandronate; the CPT-11; the topoisomerase inhibitor RFS 2000; difluoromethylornithine (DMFO); retinoids, such as retinoic acid; capecitabine; and pharmaceutically acceptable salts, acids, or derivatives of any of the above. Also included in this definition are anti-hormonal agents that act to regulate or inhibit the hormone's action on tumors, such as antiestrogens and selective estrogen receptor modulators (SERMs), including, for example, tamoxifen (including Nolvadex®), raloxifene, droloxifene, 4-hydroxy tamoxifen, trioxifene, queoxifene, LY117018, onapristone, and toremifene (Fareston®); aromatase inhibitors that inhibit the aromatase enzyme, which regulates the production of estrogen in the adrenal glands, such as, for example, 4 (5) - imidazoles, aminoglutetimide, megestrol acetate (Megace®), exemestane, formestane, fadrozole, vorozole (Rivisor®), letrozole (Femara®), and anastrozole (Arimidex®); and antiandrogens, such as flutamide, nilutamide, bicalutamide, leuprolide, and goserelin; and pharmaceutically acceptable salts, acids, or derivatives of any of the above. Any one or more of these agents can be conjugated to the humanized antibodies of the present invention to provide a therapeutic agent useful for the treatment of various disorders.
[0144] [000144] Antibodies can also be conjugated to prodrugs. A "prodrug" is a form of precursor or derivative of a pharmaceutically active substance that is less cytotoxic to tumor cells compared to the parent drug and is capable of being enzymatically activated or converted to the most active form. See, for example, Wilman, 1986, "Prodrugs in Cancer Chemotherapy", In Biochemical Society Transactions, 14, pgs. 375- 382, 615th Meeting Belfast and Stella et al., 1985, "Prodrugs: A Chemical Approach to Targeted Drug Delivery, In:" Directed Drug Delivery, Borchardt et al., (Ed.), P. 247-267, Humana Press. Useful prodrugs include, but are not limited to, phosphate-containing prodrugs, thiophosphate-containing prodrugs, sulfate-containing prodrugs, peptide-containing prodrugs, D-amino acid-modified prodrugs, glycosylated prodrugs, -lactam-containing drugs, phenoxyacetamide-containing prodrugs, and optionally substituted phenoxyacetamide and optionally substituted. containing optionally substituted phenylacetamide, 5-fluorocytosine and other 5-fluoruridine prodrugs that can be converted to the most active cytotoxic free drug. Examples of cytotoxic drugs that can be derived in a prodrug form include, but are not limited to, those chemotherapeutic agents described above.
[0145] [000145] For diagnostic as well as therapeutic monitoring purposes, the antibodies of the invention can also be conjugated to a tag, or a tag alone, or a tag and a second additional agent (prodrug, chemotherapeutic agent and the like). A label, as distinguished from the other second agents, refers to an agent that is a detectable compound or composition and can be conjugated directly or indirectly to a humanized antibody of the present invention. The tag can itself be detectable (for example, radioisotope tags or fluorescent tags) or, in the case of an enzyme tag, it can catalyze the chemical change of a compound or substrate composition that is detectable. The labeled humanized anti-CD40 antibody can be prepared and used in a variety of applications, including in vitro and in vivo diagnostics.
[0146] [000146] The antibodies of the present invention can be formulated as part of a liposomal preparation, to effect their release in vivo. A "liposome" is a small vesicle made up of several types of lipids, phospholipids, and / or surfactant. Liposomes are useful for the delivery to a mammal of a compound or formulation, such as a humanized anti-CD40 antibody disclosed herein, optionally coupled to, or in combination with, one or more pharmaceutically active agents and / or tags. The components of the liposome are commonly arranged in a bilayer formation, similar to the lipid arrangement of biological membranes.
[0147] [000147] Certain aspects of the present invention relate to isolated nucleic acids that encode one or more domains of the humanized antibodies of the present invention. An "isolated" nucleic acid molecule is a nucleic acid molecule that is identified and separated from at least one contaminating nucleic acid molecule with which it is normally associated in the antibody's natural nucleic acid origin. An isolated nucleic acid molecule is distinguished from the nucleic acid molecule in that it exists in natural cells.
[0148] [000148] In several aspects of the present invention, one or more domains of the humanized antibodies will be expressed recombinantly. Such recombinant expression can employ one or more control sequences, i.e., polynucleotide sequences necessary for the expression of an operably linked coding sequence in a particular host organism. Control sequences, suitable for use in prokaryotic cells, include, for example, the promoter, operator, and ribosome binding site sequences. Eukaryotic control sequences include, but are not limited to, promoters, polyadenylation signals, and reinforcers. These control sequences can be used for the expression and production of humanized anti-CD40 antibody in prokaryotic and eukaryotic host cells.
[0149] [000149] A nucleic acid sequence is "operably linked" when it is placed in a functional relationship with another nucleic acid sequence. For example, a nucleic acid presequence or secretory leader is operably linked to a nucleic acid encoding a polypeptide if it is expressed as a preprotein that participates in the secretion of the polypeptide; a promoter or enhancer is operably linked to a coding sequence if it affects the transcription of the sequence; or a ribosome binding site is operably linked to a coding sequence if it is positioned so as to facilitate translation. Generally, "operably linked" means that the DNA sequences being linked are contiguous, and, in the case of a secret leader, contiguous and are on the reading board. However, reinforcers are optionally contiguous. The connection can be made by connection at convenient restriction sites. If such sites do not exist, synthetic oligonucleotide adapters or linkers can be used.
[0150] [000150] As used in this document, the expressions "cell", "cell line" and "cell culture" are used interchangeably and all such designations include their progeny. Thus, "transformants" and "transformed cells" include the primary exposed cell and cultures derived from it, regardless of the number of transfers.
[0151] [000151] The term "mammal", for treatment purposes, refers to any animal classified as a mammal, including humans, domesticated and farm animals, and zoo animals, for sports, or pets , such as dogs, horses, cats, cows, and the like. Preferably, the mammal is the human being.
[0152] [000152] A "disorder", as used in this document, is any condition that would benefit from treatment with a humanized anti-CD40 antibody described here. This includes chronic and acute disorders or diseases, including those pathological conditions that predispose the mammal to the disorder in question. Non-limiting examples of the disorders to be treated in this document include cancer, hematological malignancies, benign and malignant tumors, leukemias and lymphoid malignancies and inflammatory, angiogenic, autoimmune and immunological disorders.
[0153] [000153] The terms "cancer" and "cancerous" refer to, or describe, the physiological condition in mammals that is typically characterized by unregulated cell growth. Examples of cancer include, but are not limited to, carcinoma, lymphoma, blastoma, sarcoma, and leukemia.
[0154] [000154] As used in this document, the term "CD40-associated disorder" or "CD40-associated disease" refers to a condition in which the modification or elimination of cells expressing CD40 is indicated. These include cells that express CD40 showing abnormal proliferation or cells that express CD40 that are associated with cancerous or malignant growth. The most particular examples of cancers that demonstrate abnormal expression of the CD40 antigen include B lymphoblastoid cells, Burkitt's lymphoma, multiple myeloma, T-cell lymphomas, Kaposi's sarcoma, osteosarcoma, epidermal and endothelial tumors, cancers pancreatic, lung, breast, ovarian, colon, prostate, head and neck, skin (melanoma), bladder, and kidney. Such disorders include, but are not limited to, leukemias, lymphomas, including B-cell lymphoma and non-Hodgkin's lymphoma, multiple myeloma, Waldenstrom's macroglobulinemia; solid tumors, including sarcomas, such as osteosarcoma, Ewing's sarcoma, malignant melanoma, adenocarcinoma, including ovarian adenocarcinoma, Kaposi's sarcoma / Kaposi's tumor and squamous cell carcinoma.
[0155] [000155] A disorder associated with CD40 also includes diseases and disorders of the immune system, such as autoimmune disorders and inflammatory disorders. Such conditions include, but are not limited to, rheumatoid arthritis (RA), systemic lupus erythematosus (SLE), scleroderma, Sjogren's syndrome, multiple sclerosis, psoriasis, inflammatory bowel disease (eg, ulcerative colitis and Crohn's disease), inflammation pulmonary disease, asthma, and idiopathic thrombocytopenic purpura (ITP).
[0156] [000156] The term "for the growth of" or "growth inhibitory", when used in this document, refers to the inhibition of the growth or proliferation of a cell, especially a type of neoplastic cell expressing the CD40 antigen. Thus, growth inhibition, for example, significantly reduces the percentage of neoplastic cells in the S phase.
[0157] [000157] The term "intravenous infusion" refers to the introduction of an agent into an animal or human patient's vein over a period of time greater than approximately 15 minutes, usually between approximately 30 to 90 minutes.
[0158] [000158] The term "intravenous bolus" or "intravenous" push refers to an administration of drug into a vein of an animal or human being, such that the body receives the drug in approximately 15 minutes or less, usually 5 minutes or less.
[0159] [000159] The term "subcutaneous administration" refers to the introduction of an agent under the skin of an animal or human patient, preferably into a cavity between the skin and the underlying tissue, by relatively slow, continuous release from of a drug receptacle. Tightening or pulling the skin up and out of the underlying tissue can create the cavity.
[0160] [000160] The term "subcutaneous infusion" refers to the introduction of a drug under the skin of an animal or human patient, preferably into a cavity between the skin and the underlying tissue, by relatively slow, continuous release from of a drug receptacle, for a period of time that includes, but is not limited to, 30 minutes or less, or 90 minutes or less. Optionally, the infusion can be made by subcutaneous implantation of a drug delivery pump, implanted under the skin of the animal or human patient, where the pump releases a predetermined amount of drug, for a predetermined period of time, such as 30 minutes, 90 minutes, or a period of time exceeding the duration of the treatment regimen.
[0161] [000161] The term "subcutaneous bolus" refers to the administration of the drug under the skin of an animal or human patient, where the release of the bolus drug is less than approximately 15 minutes; in another aspect, less than 5 minutes, and in yet another aspect, less than 60 seconds. Also in yet another aspect, administration is within a cavity between the skin and the underlying tissue, where the cavity can be created by squeezing or pulling the skin up and out of the underlying tissue.
[0162] [000162] The term "therapeutically effective amount" is used to refer to an amount of an active agent that alleviates or improves one or more of the symptoms of the disorder being treated. In doing so, it is this amount that has a beneficial result on the patient, for example, an effect of stopping the growth or causing the elimination of the cell. In one aspect, the therapeutically effective amount has apoptotic activity, or is capable of inducing cell death. In another aspect, the therapeutically effective amount refers to a concentration of target serum that has been shown to be effective in, for example, slowing the progression of the disease. Effectiveness can be measured in conventional ways, depending on the condition to be treated. For example, in diseases or neoplastic disorders, characterized by cells expressing CD40, effectiveness can be measured by assessing the time until disease progression, or by determining response rates.
[0163] [000163] The terms "treatment" and "therapy" and the like, as used herein, are intended to include therapeutic as well as prophylactic or suppressive measures for a disease or disorder, resulting in any clinically desirable or beneficial effect, including , but not limited to the relief or reduction of one or more symptoms, regression, reduction or cessation of the progression of the disease or disorder. Thus, for example, the term treatment includes the administration of an agent before, or after, the onset of a symptom of a disease or disorder, thereby preventing or removing one or more signs of the disease or disorder. As another example, the term includes the administration of an agent after the clinical manifestation of the disease, to combat the symptoms of the disease. In addition, administration of an agent after onset and after clinical symptoms have developed, where administration affects the clinical parameters of the disease or disorder, such as the degree of tissue damage or the amount or extent of metastasis, either whether or not treatment results in improvement of the disease, comprises "treatment" or "therapy" as used in this document. Furthermore, provided that the compositions of the invention, alone or in combination with another therapeutic agent, alleviate or ameliorate at least one symptom of a disorder being treated, compared to that symptom in the absence of the use of the humanized CD40 antibody composition, the result should be considered an effective treatment of the basic disorder, regardless of whether all symptoms of the disorder are relieved or not.
[0164] [000164] The term "package insert" is used to refer to the instructions normally included in commercial packaging of therapeutic products, which contain information on the indications, use, administration, contraindications and / or warnings regarding the use of such therapeutic products. Antibodies.
[0165] [000165] Humanized anti-CD40 antibodies, and compositions and articles of manufacture comprising one or more humanized anti-CD40 antibodies of the present invention are described and disclosed. Binding agents that include an antigen-binding fragment of a humanized anti-CD40 antibody are also described. Humanized anti-CD40 antibodies and binding agents can stop cell growth, cause CD40-expressing cells to be eliminated, or otherwise induce or cause a cytotoxic or cytostatic effect on target cells. Humanized anti-CD40 antibodies and binding agents can be used to treat a variety of diseases or disorders characterized by the proliferation of cells that express the CD40 surface antigen. A humanized anti-CD40 antibody and a CD40 binding agent each include at least one part that specifically recognizes a CD40 epitope (i.e., an antigen binding fragment).
[0166] [000166] In the initial characterization, murine antibodies were selected based on the characterization of binding to CD40.
[0167] [000167] From these initial studies, murine antibodies were selected that had the following variable regions of the heavy chain shown in Table 1 and the variable regions of the light chain shown in Table 2:
[0168] [000168] Human structure sequences were selected for each of the mouse leaders, based on the structure homology, the CDR structure, the main conserved residues, the conserved interface cluster residues and other parameters.
[0169] [000169] The CDRs of the heavy chain and the light chain of mice of the various murine antibodies selected are shown in Table 3 and Table 4, respectively:
[0170] [000170] The H-CDR1 listed above is using the sequence that uses the Chothia numbering system (Al-Lazikani et al., (1997) JMB 273,927-948). The Kabat numbering for the strings is indicated by the bold italic text, and the IMGT numbering is shown by the underlined text of the residues in the table mentioned above for CDR1 and CDR2. The sequences for H-CDR3 for each of 2H11, 10F2 and 19B10 are TTSYYVGTYGY (SEQ ID NO: 77) and for 20E2 it is ARQDGYRYAMDY (SEQ ID NO: 78).
[0171] [000171] Again, the Chothia numbering system is used in Table 4, with the Kabat numbering for the strings being indicated by the italicized text, in bold, and the IMGT numbering is shown by the underlined text.
[0172] [000172] Fabs that showed better or equal binding compared to Fab of chimeric origin were selected for conversion to IgG. The clones from the 20E2 series have been converted into two different IgG formats: a) IgG4DM (double mutant) has two mutations in the Fc / joint region, Ser228Pro, which reduces the formation of semimolecules, and Leu235Glu, which further reduces binding to FcγR b) IgG1KO (knockout of effector functions) has two mutations in the Fc region, Leu234Ala and Leu235Ala, which reduce effector function, such as binding to FcγR and complement. Both formats of IgG are described in the literature. Example 1 describes the humanization of three candidates in more detail. The results of such humanization have resulted in humanized antibody sequences, which have the heavy and light chain sequences shown below:
[0173] [000173] In some embodiments, the antigen-binding fragment can, for example, block proliferation or otherwise stop the growth of a cell or cause its depletion, death, or its elimination, for example, by binding the CD40 surface antigen. For example, in malignancies of T and B cells, antitumor effects often result (for example, growth arrest, with or without elimination or apoptosis) when the malignant cells are exposed to stimuli that result in the activation of normal lymphocytes. This activation-induced growth arrest has been observed with signals through antigen receptors or co-stimulatory receptors (see, for example, Ashwell et al., 1987, Science 237: 61; Bridges et al., 1987, J. Immunol. 139: 4242; Page and Defranco, 1988, J. Immunol. 140: 3717; and Beckwith et al., 1990, J. Natl. Cancer Inst. 82: 501). Stimulation of CD40, as a result of specific binding by antibody or soluble ligand, inhibits the growth of B cell lymphomas (see, for example, Funakoshi et al., 1994, Blood 83: 2787-2794). Agents that inhibit the growth of malignant cells in this mode and that are directed against the CD40 surface antigen are examples of suitable agents.
[0174] [000174] CD40-specific agents include an antigen-binding fragment of a humanized anti-CD40 antibody that binds to CD40 (for example, human CD40 or a variant thereof). CD40-specific agents and antibodies can optionally be conjugated to, or fused to, a cytotoxic or chemotherapeutic agent. In aspects where the humanized antibody binds to the CD40 surface antigen and causes the elimination of cell types that express CD40, the binding is generally characterized by targeting the CD40 surface antigen cell in vivo. Suitable binding agents bind the CD40 antigen with sufficient affinity and / or avidity, such that the CD40-specific agent is useful as a therapeutic agent specifically targeting a cell expressing the antigen.
[0175] [000175] In some ways, the humanized antibody decreases the binding of the CD40 ligand to CD40 by at least 45%, at least 50%, at least 60% or at least 75% or at least 80%, or at least least 90%, or at least 95%.
[0176] [000176] In some embodiments, humanized anti-CD40 antibodies, including their antigen-binding fragments, such as the heavy and light chain variable domains, comprise an amino acid sequence of residues derived from Antibody A CDRs (sequence of heavy chain = SEQ ID NO: 27; SEQ ID NO: 28; SEQ ID NO: 29 or SEQ ID NO: 30; light chain sequence = SEQ ID NO: 26), Antibody B (heavy chain sequence = SEQ ID NO : 32; SEQ ID NO: 33; SEQ ID NO: 34; or SEQ ID NO: 35; light chain sequence = SEQ ID NO: 31) and Antibody C (heavy chain sequence = SEQ ID NO: 37; SEQ ID NO: 38; SEQ ID NO: 39 or SEQ ID NO: 40; light chain sequence = SEQ ID NO: 36;) described herein above and amino acid residues derived from framework regions of a human immunoglobulin. Humanized anti-CD40 antibodies optionally include specific amino acid substitutions in regions of consensus structure or germline.
[0177] [000177] Specific substitution of amino acid residues at these positions in the structure can improve several aspects of antibody performance, including binding affinity and / or stability, over that demonstrated in humanized antibodies formed by "direct exchange" of CDRs or HVLs in the human germline structure regions, as shown in the examples below.
[0178] [000178] In some embodiments, the present invention describes other monoclonal antibodies with heavy chain (VH) sequences from SEQ ID NO: 1 to SEQ ID NO: 4 and light chain (VL) sequences from SEQ ID NO: 5 to SEQ ID NO: 8 (see Tables 1 and 2 above). The CDR sequence of these murine antibodies is shown in Tables 3 and 4 placing such CDRs in the FRs of the variable domains of the human consensus heavy and light chains will produce the useful humanized antibodies of the present invention.
[0179] [000179] In some specific embodiments, the humanized anti-CD40 antibodies disclosed in this document comprise at least one variable domain of the heavy or light chain comprising the CDRs or HVLs of the murine monoclonal antibodies, as shown in Tables 1 through 4 above, and the FRs of the variable domains of the heavy and light chains of the human germline. In the illustrative modalities, the humanized antibodies created in this document are: Antibody A, Antibody B and Antibody C, and the various sequences of the heavy and light chains of the same are shown in SEQ ID NOs 26 through SEQ ID NO: 40.
[0180] [000180] In the specific embodiments, antibodies are contemplated that have a heavy chain sequence of any of SEQ ID NO: 27, SEQ ID NO: 28, SEQ ID NO: 29 or SEQ ID NO: 30 in combination with a chain sequence of SEQ ID NO: 26. Alternative antibodies include those that have a SEQ ID NO: 32, SEQ ID NO: 33, SEQ ID NO: 34 or SEQ ID NO SEQ ID NO: 35 heavy chain sequence, in combination with a SEQ light chain sequence ID NO: 31. In still further embodiments, humanized antibodies are provided which have a heavy chain sequence of SEQ ID NO: 37, SEQ ID NO: 38; SEQ ID NO: 39 or SEQ ID NO: 40, in combination with a light chain sequence of SEQ ID NO: 36.
[0181] [000181] The CDRs of these sequences are shown in Tables 3 and 4. In the specific modalities, it is contemplated that chimeric antibodies with the CDR regions exchanged (that is, for example, exchanging one or two CDRs of Antibody A for CDR analogous to that of Antibody C) among these illustrative immunoglobulins can produce useful antibodies.
[0182] [000182] In certain embodiments, the humanized anti-CD40 antibody is an antibody fragment. The various antibody fragments have, in general, been discussed above and there are techniques that have been developed for the production of antibody fragments. The fragments can be derived by means of proteolytic digestion of intact antibodies (see, for example, Morimoto et al., 1992, Journal of Biochemical and Biophysical Methods 24: 107-117; and Brennan et al., 1985, Science 229: 81 ). Alternatively, the fragments can be produced directly in recombinant host cells. For example, Fab'-SH fragments can be directly recovered from E. coli and chemically coupled to form F (ab ') 2 fragments (see, for example, Carter et al., 1992, Bio / Technology 10: 163-167 ). By another approach, F (ab ') 2 fragments can be isolated directly from recombinant host cell culture. The other techniques for producing antibody fragments will be apparent to the skilled practitioner.
[0183] [000183] Certain embodiments include an F (ab ') 2 fragment of a humanized anti-CD40 antibody comprising a heavy chain sequence of any of SEQ ID NO: 27, SEQ ID NO: 28, SEQ ID NO: 29 or SEQ ID NO: 30 in combination with a light chain sequence of SEQ ID NO: 26. Alternative antibodies include those that have a heavy chain sequence of SEQ ID NO: 32, SEQ ID NO: 33, SEQ ID NO: 34 or SEQ ID NO: 35, in combination with a light chain sequence of SEQ ID NO: 31. In still further embodiments, humanized antibodies are provided which have a heavy chain sequence of SEQ ID NO: 37, SEQ ID NO: 38; SEQ ID NO: 39 or SEQ ID NO: 40, in combination with a light chain sequence of SEQ ID NO: 36. Such modalities can include an intact antibody comprising such F (ab ') 2.
[0184] [000184] In some embodiments, the antibody or antibody fragment includes a constant region that mediates the effector function. The constant region can provide antibody-dependent cell cytotoxicity (ADCC), antibody-dependent cell phagocytosis (ADCP) and / or complement-dependent cytotoxicity (CDC) against a target cell that expresses CD40. The effector domain (s) can, for example, be an Fc region of an Ig molecule. Typically, the CD40-binding agent recruits and / or activates cytotoxic leukocytes (for example, natural killer cells (NK), phagocytic cells (for example, macrophages), and / or serum complement components).
[0185] [000185] The effector domain of an antibody can be from any suitable vertebrate animal species and isotypes. The isotypes of different animal species differ in their ability to mediate effector functions. For example, the ability of human immunoglobulin to mediate CDC and ADCC / ADCP is generally in the order of IgG1≈IgG3> IgG2 / IgM / IgG4, respectively. The immunoglobulins of mice mediate CDC and ADCC / ADCP generally in the order of IgM≈IgG3 >> IgG2b> IgG2a >> IgG1 and IgG2b> IgG2a> IgG1 >> IgG3 of mice, respectively. In another example, mouse IgG2a measured ADCC, while both mouse IgG2a and IgM measured CDC. Antibody Modifications
[0186] [000186] Humanized anti-CD40 antibodies and agents can include modifications of the humanized anti-CD40 antibody or its antigen binding fragment. For example, it may be desirable to modify the antibody in relation to the effector function, in order to increase the effectiveness of the antibody in the treatment of cancer. Such modification is the introduction of cysteine residue (s) in the Fc region, thereby allowing disulfide bond formation between the chains in this region. The homodimeric antibody thus generated may have improved internalization capacity and / or complement-mediated cell death and / or increased antibody-dependent cell cytotoxicity (ADCC). See, for example, Caron et al., 1992, J. Exp Med. 176: 1191-1195; and Shopes, 1992, J. Immunol. 148: 2918-2922. Homodimeric antibodies having increased antitumor activity can also be prepared using heterobifunctional crosslinkers, as described in Wolff et al., 1993, Cancer Research 53: 2560-2565. Alternatively, an antibody can be engineered to contain double Fc regions, increasing complement lysis and the ADCC capabilities of the antibody. See Stevenson et al., 1989, Anti-Cancer Drug Design 3: 219-230.
[0187] [000187] Antibodies with improved ability to support ADCC have been generated by modifying the glycosylation pattern of their Fc region. This is possible since the glycosylation of the antibody in the asparagine residue, N297, in the CH2 domain is involved in the interaction between the IgG and Fc receptors, a prerequisite for ADCC. Host cell lines have been engineered to express antibodies with altered glycosylation, such as increased bisection nacetylglycosamine or reduced fucose. The reduction of fucose provides a greater increase in ADCC activity than does the increase in the presence of bisection N-acetylglycosamine. In addition, the increase in ADCC by antibodies with little fucose is independent of the FcγRIIIa V / F polymorphism.
[0188] [000188] Modification of the amino acid sequence of the Fc region of the antibodies is an alternative to glycosylation engineering to increase ADCC. The IgG1 binding site for Fcγ receptors was determined by extensive mutational analysis. This resulted in the generation of humanized IgG1 antibodies with Fc mutations that increase binding affinity for FcγRIIIa and increase ADCC in vitro. In addition, Fc variants with many different permutations of binding properties have been obtained, for example, improved binding to specific FcγR receptors with unchanged or decreased binding to other FcγR receptors.
[0189] [000189] Another aspect includes immunoconjugates comprising the humanized antibody or fragments conjugated to a cytotoxic agent, such as a chemotherapeutic agent, a toxin (e.g., an enzymatically active toxin of bacterial, fungal, plant, or animal origin, or fragments), or a radioactive isotope (ie, a radioconjugate).
[0190] [000190] Chemotherapeutic agents useful in the generation of such immunoconjugates have been described above. Enzymatically active toxins and fragments that can be used to form useful immunoconjugates include diphtheria A chain, active diphtheria toxin non-binding fragments, exotoxin A chain (from Pseudomonas aeruginosa), ricin A chain, the abrin A chain, the modecin A chain, the alpha-sarcin, the Aleurites fordii proteins, the diantina proteins, the American Phytolaca proteins (PAPI, PAPII, and PAP-S), the inhibitor of Momordica charantia, the curcine, crotin, the Sapaonaria officinalis inhibitor, gelonin, mitogelin, restrictocin, phenomycin, enomycin, trichothecenes, and the like. A variety of radionuclides are available for the production of radioconjugated humanized anti-CD40 antibodies. Examples include 212Bi, 131I, 131In, 90Y, and 186Re.
[0191] [000191] The conjugates of the humanized anti-CD40 antibody and the cytotoxic or chemotherapeutic agent can be prepared by known methods, using a variety of bifunctional protein coupling agents, such as N-succinimidyl-3- (2-pyridyldithiol) propionate ) (SPDP), iminothiolane (IT), bifunctional derivatives of imidoesters (such as dimethyl adipimidate HCL), active esters (such as disuccinimidyl suberate), aldehydes (such as glutaraldehyde), compounds of bis-azido (such as bis (p-azidobenzoyl) hexanediamine), bisdiazonium derivatives (such as bis- (p-diazoniobenzoyl) -ethylenediamine), diisocyanates (such as toluene 2,6-diisocyanate), and bis-active fluorine compounds (such as 1,5-difluor-2,4-dinitrobenzene). For example, a ricin immunotoxin can be prepared as described in Vitetta et al., 1987, Science 238: 1098. Carbon-labeled 1-isothiocyanatobenzyl-3-methylldiethylene (MX-DTPA) 14 is an illustrative chelating agent for the conjugation of the radionuclide to the antibody. Conjugates can also be formed with a cleavable linker.
[0192] [000192] In another embodiment, the antibody can be conjugated to a "receptor" (such as streptavidin) for use in pre-targeting the tumor. In this procedure, the anticorporeceptor conjugate is administered to a patient, followed by removal of the unbound conjugate from the circulation using a purifying agent and then administration of a "ligand" that selectively binds the receptor (for example, avidin), the ligand being conjugated to a cytotoxic agent (for example, a radionuclide).
[0193] [000193] The humanized anti-CD40 antibodies disclosed in this document can also be formulated as immunoliposomes. Liposomes containing the antibody are prepared by methods known in the art, as described in Epstein et al., 1985, Proc. Natl. Acad. Sci. USA 82: 3688; Hwang et al., 1980, Proc. Natl. Acad. Sci. USA 77: 4030; and in Pats. U.S. Nos. 4,485,045 and 4,544,545. Liposomes having increased circulation time are disclosed, for example, in U.S. Pat. No. 5,013,556.
[0194] [000194] Particularly useful liposomes can be generated by the reverse phase evaporation method, with a lipid composition comprising phosphatidylcholine, cholesterol and PEG-derived phosphatidylethanolamine (PEG-PE). Liposomes are extruded through filters of defined pore size, to produce liposomes with the desired diameter. Fab 'fragments of an antibody disclosed in this document can be conjugated to liposomes as described in Martin et al., 1982, J. Biol. Chem. 257: 286-288, through a disulfide exchange reaction. A chemotherapeutic agent (such as doxorubicin) is optionally contained in the liposome. See, for example, Gabizon et al., 1989, J. National Cancer Inst. 81 (19): 1484.
[0195] [000195] The antibodies described and disclosed in this document can also be used in ADEPT (Antibody Directed Enzyme Therapy) conjugating the antibody to a prodrug activating enzyme that converts a prodrug (for example, a chemotherapeutic agent from peptidyl) in an active anticancer drug. See, for example, WO 81/01145, WO 88/07378, and Pat. No. 4,975,278. The enzyme component of the immunoconjugate, useful for ADEPT, is an enzyme capable of acting on a prodrug in such a way as to convert it into its more active cytotoxic form. Specific enzymes that are useful in ADEPT include, but are not limited to, alkaline phosphatase to convert phosphate-containing prodrugs into free drugs, arylsulfatase to convert sulfate-containing prodrugs into free drugs; cytosine deaminase to convert non-toxic 5-fluorocytosine into the anticancer drug, 5-fluorouracil; proteases, such as serratia protease, thermolysin, subtilisin, carboxypeptidases, and cathepsins (such as cathepsins B and L), to convert peptide-containing prodrugs into free drugs; D-alanylcarboxypeptidases, for converting prodrugs containing D-amino acid substituents; enzymes that cleave carbohydrates, such as β-galactosidase and neuraminidase, to convert glycosylated prodrugs into free drugs; β-lactamase, to convert drugs derived with β-lactams into free drugs; and penicillin amidase, such as penicillin V amidase or penicillin G amidase, to convert the derived drugs into their amine nitrogens with phenoxyacetyl or phenylacetyl groups, respectively, into free drugs. Alternatively, antibodies having enzymatic activity ("abzymes") can be used to convert the prodrugs into free active drugs (see, for example, Massey, 1987, Nature 328: 457-458). The antibody-enzyme conjugates can be prepared by known methods for the release of the enzyme to a population of tumor cells, for example, covalently linking the enzyme to the humanized anti-CD40 antibody / heterobifunctional cross-linking reagents discussed above. Alternatively, fusion proteins comprising at least the antigen-binding region of an antibody disclosed herein, linked to at least a functionally active part of an enzyme as described above can be constructed using recombinant DNA techniques (see, for example, Neuberger et al., 1984, Nature 312: 604-608).
[0196] [000196] In certain embodiments, it may be desirable to use an anti-CD40 antibody fragment, instead of an intact antibody, to increase tumor penetration, for example. It may be desirable to modify the antibody fragment to increase its serum half-life. This can be achieved, for example, by incorporating a recovery receptor-binding epitope into the antibody fragment. In one method, the appropriate region of the antibody fragment can be altered (for example, mutated), or the epitope can be incorporated into a peptide tag that is then fused to the antibody fragment at the end or in the middle, for example, through of DNA or peptide synthesis. See, for example, WO 96/32478.
[0197] [000197] In other embodiments, covalent modifications of the humanized anti-CD40 antibody are also included. Covalent modifications include the modification of cysteinyl residues, histidyl residues, lysinyl and amino terminal residues, arginyl residues, tyrosyl residues, carboxyl side groups (aspartyl or glutamyl), glutaminyl and asparaginyl residues, or seryl residues , or threonyl. Another type of covalent modification involves chemically or enzymatically coupling glycosides to the antibody. Such modifications can be made by chemical synthesis or by enzymatic or chemical cleavage of the antibody, if applicable. Other types of covalent modifications of the antibody can be introduced into the molecule by reacting targeted amino acid residues of the antibody with an organic derivative agent that is capable of reacting with selected side chains or the terminal amino or carboxy residues.
[0198] [000198] The removal of any portions of carbohydrates present on the antibody can be carried out chemically or enzymatically. Chemical deglycosylation is described by Hakimuddin et al., 1987, Arch. Biochem. Biophys. 259: 52 and by Edge et al., 1981, Anal. Biochem., 118: 131. The enzymatic cleavage of the carbohydrate moieties on the antibodies can be achieved by using a variety of endo- and exoglycosidases, as described by Thotakura et al., 1987, Meth. Enzymol 138: 350.
[0199] [000199] Another type of useful covalent modification comprises binding the antibody to one of a variety of non-proteinaceous polymers, for example, polyethylene glycol, polypropylene glycol, or polyoxyalkylenes, in the manner presented in one or more of Pat. No. 4,640,835, Pat. No. 4,496,689, Pat. No. 4,301,144, Pat. No. 4,670,417, Pat. No. 4,791,192 and U.S. Pat. No. 4,179,337. Humanization and Variants of Amino Acid Sequences
[0200] [000200] The amino acid sequence variants of the anti-CD40 antibody can be prepared by introducing appropriate nucleotide changes into the DNA of the anti-CD40 antibody, or by peptide synthesis. Such variants include, for example, the removal of, and / or insertions in and / or substitutions of, residues within the amino acid sequences of the anti-CD40 antibodies of the examples contained herein. Any combination of removals, insertions, and replacements is made to arrive at the final construction, as long as the final construction has the desired characteristics. Amino acid changes can also alter the post-institutional processes of the humanized or variant anti-CD40 antibody, such as changing the number or position of glycosylation sites.
[0201] [000201] A useful method for identifying certain residues or regions of the anti-CD40 antibody that are preferred positions for mutagenesis is called "alanine scan mutagenesis", as described by Cunningham and Wells (Science, 244: 1081-1085 (1989)). Here, a target residue or group of residues is identified (for example, charged residues, such as arg, asp, his, lys, and glu) and replaced with a neutral or negatively charged amino acid (typically alanine), to affect the interaction of the amino acids with the CD40 antigen. These amino acid positions demonstrating functional sensitivity to substitutions are then refined by introducing additional variants or other variants at, or for, the substitution sites. Thus, although the site for introducing a variation in the amino acid sequence is predetermined, the nature of the mutation per se does not need to be predetermined. For example, to analyze the performance of a mutation at a given site, alanine scan or random mutagenesis is conducted on the target codon or region and the expressed anti-CD40 antibody variants are examined for the desired activity.
[0202] [000202] Amino acid sequence inserts include terminal amino and / or carboxyl fusions that vary in length from a residue to polypeptides containing one hundred or more residues, as well as insertions within the individual or multiple amino acid residue sequences. Examples of terminal inserts include an anti-CD40 antibody fused to an epitope tag. Other variants of insertion of the anti-CD40 antibody molecule include a fusion at the N or C end of the anti-CD40 antibody of an enzyme or a polypeptide that increases the serum half-life of the antibody.
[0203] [000203] Another type of variant is an amino acid substitution variant. These variants have at least one amino acid residue in the anti-CD40 antibody molecule removed and a different residue inserted in its place. The sites of greatest interest for substitution mutagenesis include hypervariable regions, but changes in RF are also contemplated. Conservative substitutions are shown in Table 5 under the heading "preferred substitutions". If such substitutions result in a change in biological activity, then further substantial changes, called "illustrative substitutions", or as further described below in reference to the classes of amino acids, can be introduced and the products examined.
[0204] (1) hidrofóbicos: norleucina, met, ala, val, leu, ile; (2) hidrofílicos neutros: cys, ser, thr; (3) ácidos: asp, glu; (4) básicos: asn, gin, his, lys, arg; (5) resíduos que influenciam a orientação da cadeia: gly, pro; e (6) aromáticos: trp, tyr, phe. [000204] In protein chemistry, it is generally accepted that the biological properties of the antibody can be accomplished by selecting substitutions that differ significantly in their effect on maintaining (a) the structure of the polypeptide backbone in the substitution area, for example, as a leaf or helical conformation, (b) the charge or hydrophobicity of the molecule at the target site, or (c) the size of the side chain. Naturally occurring residues are divided into groups based on the common properties of the side chains: (1) hydrophobic: norleucine, met, ala, val, leu, ile; (2) neutral hydrophilic: cys, ser, thr; (3) acids: asp, glu; (4) basic: asn, gin, his, lys, arg; (5) residues that influence the orientation of the chain: gly, pro; and (6) aromatics: trp, tyr, phe.
[0205] [000205] Non-conservative substitutions will imply exchanging a member of one of these classes for another class.
[0206] [000206] Any cysteine residue not involved in maintaining the appropriate conformation of the humanized anti-CD40 antibody or variant can also be replaced, usually by serine, to improve the oxidative stability of the molecule, prevent abnormal cross-linking, or provide established points of conjugation to a cytotoxic or cytostatic compound. Conversely, cysteine bond (s) can be added to the antibody to improve its stability (particularly where the antibody is an antibody fragment, such as an Fv fragment).
[0207] [000207] One type of substitution variant involves replacing one or more residues in the hypervariable region of an antibody of origin (for example, a humanized or human antibody). Generally, the resulting variant (s), selected for further development, will have improved biological properties over the source antibody from which it (s) is (are) generated (s). A convenient way to generate such substitution variants is affinity maturation using phage exposure. Briefly, several sites in the hypervariable region (for example, 6-7 sites) are mutated to generate all possible amino substitutions at each site. The antibody variants thus generated are exposed in a monovalent manner from filamentous phage particles as fusions to the M13 gene III product packed within each particle. Variants exposed in the phages are then examined for their biological activity (for example, binding affinity). To identify sites in the hypervariable region that are candidates for modification, alanine scanning mutagenesis can be performed to identify residues in the hypervariable region that contribute significantly to antigen binding. Alternatively, or in addition, it may be beneficial to analyze a crystal structure of the antigen-antibody complex to identify points of contact between the antibody and human CD40. Such contact wastes and the nearby wastes are candidates for replacement according to the techniques elaborated in this document. Once such variants are generated, the panel of variants is subjected to examination, as described in this document, and antibodies with superior properties in one or more relevant assays can be selected for further development.
[0208] [000208] Another type of amino acid variant of the antibody alters the original glycosylation pattern of the antibody. By "altering" it is intended to remove one or more portions of carbohydrates found in the antibody, and / or to add one or more glycosylation sites that are not present in the antibody.
[0209] [000209] In some embodiments, it may be desirable to modify the antibodies of the invention to add glycosylation sites. Glycosylation of antibodies is typically N-linked or O-linked. N-linked refers to the joining of the carbohydrate moiety to the side chain of an asparagine residue. The asparagine-X-serine and asparagine-X-threonine tripeptide sequences, where X is any amino acid except proline, are the recognition sequences for the enzymatic union of the carbohydrate moiety to the asparagine side chain. Thus, the presence of any of these tripeptide sequences in a polypeptide creates a potential glycosylation site. O-linked glycosylation refers to the union of one of the N-acetylgalactosamine, galactose, or xylose sugars to a hydroxyamino acid, most commonly serine or threonine, although 5-hydroxyproline or 5-hydroxylisine can also be used. Thus, to glycosylate a given protein, for example, an antibody, the amino acid sequence of the protein is engineered to contain one or more of the tripeptide sequences described above (for the N-linked glycosylation sites). The change can also be made by adding, or replacing, one or more serine or threonine residues to the original antibody sequence (for O-linked glycosylation sites).
[0210] [000210] The nucleic acid molecules encoding the amino acid sequence variants of the anti-CD40 antibody are prepared by a variety of methods known in the art. These methods include, but are not limited to, isolation from a natural source (in the case of naturally occurring amino acid sequence variants) or preparation by oligonucleotide-mediated (or site-directed) mutagenesis, PCR mutagenesis, and cassette mutagenesis. a variant prepared in advance or a non-variant version of the anti-CD40 antibody. Polynucleotides, Vectors, Host Cells, and Recombinant Methods
[0211] [000211] Other modalities include isolated polynucleotides that comprise a sequence encoding a humanized anti-CD40 antibody, the vectors, and host cells comprising the polynucleotides, and recombinant techniques for producing the humanized antibody. Isolated polynucleotides can encode any desired form of the anti-CD40 antibody, including, for example, life-size monoclonal antibodies, Fab, Fab ', F (ab') 2 fragments, and Fv, diabodies, linear antibodies, antibody molecules from a single chain, and multispecific antibodies formed from antibody fragments.
[0212] [000212] Some embodiments include isolated polynucleotides comprising the sequences encoding an antibody or antibody fragment having the amino acid sequence of the heavy chain variable region of any of SEQ ID NO: 1 to 4, SEQ ID NO: 27, SEQ ID NO: 28, SEQ ID NO: 29, SEQ ID NO: 30, SEQ ID NO: 32, SEQ ID NO: 33, SEQ ID NO: 34, SEQ ID NO: 35, SEQ ID NO: 37, SEQ ID NO: 38, SEQ ID NO: 39, or SEQ ID NO: 40. Some embodiments include isolated polynucleotides comprising sequences that encode an antibody or antibody fragment having the amino acid sequence of the light chain variable domain of SEQ ID NO: 26, SEQ ID NO: 31, or SEQ ID NO: 36.
[0213] [000213] In one aspect, the sequence (s) of the isolated polynucleotides encodes an antibody or antibody fragment having a heavy chain variable domain and a light chain variable region comprising the amino acid sequences of SEQ ID NO: 27 and SEQ ID NO: 26, respectively; SEQ ID NO: 28 and SEQ ID NO: 26, respectively; SEQ ID NO: 29 and SEQ ID NO: 26, respectively; SEQ ID NO: 30 and SEQ ID NO: 26, respectively; SEQ ID NO: 32 and SEQ ID NO: 31, respectively; SEQ ID NO: 33 and SEQ ID NO: 31, respectively; SEQ ID NO: 34 and SEQ ID NO: 31, respectively; SEQ ID NO: 35 and SEQ ID NO: 31, respectively; SEQ ID NO: 37 and SEQ ID NO: 36, respectively; SEQ ID NO: 38 and SEQ ID NO: 36, respectively; SEQ ID NO: 39 and SEQ ID NO: 36, respectively; SEQ ID NO: 40 and SEQ ID NO: 36, respectively.
[0214] [000214] The polynucleotide (s) comprising a sequence encoding a humanized anti-CD40 antibody or a fragment or chain thereof may be fused to one or more regulatory or control sequences, as is known in the art, and may be contained in expression vectors or the appropriate host cell, as is known in the art. Each of the polynucleotide molecules encoding the variable domains of the heavy or light chains can be independently fused to a sequence of polynucleotides encoding a constant domain, such as a human constant domain, enabling the production of intact antibodies. Alternatively, the polynucleotides, or parts of them, can be fused together, providing a template for the production of a single chain antibody.
[0215] [000215] For recombinant production, a polynucleotide encoding the antibody is inserted into a replicable vector for cloning (amplification of DNA) or for expression. Many suitable vectors are available to express the recombinant antibody. The components of the vector generally include, but are not limited to, one or more of the following: a signal sequence, an origin of replication, one or more marker genes, a reinforcer element, a promoter, and a transcription termination sequence.
[0216] [000216] Humanized anti-CD40 antibodies can also be produced as fusion polypeptides, wherein the antibody is fused to a heterologous polypeptide, such as a signal sequence or other polypeptide having a specific cleavage site at the amino end of the protein or protein. mature polypeptide. The selected heterologous signal sequence is typically one that is recognized and processed (i.e., cleaved by a signal peptidase) by the host cell. For prokaryotic host cells that do not recognize and process the humanized anti-CD40 antibody signal sequence, the signal sequence can be replaced with a prokaryotic signal sequence. The signal sequence can be, for example, alkaline phosphatase, penicillinase, lipoprotein, heat stable enterotoxin II leaders, and the like. For yeast secretion, the native signal sequence can be replaced, for example, by a leader sequence obtained from the yeast invertase alpha factor (including the Saccharomyces and Kluyveromyces factor  leaders), acid phosphatase C, albicans glycoamylase, or by the signal described in WO90 / 13646. In mammalian cells, mammalian signal sequences, as well as viral secretory leaders, for example, the herpes simplex gD signal, can be used. The DNA for such a precursor region is linked in the reading frame to the DNA encoding the humanized anti-CD40 antibody.
[0217] [000217] The expression and cloning vectors contain a sequence of nucleic acids that enables the vector to replicate in one or more selected host cells. Generally, in cloning vectors this sequence is one that enables the vector to replicate independently of the host's chromosomal DNA, and includes the origins of replication or sequences that replicate autonomously. Such sequences are well known for a variety of bacteria, yeasts, and viruses. The origin of replication of plasmid pBR322 is suitable for most Gramnegative bacteria, the origin of plasmid 2-. it is suitable for yeast, and the various viral sources (SV40, polyoma, adenovirus, VSV, and BPV) are useful for cloning vectors in mammalian cells. Generally, the origin of replication component is not required for mammalian expression vectors (the SV40 origin can typically be used only because it contains the initial promoter).
[0218] [000218] Expression and cloning vectors may contain a gene that encodes a selectable marker to facilitate identification of the expression. Typical selectable marker genes encode proteins that confer resistance to antibiotics or other toxins, for example, ampicillin, neomycin, methotrexate, or tetracycline, or alternatively, are complement auxotrophic deficiencies, or in other alternatives, supply specific nutrients that are not present in complex media, for example, the gene encoding the D-alanine racemase for Bacilli.
[0219] [000219] An example of a selection scheme uses a drug to stop the growth of a host cell. These cells that are successfully transformed with a heterologous gene produce a protein that confers resistance to drugs and, thus, survive the selection regime. Examples of such a dominant selection use the drugs neomycin, mycophenolic acid, and hygromycin. Common selectable markers for mammalian cells are those that enable the identification of cells competent to absorb a nucleic acid encoding a humanized anti-CD40 antibody, such as DHFR (dihydrofolate reductase), thymidine kinase, metallothionein I and II (such such as primate metallothionein genes), adenosine deaminase, ornithine decarboxylase, and the like. Cells transformed with the DHFR selection gene are first identified by culturing all transformants in a culture medium containing methotrexate (Mtx), a competitive DHFR antagonist. An appropriate host cell when wild-type DHFR is employed is the Chinese hamster ovary (CHO) cell line, deficient in DHFR activity (eg, DG44).
[0220] [000220] Alternatively, host cells (particularly wild-type hosts that contain endogenous DHFR) transformed or co-transformed with DNA sequences encoding anti-CD40 antibody, wild-type DHFR protein, and another selectable marker, such as the 3'-phosphotransferase aminoglycoside (APH), can be selected by cell growth in medium containing a selectable marker selection agent, such as an aminoglycoside antibiotic, for example, kanamycin, neomycin, or G418. See, for example, Pat. No. 4,965,199.
[0221] [000221] Where recombinant production is carried out in a yeast cell as a host cell, the TRP1 gene present in the YRp7 yeast plasmid (Stinchcomb et al., 1979, Nature 282: 39) can be used as a selectable marker. The TRP1 gene provides a selection marker for a mutant strain of yeast that lacks the ability to grow on tryptophan, for example, ATCC No. 44076 or PEP4-1 (Jones, 1977, Genetics 85:12). The presence of the trp1 lesion in the yeast host cell genome then provides an effective environment for detecting transformation by growth in the absence of tryptophan. Similarly, Leu2p deficient yeast strains, such as ATCC 20,622 and 38,626, are complemented by known plasmids containing the LEU2 gene.
[0222] [000222] In addition, vectors derived from the 1.6 µm circular plasmid pKD1 can be used for the transformation of Kluyveromyces yeasts. Alternatively, an expression system for the large-scale production of recombinant calf chymosin has been described by K. lactis (Van den Berg, 1990, Bio / Technology 8: 135). Stable multiple copy expression vectors for the secretion of mature recombinant human serum albumin by industrial Kluyveromyces strains have also been disclosed (Fleer et al., 1991, Bio / Technology 9: 968-975).
[0223] [000223] Expression and cloning vectors usually contain a promoter that is recognized by the host organism and is operably linked to the nucleic acid molecule encoding an anti-CD40 antibody or its polypeptide chain. Promoters suitable for use with prokaryotic hosts include the phoA promoter, β-lactamase and lactose promoter systems, alkaline phosphatase, tryptophan (trp) promoter system and hybrid promoters, such as the tac promoter . Other known bacterial promoters are also suitable. Promoters for use in bacterial systems will also contain a Shine-Dalgamo (S.D.) sequence operably linked to the DNA encoding the humanized anti-CD40 antibody.
[0224] [000224] Many sequences of eukaryotic promoters are known. Virtually all eukaryotic genes have an AT-rich region located approximately 25 to 30 bases upstream from the site where transcription is initiated. Another sequence found 70 to 80 bases upstream from the start of transcription of many genes is a CNCAAT region, where N can be any nucleotide. At the 3 'end of most eukaryotic genes is the AATAAA sequence which may be the signal for the addition of the poly A end to the 3' end of the coding sequence. All of these sequences are properly inserted into eukaryotic expression vectors.
[0225] [000225] Examples of suitable promoter sequences, for use with yeast hosts, include promoters for 3-phosphoglycerate kinase or other glycolytic enzymes, such as enolase, glyceraldehyde-3-phosphate dehydrogenase, hexokinase, pyruvate decarboxylase, phosphofrutocinase, glucose-6-phosphate isomerase, 3-phosphoglycerate mutase, pyruvate kinase, triosphosphate isomerase, phosphoglycosis isomerase, and glucokinase.
[0226] [000226] Inducible promoters have the added advantage of transcription controlled by growing conditions. These include the regions of yeast promoters for alcohol dehydrogenase 2, isocytochrome C, phosphatase acid, derivative enzymes associated with nitrogen metabolism, metallothionein, glyceraldehyde-3-phosphate dehydrogenase, and enzymes responsible for the use of maltose and galactose. Suitable vectors and promoters, for use in yeast expression, are further described in EP 73,657. Yeast boosters are also advantageously used with yeast promoters.
[0227] [000227] Transcription of the humanized anti-CD40 antibody from vectors in mammalian host cells is controlled, for example, through promoters obtained from virus genomes, such as the polyomavirus, the contagious epithelioma virus, adenovirus ( such as Adenovirus 2), bovine papillomavirus, avian sarcoma virus, cytomegalovirus, retrovirus, hepatitis B virus and Simian Virus 40 (SV40), from heterologous mammalian promoters, for example, the promoter of actin or an immunoglobulin promoter, or from heat shock promoters, as long as such promoters are compatible with host cell systems.
[0228] [000228] The early and late promoters of the SV40 virus are conveniently obtained as an SV40 restriction fragment that also contains the SV40 viral origin of replication. The immediate human cytomegalovirus initial promoter is conveniently obtained as a restriction fragment of HindIII E. A system for expressing DNA in mammalian hosts using bovine papillomavirus as a vector is disclosed in U.S. Pat. No. 4,419,446. A modification of this system is described in Pat. No. 4,601,978. See also Reyes et al., 1982, Nature 297: 598-601, disclosing human p-interferon cDNA expression in mouse cells under the control of a thymidine kinase promoter from the herpes simplex virus. Alternatively, the long terminal repeat of the rous sarcoma virus can be used as the promoter.
[0229] [000229] Another useful element that can be used in a recombinant expression vector is a reinforcing sequence, which is used to increase the transcription of a DNA encoding an anti-CD40 antibody humanized by higher eukaryotes. Many reinforcing sequences are now known from mammalian genes (for example, globin, elastase, albumin, α-fetoprotein, and insulin). Typically, however, a reinforcing sequence from a eukaryotic cell virus is used. Examples include the SV40 booster sequence on the back side of the replication origin (bp 100-270), the booster sequence from the cytomegalovirus initial promoter, the polyoma booster sequence on the back side of the replication origin, and the booster sequences of adenovirus. See also Yaniv, 1982, Nature 297: 17-18 for a description of the reinforcing elements for the activation of eukaryotic promoters. The reinforcer sequence can be spliced into the vector at a position 5 'or 3' to the coding sequence of the humanized antiCD40 antibody, but is preferably located at a site 5 'from the promoter.
[0230] [000230] Expression vectors used in eukaryotic host cells (yeast cells, fungi, insects, plants, animals, humans, or nucleated from other multicellular organisms) may also contain sequences necessary for termination of transcription and to stabilize the mRNA. Such sequences are commonly available from the 5 'and, occasionally, 3', untranslated regions, from eukaryotic or viral DNAs or cDNAs. These regions contain nucleotide segments transcribed as polyadenylated fragments in the untranslated part of the mRNA encoding the anti-CD40 antibody. A useful transcription termination component is the bovine growth hormone polyadenylation region. See WO94 / 11026 and the expression vector disclosed therein. In some embodiments, humanized anti-CD40 antibodies can be expressed using the CHEF system. (See, for example, U.S. Pat. No. 5,888,809; the disclosure of which is incorporated by reference in this document.)
[0231] [000231] The host cells suitable for cloning or expressing the DNA in the vectors contained herein are the prokaryotic, yeast, or higher eukaryotic cells described above. Suitable prokaryotes for this purpose include eubacteria, such as Gram-negative or Gram-positive organisms, for example, Enterobacteriaceae, such as Escherichia, for example, E. coli, Enterobacter, Erwinia, Klebsiella, Proteus, Salmonella, for example , Salmonella typhimurium, Serratia, for example, Serratia marcescans, and Shigella, as well as Bacilli, such as B. subtilis and B. licheniformis (for example, B. licheniformis 41 P published in DD 266,710, published on April 12, 1989) , Pseudomonas, such as P. aeruginosa, and Streptomyces. A preferred E. coli cloning host is E. coli 294 (ATCC 31,446), although other strains, such as E. coli B, E. coli X1776 (ATCC 31,537), and E. coli W3110 (ATCC 27,325), are appropriate. These examples are illustrative, rather than limiting.
[0232] [000232] In addition to prokaryotes, eukaryotic microbes, such as filamentous fungi or yeasts, are suitable cloning or expression hosts for the humanized anti-CD40 antibody coding vectors. Saccharomyces cerevisiae, or common baker's yeast, is the most commonly used among lower eukaryotic host microorganisms. However, several other genera, species, and strains are commonly available and are useful in this document, such as Schizosaccharomyces pombe; hosts of Kluyveromyces, such as, for example, K. lactis, K. fragilis (ATCC 12.424), K. bulgaricus (ATCC 16.045), K. wickeramii (ATCC 24.178), K. waltii (ATCC 56.500), K. drosophilarum ( ATCC 36.906), K. thermotolerans, and K. marxianus; yarrowia (EP 402,226); Pichia pastors (EP 183,070); Candida; Trichoderma reesia (EP 244,234); Neurospora crassa; Schwanniomyces, such as Schwanniomyces occidentalis; and filamentous fungi, such as, for example, Neurospora, Penicillium, Tolypocladium, and hosts of Aspergillus, such as A. nidulans and A. niger.
[0233] [000233] Host cells suitable for the expression of glycosylated humanized anti-CD40 antibody are derived from multicellular organisms. Examples of invertebrate cells include plant and insect cells, including, for example, the various baculoviral strains and variants, and the corresponding facultative insect host cells of hosts such as Spodoptera frugiperda (caterpillar), Aedes aegypti (mosquito) , Aedes albopictus (mosquito), Drosophila melanogaster (fruit fly), and Bombyx mori (silkworm). A variety of viral strains for transfection are publicly available, for example, the L-1 variant of Autographa californica NPV and the Bm-5 strain of Bombyx mori NPV, and such viruses can be used, particularly for the transfection of Spodoptera cells. frugiperda.
[0234] [000234] Plant cell cultures of cotton, corn, potatoes, soybeans, petunia, tomatoes, and tobacco can also be used as hosts.
[0235] [000235] In another aspect, the expression of humanized anti-CD40 is carried out in vertebrate cells. The propagation of vertebrate cells in culture (tissue culture) has become a routine procedure and techniques are widely available. Examples of useful mammalian host cell lines are the CV40 monkey kidney lineage transformed by SV40 (COS-7, ATCC CRL 1651), the human embryonic kidney lineage (293 or 293 cells subcloned for growth in suspension culture , (Graham et al., 1977, J. Gen Virol. 36: 59), young hamster kidney cells (BHK, ATCC CCL 10), Chinese hamster ovary cells / -DHFR1 (CHO, Urlaub et al ., 1980, Proc. Natl. Acad. Sci. USA 77: 4216; for example, DG44), mouse sertoli cells (TM4, Mather, 1980, Biol. Reprod. 23: 243-251), mouse cells monkey kidneys (CV1 ATCC CCL 70), kidney cells from African green monkeys (VERO-76, ATCC CRL-1587), human cervical carcinoma cells (HELA, ATCC CCL 2), canine kidney cells (MDCK , ATCC CCL 34), rat liver cells in buffaloes (BRL 3A, ATCC CRL 1442), human lung cells (W138, ATCC CCL 75), human liver cells (Hep G2, HB 8065), mouse breast tumor (MMT 060562, ATCC CCL51), TR1 cells (Mather et al., 1982, Annals N.Y. Acad. Sci. 383: 44-68), MRC 5 cells, FS4 cells, and the human hepatoma lineage (Hep G2).
[0236] [000236] Host cells are transformed with the expression or cloning vectors described above, to produce humanized anti-CD40 antibodies, and cultured in conventional nutrient media, modified as appropriate to induce promoters, select transformants, or amplify the genes that encode the desired strings.
[0237] [000237] Host cells used to produce a humanized anti-CD40 antibody described in this document can be grown in a variety of media. Commercially available media such as Ham's F10 (Sigma-Aldrich Co., St. Louis, Mo.), Minimum Essential Medium ((MEM), (Sigma-Aldrich Co.), RPMI-1640 (Sigma-Aldrich Co. ), and Dulbecco's Modified Eagle's Medium ((DMEM), Sigma-Aldrich Co.), are suitable for culturing host cells In addition, any of the media described in one or more of Ham et al., 1979, Meth. Enz. 58: 44, Barnes et al., 1980, Anal. Biochem. 102: 255, US Pat. No. 4,767,704, US Pat. No. 4,657,866, US Pat. No. 4,927,762, Pat. US No. 4,560,655, U.S. Pat. No. 5,122,469, WO 90/103430, and WO 87/00195 can be used as culture media for host cells, and any of these media can be supplemented, as needed, with hormones and / or other growth factors (such as insulin, transferrin, or epidermal growth factor), salts (such as sodium chloride, calcium, magnesium, and phosphate), buffers (such as HEPES), nucleotides (such as adenosine and thymidine), antibiotics (such as gentamicin), microelements (defined as inorganic compounds normally present in final concentrations in the micromolar range), and glucose or an equivalent energy source. Other supplements can also be included in appropriate concentrations, which would be known to those skilled in the art. Culture conditions, such as temperature, pH, and the like, are those previously used with the host cell selected for expression, and will be apparent to the technician commonly versed.
[0238] [000238] When using recombinant techniques, the antibody can be produced intracellularly, in the periplasmic space, or directly secreted in the medium. If the antibody is produced intracellularly, cells can be disrupted to release the protein as a first step. Particulate residues, host cells or lysed fragments can be removed, for example, by centrifugation or ultrafiltration. Carter et al., 1992, Bio / Technology 10: 163-167, describe a procedure for isolating antibodies that are secreted into the E. coli periplasmic space. Briefly, the cell paste is thawed in the presence of sodium acetate (pH 3.5), EDTA, and phenylmethylsulfonyl fluoride (PMSF), for about 30 minutes. Cell residues can be removed by centrifugation. Where the antibody is secreted into the medium, supernatants from such expression systems are generally first concentrated using a commercially available protein concentration filter, for example, an Amicon or Millipore Pellicon ultrafiltration unit. A protease inhibitor, such as PMSF, can be included in any of the preceding steps to inhibit proteolysis and antibiotics can be included to prevent the growth of foreign contaminants. A variety of methods can be used to isolate the antibody from the host cell.
[0239] [000239] The antibody composition prepared from the cells can be purified using, for example, hydroxylapatite chromatography, gel electrophoresis, dialysis, and affinity chromatography, with affinity chromatography being a typical purification technique. The suitability of protein A as an affinity linker depends on the species and isotype of any immunoglobulin Fc domain that is present in the antibody. Protein A can be used to purify antibodies that are based on human gamma1, gamma2, or gamma4 heavy chains (see, for example, Lindmark et al., 1983 J. Immunol. Meth. 62: 1-13). Protein G is recommended for all mouse isotypes and for human gamma3 (see, for example, Guss et al., 1986 EMBO J. 5: 1567-1575). A matrix to which an affinity ligand is attached is most often agarose, but other matrices are available. Mechanically stable matrices, such as controlled pore glass or poly (styrenedivinyl) benzene, allow for faster flow rates and shorter processing times than can be achieved with agarose. Where the antibody comprises a CH3 domain, Bakerbond ABX® resin (J. T. Baker, Phillipsburg, N.J.) is useful for purification. Other techniques for the purification of proteins, such as fractionation on an ion exchange column, precipitation with ethanol, reverse phase HPLC, chromatography on silica, chromatography on heparin SEPHAROSE®, chromatography on an anion exchange resin or cation (such as a poly (aspartic acid) column), chromatofocalization, SDS-PAGE, and precipitation with ammonium sulfate, are also available depending on the antibody to be recovered.
[0240] [000240] After any (any) preliminary purification step (s), the mixture comprising the antibody of interest and the contaminants can be subjected to chromatography by hydrophobic interaction at low pH, using an elution buffer, at a pH between about 2.5-4.5, typically carried out at low salt concentrations (for example, about 0-0.25 M salt).
[0241] [000241] Also included are nucleic acids that hybridize under severe low, moderate, and high conditions, as defined in this document, with all or part (for example, the part encoding the variable region) of the nucleotide sequence represented by isolated polynucleotide sequence (s) encoding an antibody or antibody fragment having a heavy chain variable domain and a light chain variable region comprising the amino acid sequences of SEQ ID NO: 27 and SEQ ID NO: 26, respectively; SEQ ID NO: 28 and SEQ ID NO: 26, respectively; SEQ ID NO: 29 and SEQ ID NO: 26, respectively; SEQ ID NO: 30 and SEQ ID NO: 26, respectively; SEQ ID NO: 32 and SEQ ID NO: 31, respectively; SEQ ID NO: 33 and SEQ ID NO: 31, respectively; SEQ ID NO: 34 and SEQ ID NO: 31, respectively; SEQ ID NO: 35 and SEQ ID NO: 31, respectively; SEQ ID NO: 37 and SEQ ID NO: 36, respectively; SEQ ID NO: 38 and SEQ ID NO: 36, respectively; SEQ ID NO: 39 and SEQ ID NO: 36, respectively; SEQ ID NO: 40 and SEQ ID NO: 36, respectively. The hybridizing portion of the nucleic acid that hybridizes is typically at least 15 (e.g., 20, 25, 30 or 50) nucleotides in length. The hybridizing portion of the nucleic acid that hybridizes is at least 80%, for example, at least 90%, at least 95%, or at least 98% identical to the sequence of a part of, or all, nucleic acid encoding a polypeptide anti-CD40 (for example, a variable region of the heavy chain or light chain), or its complement. Hybridization nucleic acids of the type described in this document can be used, for example, as a cloning probe, a primer, for example, a PCR primer, or a diagnostic probe.
[0242] [000242] Some embodiments include isolated polynucleotides including sequences encoding an antibody or antibody fragment having the amino acid sequence of the variable region of the heavy chain that is at least 80%, at least 90%, at least 95%, at least 98 %, or at least 99% identical with the amino acid sequence of any of SEQ ID NO: 1 to 4, SEQ ID NO: 27, SEQ ID NO: 28, SEQ ID NO: 29, SEQ ID NO: 30, SEQ ID NO: 32, SEQ ID NO: 33, SEQ ID NO: 34, SEQ ID NO: 35, SEQ ID NO: 37, SEQ ID NO: 38, SEQ ID NO: 39, or SEQ ID NO: 40. Some modalities include isolated polynucleotides including sequences encoding an antibody or antibody fragment having the amino acid sequence of the light chain variable domain that is at least 80%, at least 90%, at least 95%, at least 98%, or at least 99 % identical to the amino acid sequence of any of SEQ ID NO: 5 to 8, SEQ ID NO: 26, SEQ ID NO: 31, or SEQ ID NO: 36.
[0243] [000243] In one aspect, the isolated polynucleotide sequence (s) encode an antibody or antibody fragment having a heavy chain variable domain and a light chain variable region, each including an amino acid sequence which is at least 80%, at least 90%, at least 95%, at least 98%, or at least 99% identical to the amino acid sequence of an antibody or antibody fragment having a heavy chain variable domain and a variable region the light chain comprising the amino acid sequences of SEQ ID NO: 27 and SEQ ID NO: 26, respectively; SEQ ID NO: 28 and SEQ ID NO: 26, respectively; SEQ ID NO: 29 and SEQ ID NO: 26, respectively; SEQ ID NO: 30 and SEQ ID NO: 26, respectively; SEQ ID NO: 32 and SEQ ID NO: 31, respectively; SEQ ID NO: 33 and SEQ ID NO: 31, respectively; SEQ ID NO: 34 and SEQ ID NO: 31, respectively; SEQ ID NO: 35 and SEQ ID NO: 31, respectively; SEQ ID NO: 37 and SEQ ID NO: 36, respectively; SEQ ID NO: 38 and SEQ ID NO: 36, respectively; SEQ ID NO: 39 and SEQ ID NO: 36, respectively SEQ ID NO: 40 and SEQ ID NO: 36, respectively.
[0244] [000244] In another aspect, the invention relates to a polynucleotide in the embodiment described immediately above, where the heavy chain variable domain and the encoded antibody light chain variable region include an amino acid sequence that is at least 95%, at least 98%, or at least 99% identical to the amino acid sequence of an antibody or antibody fragment having a heavy chain variable domain and a light chain variable region comprising the amino acid sequences of, in a modality, SEQ ID NO: 27 and SEQ ID NO: 26, respectively; in another embodiment, SEQ ID NO: 28 and SEQ ID NO: 26, respectively; in another embodiment, SEQ ID NO: 29 and SEQ ID NO: 26, respectively; in another embodiment, SEQ ID NO: 30 and SEQ ID NO: 26, respectively; in another embodiment, SEQ ID NO: 32 and SEQ ID NO: 31, respectively; in another embodiment, SEQ ID NO: 33 and SEQ ID NO: 31, respectively; in another embodiment, SEQ ID NO: 34 and SEQ ID NO: 31, respectively; in another embodiment, SEQ ID NO: 35 and SEQ ID NO: 31, respectively; in another embodiment, SEQ ID NO: 37 and SEQ ID NO: 36, respectively; in another embodiment, SEQ ID NO: 38 and SEQ ID NO: 36, respectively; in another embodiment, SEQ ID NO: 39 and SEQ ID NO: 36, respectively; and in another embodiment, SEQ ID NO: 40 and SEQ ID NO: 36, respectively.
[0245] [000245] As used herein, the terms "identical" or "percent identity", in the context of two or more nucleic acids or polypeptide sequences, refer to two or more sequences or subsequences that are the same or have a percentage specified number of nucleotides or amino acid residues that are the same when compared and aligned for maximum match. To determine the percentage of identity, the sequences are aligned for optimal comparison purposes (for example, spaces can be introduced in the sequence of a first amino acid or nucleic acid sequence for optimal alignment with a second amino acid or nucleic acid sequence). Amino acid residues or nucleotides at corresponding amino acid positions or nucleotide positions are then compared. When a position in the first sequence is occupied by the same amino acid or nucleotide residue as the corresponding position in the second sequence, then the molecules are identical at that position. The percentage of identity between the two strings is a function of the number of identical positions shared by the strings (that is,% identity = # of identical positions / # total positions (for example, overlapping positions) x 100). In some embodiments, the two strings that are compared are of the same length after spaces are introduced into the strings, as appropriate (for example, excluding the additional string that extends beyond the strands being compared). For example, when the sequences of the variable regions are compared, the leader and / or constant domain sequences are not considered. For sequence comparisons between two sequences, a "corresponding" CDR refers to a CDR in the same position in both sequences (for example, CDR-H1 from each sequence).
[0246] [000246] The determination of the percentage of identity or percentage of similarity between two sequences can be carried out using a mathematical algorithm. A preferred, non-limiting example of a mathematical algorithm used for the comparison of two sequences is the algorithm by Karlin and Altschul, 1990, Proc. Natl. Acad. Sci. USA 87: 2264-2268, modified as in Karlin and Altschul, 1993, Proc. Natl. Acad. Sci. USA 90: 5873-5877. Such an algorithm is incorporated into the NBLAST and XBLAST programs by Altschul et al., 1990, J. Mol. Biol. 215: 403-410. BLAST nucleotide searches can be performed with the NBLAST program, punctuation = 100, word length = 12, to obtain nucleotide sequences homologous to a nucleic acid encoding a protein of interest. Protein searches by BLAST can be performed with the XBLAST program, punctuation = 50, word length = 3, to obtain sequences of amino acids homologous to the protein of interest. To obtain alignments with spaces for comparison purposes, Gapped BLAST can be used, as described in Altschul et al., 1997, Nucleic Acids Res. 25: 3389-3402. Alternatively, PSI-Blast can be used to perform a repeated search that detects distant relationships between molecules (Id.). When using the BLAST, Gapped BLAST, and PSI-Blast programs, the preset parameters of the respective programs (for example, XBLAST and NBLAST) can be used. Another non-limiting, preferred example of a mathematical algorithm used for sequence comparison is the algorithm by Myers and Miller, CABIOS (1989). This algorithm is incorporated into the ALIGN program (version 2.0), which is part of the GCG sequence alignment software package. When using the ALIGN program to compare amino acid sequences, a PAM120 weight residue table, a space length penalty of 12, and a space penalty of 4 can be used. Additional algorithms for sequence analysis are known in the art and include ADVANCE and ADAM, as described in Torellis and Robotti, 1994, Comput. Appl. Biosci. 10: 3-5; and FASTA, described in Pearson and Lipman, 1988, Proc. Natl. Acad. Sci. USA 85: 2444-8. Within FASTA, ktup is a control option that adjusts the sensitivity and speed of the search. If ktup = 2, similar regions in the two sequences being compared are found considering pairs of aligned residues; if ktup = 1, individual aligned amino acids are examined. ktup can be adjusted to 2 or 1 for protein sequences, or 1 to 6 for DNA sequences. The default if ktup is not specified is 2 for proteins and 6 for DNA. Alternatively, protein sequence alignment can be performed using the CLUSTAL W algorithm, as described by Higgins et al., 1996, Methods Enzymol. 266: 383-402. Non-Therapeutic Uses
[0247] [000247] The antibodies described in this document are useful as affinity purifying agents. In this process, the antibodies are immobilized on a solid phase, such as a Protein A resin, using methods well known in the art. The immobilized antibody is contacted with a sample containing the CD40 protein (or its fragment) to be purified and, after that, the support is washed with a suitable solvent that will remove substantially all the material in the sample, except for the CD40 protein, which is bound to the immobilized antibody. Finally, the support is washed with another suitable solvent that will release the CD40 protein from the antibody.
[0248] [000248] Humanized anti-CD40 antibodies are also useful in diagnostic assays to detect and / or quantify the CD40 protein, for example, by detecting CD40 expression in specific cells, tissues or serum.
[0249] [000249] It will be advantageous in some modalities, for example, for diagnostic purposes, to label the antibody with a detectable portion. Several detectable tags are available, including radioisotopes, fluorescent tags, substrate enzyme tags and the like. The tag can be indirectly conjugated to the antibody using several known techniques. For example, the antibody can be conjugated to biotin and any of the three broad categories of brands mentioned above can be conjugated to avidin, or vice versa. Biotin selectively binds to avidin and thus the tag can be conjugated to the antibody in this indirect way. Alternatively, to obtain indirect conjugation of the tag with the antibody, the antibody can be conjugated to a small hapten (such as digoxin) and one of the different types of marks mentioned above is conjugated to an anti-hapten antibody (for example, anti-digoxin antibody). In this way, the indirect conjugation of the tag with the antibody can be obtained.
[0250] [000250] Illustrative radioisotope brands include 35S, 14C, 125I, 3H, and 131I. The antibody can be labeled with the radioisotope, using the techniques described, for example, in Current Protocols in Immunology, Volumes 1 and 2, 1991, Coligen et al., Ed. Wiley-Interscience, New York, N.Y., Pubs. Radioactivity can be measured, for example, by scintillation counting.
[0251] [000251] Illustrative fluorescent brands include brands derived from rare earth chelates (europium chelates) or fluorescein and its derivatives, rhodamine and its derivatives, dansila, Lissamine, phycoerythrin, and Texas Red are available. The fluorescent labels can be conjugated to the antibody using known techniques, such as those disclosed in Current Protocols in Immunology, supra, for example. Fluorescence can be quantified using a fluorimeter.
[0252] [000252] There are several well-characterized enzyme-substrate brands known in the art (see, for example, U.S. Pat. No. 4,275,149 for a review). The enzyme generally catalyzes a chemical change to the chromogenic substrate that can be measured using a variety of techniques. For example, the change can be a color change in a substrate that can be measured spectrophotometrically. Alternatively, the enzyme can alter the fluorescence or chemiluminescence of the substrate. Techniques for quantifying a change in fluorescence are described above. The chemiluminescent substrate becomes electronically excited by a chemical reaction and can then emit light that can be measured, using a chemiluminometer, for example, or donates energy to a fluorescent receiver.
[0253] [000253] Examples of enzyme tags include luciferases, such as firefly luciferase and bacterial luciferase (US Pat. No. 4,737,456), luciferin, 2,3-dihydrophthalazinadiones, malate dehydrogenase, urease, a peroxidase, such as horseradish peroxidase (HRPO), alkaline phosphatase, β-galactosidase, glycoamylase, lysozyme, saccharide oxidase (such as glucose oxidase, galactose oxidase, and glucose-6-phosphate dehydrogenase), heterocyclic oxidases (such as uricase and xanthine oxidase), lactoperoxidase, microperoxidase, and the like. Techniques for conjugating enzymes to antibodies are described, for example, in O'Sullivan et al., 1981, Methods for the Preparation of EnzymeAntibody Conjugates for use in Enzyme Immunoassay, in Methods in Enzym. (J. Langone and H. Van Vunakis, eds.), Academic press, N.Y., 73: 147-166.
[0254] [000254] Examples of enzyme-substrate combinations include, for example: horseradish peroxidase (HRPO) with hydrogen peroxidase as a substrate, where hydrogen peroxidase oxidizes a dye precursor, such as orthophenylene diamine (OPD) or the 3,3 ', 5,5'-tetramethyl benzidine hydrochloride (TMB); alkaline phosphatase (AP) with para-nitrophenyl phosphate as a chromogenic substrate; and β-D-galactosidase (β-D-Gal) with a chromogenic substrate, such as p-nitrophenyl-β-D-galactosidase or the fluorogenic substrate 4-methylumbelliferyl-β-D-galactosidase.
[0255] [000255] Several other enzyme-substrate combinations are available for those skilled in the art. For a general review of these, see Pat. No. 4,275,194 and U.S. Pat. No. 4,318,980.
[0256] [000256] In another embodiment, the humanized anti-CD40 antibody is used unlabeled and detected with a labeled antibody that binds the humanized anti-CD40 antibody.
[0257] [000257] The antibodies described in this document can be used in any known test method, such as competitive binding assays, direct and indirect fitting assays, and immunoprecipitation assays. See, for example, Zola, Monoclonal Antibodies: A Manual of Techniques, p. 147-158 (CRC Press, Inc. 1987). Diagnostic Kits
[0258] [000258] A humanized anti-CD40 antibody can be used in a diagnostic kit, that is, a packaged combination of reagents in predetermined quantities, with instructions for carrying out the diagnostic assay. Where the antibody is labeled with an enzyme, the kit can include substrates and cofactors required by the enzyme, such as a substrate precursor that provides the detectable chromophore or fluorophore. In addition, other additives can be included, such as stabilizers, buffers (for example, a blocking buffer or lysis buffer), and the like. The relative quantities of the various reagents can be varied widely to provide solution concentrations of the reagents that substantially optimize the sensitivity of the assay. The reagents can be provided as dry powders, usually freeze-dried, including excipients which, upon dissolution, will provide a reagent solution having the appropriate concentration. Therapeutic Uses
[0259] [000259] In another embodiment, a humanized anti-CD40 antibody disclosed in this document is useful in the treatment of various disorders associated with CD40 expression, as described in this document.
[0260] [000260] The humanized anti-CD40 antibody or agent is administered by any suitable means, including parenteral, subcutaneous, intraperitoneal, intrapulmonary, and intranasal administration, and, if desired for local, intralesional immunosuppressive treatment (including infusion or, of otherwise, contact of the graft with the antibody before transplantation). The humanized anti-CD40 antibody or agent can be administered, for example, as an infusion or as a cake. Parenteral infusions include intramuscular, intravenous, intraarterial, intraperitoneal, or subcutaneous administration. In addition, the humanized anti-CD40 antibody is suitably administered by pulse infusion, particularly with declining doses of the antibody. In one aspect, the dosage is given by injections, more preferably intravenous or subcutaneous injections, depending in part on whether the administration is short or chronic.
[0261] [000261] For the prevention or treatment of the disease, the appropriate dosage of antibody will depend on a variety of factors, such as the type of disease to be treated, as defined above, the severity and course of the disease, whether the antibody is administered for preventive or therapeutic purposes, prior therapy, the patient's medical history and antibody response, and the discretion of the responsible physician. The antibody is properly administered to the patient at one time or during a series of treatments.
[0262] [000262] Depending on the type and severity of the disease, about 1 µg / kg to 20 mg / kg (eg, 0.1-15 mg / kg) of antibody is an initial candidate dosage for administration to the patient, either either, for example, by one or more separate administrations, or by continuous infusion. A typical daily dosage could range from about 1 µg / kg to 100 mg / kg or more, depending on the factors mentioned above. For repeated administrations over several days or more, depending on the condition, treatment is continued until a desired suppression of the symptoms of the disease occurs. However, other dosage regimens may be useful. The progress of this therapy is easily monitored by conventional techniques and trials. An illustrative dosage regimen is that disclosed in WO 94/04188.
[0263] [000263] The term "suppression" is used in this document in the same context as "improvement" and "relief" to mean a decrease in one or more characteristics of the disease.
[0264] [000264] The antibody composition will be formulated, dosed, and administered in a manner consistent with good medical practice. Factors for consideration in this context include the particular disorder being treated, the particular mammal being treated, the clinical condition of the individual patient, the cause of the disorder, the location of the agent's release, the method of administration, the treatment program. administration, and other factors known to physicians. The "therapeutically effective amount" of the antibody to be administered will be governed by such considerations, and is the minimum amount necessary to prevent, ameliorate, or treat the disorder associated with CD40 expression.
[0265] [000265] The antibody does not need to be, but is optionally, formulated with one or more agents currently used to prevent or treat the disorder in question. The effective amount of such other agents depends on the amount of humanized anti-CD40 antibody present in the formulation, the type of disorder or treatment, and the other factors discussed above. These are generally used in the same dosages and with the routes of administration as used above or about 1 to 99% of the dosages used before. Disorders Associated with CD40
[0266] [000266] Anti-CD40 antibodies or agents are useful to treat or prevent cancer that expresses CD40 or an immune disorder characterized by expression of CD40, for example, by inadequate activation of immune cells (eg, lymphocytes or dendritic cells ). Such CD40 expression may be due, for example, to the increased levels of CD40 protein on the cell surface and / or to the altered antigenicity of the expressed CD40. The treatment or prevention of the immune disorder, according to the methods described in this document, is achieved by administering to a patient in need of such treatment or prevention an effective amount of the anti-CD40 antibody or agent, whereby the antibody (i) is it binds to the activated immune cells that express CD40 and that are associated with the disease state and (ii) exerts a cytotoxic, cytostatic, or immunosuppressive effect on the activated immune cells.
[0267] [000267] Immune diseases that are characterized by inadequate activation of immune cells and that can be treated or prevented by the methods described in this document can be classified, for example, by the type (s) of hypersensitivity reaction (s) that form (m) the basis of the disorder. These reactions are typically classified into four types: anaphylactic reactions, cytotoxic (cytolytic) reactions, immune complex reactions, or cell-mediated immunity (CMI) reactions (also referred to as delayed type hypersensitivity reactions (DTH)). (See, for example, Fundamental Immunology (William E. Paul ed., Raven Press, N.Y., 3rd ed. 1993).)
[0268] [000268] Specific examples of such immunological diseases include the following: rheumatoid arthritis, autoimmune demyelinating diseases (e.g., multiple sclerosis, allergic encephalomyelitis), endocrine ophthalmopathy, uveorethinitis, systemic lupus erythematosus, myasthenia gravis, severe myeloma, glomerulonephritis, glomerulonephritis, severe disease, glomerulonephritis, severe myeloma autoimmune hepatological, inflammatory bowel disease (e.g. Crohn's disease or ulcerative colitis), anaphylaxis, allergic reaction, Sjogren's syndrome, type I diabetes mellitus, primary biliary cirrhosis, Wegener's granulomatosis, fibromyalgia, polymyositis, dermatomyositis, inflammatory myositis, insufficiency multiple endocrine, Schmidt's syndrome, autoimmune uveitis, Addison's disease, adrenalitis, thyroiditis, Hashimoto's thyroiditis, autoimmune thyroid disease, pernicious anemia, gastric atrophy, chronic hepatitis, lupoid hepatitis, atherosclerosis, subacute cutaneous lupus erythematosus, hypoparathyroidism Dressler, autoimmune thrombocytopenia, purple idiopathic thrombocytopenic, hemolytic anemia, pemphigus vulgaris, pemphigus, dermatitis herpetiformis, alopecia in areas, pemphigoid, scleroderma, progressive systemic sclerosis, CREST syndrome (calcinosis, Raynaud's phenomenon, esophageal dysmotility, sclerodactia), and telangitis female, ankylosing spondylitis, ulcerative colitis, mixed connective tissue disease, polyarteritis nodosa, systemic necrotizing vasculitis, atopic dermatitis, atopic rhinitis, Goodpasture syndrome, Chagas disease, sarcoidosis, rheumatic fever, asthma, recurrent abortion, antiphospholipid syndrome, lung farmer, erythema multiforme, post-cardiotomy syndrome, Cushing's syndrome, autoimmune chronic active hepatitis, avian lung, toxic epidermal necrolysis, Alport's syndrome, alveolitis, allergic alveolitis, fibrosing alveolitis, interstitial lung disease, nodular erythema, ganoderma, ganoderma transfusion reaction, Takayasu's arteritis, polymyalgia re ummatic, temporal arteritis, schistosomiasis, giant cell arteritis, ascariasis, aspergillosis, Sampter's syndrome, eczema, lymphomatoid granulomatosis, Behcet's disease, Calan's syndrome, Kawasaki's disease, dengue, encephalomyelitis, endocarditis, high endomyocardial fibrosis, endophthalmitis, high endomyocarditis and diutine, psoriasis, fetal erythroblastosis, eosinophilic faciitis, Shulman's syndrome, Felty's syndrome, filariasis, cyclitis, chronic cyclitis, heterochronic cyclitis, Fuch's cyclitis, IgA nephropathy, Henoch-Schonlein purple, graft versus host disease, rejection transplantation, cardiomyopathy, Eaton-Lambert syndrome, recurrent polychondritis, cryoglobulinemia, Waldenstrom macroglobulemia, Evan's syndrome, adult respiratory distress syndrome, lung inflammation, osteoporosis, delayed type hypersensitivity and autoimmune gonadal insufficiency.
[0269] [000269] Consequently, the methods described in this document include the treatment of B lymphocyte disorders (e.g., systemic lupus erythematosus, Goodpasture's syndrome, rheumatoid arthritis, and type I diabetes), Th1 lymphocytes (e.g., rheumatoid arthritis, multiple sclerosis) , psoriasis, Sjorgren's syndrome, Hashimoto's thyroiditis, Grave's disease, primary biliary cirrhosis, Wegener's granulomatosis, tuberculosis, or graft versus host disease), or Th2 lymphocytes (for example, atopic dermatitis, systemic lupus erythematosus, atopic asthma, rhinoconjunctivitis, allergic rhinitis, Omenn syndrome, systemic sclerosis, or graft versus chronic host disease). Generally, disorders involving dendritic cells involve disorders of Th1 lymphocytes or Th2 lymphocytes.
[0270] 1) Inibir a diferenciação das células B e a troca de isótipo do anticorpo; 2) Inibir a produção de citocina e quimiocina e a suprarregulação de moléculas de adesão em células T e macrófagos; 3) Inibir a ativação de células dendríticas e 4) Inibir a produção de citocinas proinflamatórias, quimiocinas, metaloproteinases de matriz, prostaglandinas, e infrarregular as moléculas de adesão em células não imunes (por exemplo, células epiteliais, endoteliais e mesenquimais). [000270] Rheumatoid arthritis (RA) is one of the most common inflammatory autoimmune diseases, affecting approximately 1% of the population. Although effective treatments are available (for example, MTX and anti-TNF agents), there is a major unmet medical need, especially for those patients who do not respond adequately to anti-TNF therapies (approximately 30% of patients). In addition, up to 50% of patients discontinue treatment with TNF antagonist within 5 years, mainly due to adverse events, but also because an increasingly recognized number of patients lose their therapeutic benefit. Thus, it is important to establish effective therapies that target the inflammation and destruction of joints in RA, but do not rely only on direct inhibition of TNF. A very attractive approach is to target co-stimulating cell pathways. One of the key receptor-ligand pairs in costimulation is CD40 / CD40L. This system allows interactions between immune cells, and between immune and non-immune cells, all of which are important in the pathogenesis of RA. Blocking CD40 with an antagonistic antibody of the present invention can have one or more of the following effects on RA: 1) Inhibit B cell differentiation and antibody isotype exchange; 2) Inhibit the production of cytokine and chemokine and the uptake of adhesion molecules in T cells and macrophages; 3) Inhibit the activation of dendritic cells and 4) Inhibit the production of proinflammatory cytokines, chemokines, matrix metalloproteinases, prostaglandins, and infraregulate adhesion molecules in non-immune cells (for example, epithelial, endothelial and mesenchymal cells).
[0271] [000271] The methods of achieving one or more of the effects mentioned above are expressly contemplated in this document. In addition to RA, the compositions of the present invention will be particularly useful in the methods of treating Multiple Sclerosis, Psoriasis (including Psoriatic Arthritis), Juvenile Rheumatoid Arthritis, Inflammatory Bowel Disease, Systemic Lupus Erythematosus, and Solid Organ Transplantation.
[0272] [000272] Rheumatoid Arthritis (RA) is a chronic, systemic autoimmune disease, with a prevalence of approximately 1% in adults. The disease continues to cause significant morbidity and premature mortality (mortality is predominantly due to accelerated cardiovascular disease). It has now been identified that joint damage occurs very early in the course of the disease, with up to 30% of patients showing radiographic evidence of bone wear at the time of diagnosis, increasing to 60% after 1 year. Current guidelines recommend starting therapy with traditional disease-modifying antirheumatic drugs (DMARDs) within 3 months after a definitive diagnosis has been established. DMARDS have the potential to reduce or prevent joint damage and retain joint function. Currently, rheumatologists select methotrexate (MTX) as the initial DMARD therapy for most patients.
[0273] [000273] TNF antagonists etanercept (Enbrel®), infliximab (Remicade®), adalimumab (Humira®), CTLA4 abatacept antagonist (Orencia®), IL-6 tocilizumab antireceptor mAb and rituximabb anti-CD20 mAb (Rituxan®) are effective in the treatment of RA. Current guidelines generally recommend using biological DMARDs for the treatment of active RA after an inadequate response to traditional DMARDs.
[0274] [000274] Recent studies in patients with initial aggressive RA, without prior treatment with MTX, showed that the combination of MTX with a TNF antagonist was superior to each when used as monotherapy. The most extraordinary result was the significant radiological benefit of combination therapy. Therefore, the combination of MTX and TNF inhibitors should be used in patients at increased risk for aggressive disease and aggressive phenotype (for example, high activity score, functional deterioration, seropositivity for rheumatoid factor (RF) or cyclic citrullinated anti-peptide antibody (CCP), elevated CRP, radiographic wear). However, we anticipate that, in clinical practice, it will be rare for TNF antagonists to be used as a first-line therapy. A survey by US rheumatologists conducted in April 2005 showed that the factors that most influenced the decision to use a TNF antagonist were: failure of MTX or multiple DMARDs, overall physician assessment, functional deterioration, and worsening or radiographic wear. Currently, an estimated 20% of RA patients receive TNF inhibitor therapy in the US.
[0275] [000275] A substantial percentage of patients with RA are not adequately assisted with current treatments, including biological therapies, because of intolerance and toxicity of the drug or lack of response. Up to 50% of patients discontinue treatment with a TNF antagonist within 5 years, mainly due to adverse events, but also because an increasingly recognized number of patients lose their response.
[0276] [000276] In some embodiments, the immune disorder is an immune disorder mediated by T cells, such as a T cell disorder, in which the activated T cells, associated with the disorder, express CD40. Anti-CD40 antibodies or agents can be administered to reduce such activated T cells that express CD40. In a specific embodiment, administration of anti-CD40 antibodies or agent can reduce activated T cells that express CD40, while the remaining T cells are not substantially reduced by anti-CD40 or the agent. In this context, "are not substantially reduced" means that less than about 60%, or less than about 70% or less than about 80% of the remaining T cells are not reduced.
[0277] [000277] Anti-CD40 antibodies and agents as described in this document are also useful for treating or preventing cancer that expresses CD40. The treatment or prevention of a cancer that expresses CD40, according to the methods described in this document, is achieved by administration to a patient who needs such treatment or prevention of an effective amount of the anti-CD40 antibody or agent, by that the antibody (i) binds to cancer cells that express CD40 and (ii) exerts a cytotoxic or cytostatic effect to reduce or inhibit the proliferation of cancer cells that express CD40.
[0278] [000278] CD40-expressing cancers that can be treated or prevented by the methods described in this document include, for example, leukemia, such as acute leukemia, acute lymphocytic leukemia, acute myelocytic leukemia (e.g. myeloblastic, promyelocytic , myelomonocytic, monocytic, or erythroleukemia), chronic leukemia, chronic myelocytic (granulocytic) leukemia, or chronic lymphocytic leukemia; red polycythemia; o Lymphoma (for example, Hodgkin's disease or Non-Hodgkin's disease); multiple myeloma, Waldenstrom's macroglobulinemia; heavy chain disease; solid tumors, such as sarcomas and carcinomas (e.g., fibrosarcoma, myxosarcoma, liposarcoma, chondrosarcoma, osteogenic sarcoma, osteosarcoma, chordoma, angiosarcoma, endotheliosarcoma, lymphangiosarcoma, colon carcinoma, colorectal carcinoma, pancreatic cancer, breast cancer, ovarian cancer, prostate cancer, squamous cell carcinoma, basal cell carcinoma, adenocarcinoma, sweat gland carcinoma, sebaceous gland carcinoma, papillary carcinoma, papillary carcinoma, papillary carcinoma , medullary carcinoma, bronchogenic carcinoma, renal cell carcinoma, hepatoma, bile duct carcinoma, choriocarcinoma, seminoma, embryonic carcinoma, Wilms tumor, cervical cancer, uterine cancer, testicular tumor, lung carcinoma, small cell lung carcinoma, non-small cell lung carcinoma, carcinoma bladder, epithelial carcinoma, glioma, astrocytoma, medulloblastoma, craniopharyngioma, ependymoma, pinealoma, hemangioblastoma, acoustic neuroma, oligodendroglioma, menangioma, melanoma, neuroblastoma, retinoblastoma, carcinoma, nasopharyngeal carcinoma, nasopharyngeal carcinoma, nasopharyngeal carcinoma. Pharmaceutical Compositions and Their Administration
[0279] [000279] A composition comprising a CD40 binding agent (for example, an anti-CD40 antibody) can be administered to a patient having, or at risk of, having an immune disorder or a cancer that expresses CD40. The invention further provides for the use of a CD40 binding agent (for example, an anti-Cd40 antibody) in the manufacture of a medicament for the prevention or treatment of a cancer that expresses CD40 or an immune disorder. The term "patient", as used herein, means any mammalian patient to whom the CD40 binding agent may be administered, including, for example, humans and non-human mammals, such as primates, rodents, and the dogs. Patients specifically intended for treatment using the methods described in this document include humans. The antibodies or agents can be administered alone or in combination with other compositions to prevent or treat the immune disorder or cancer that expresses CD40.
[0280] [000280] Preferred antibodies for use in such pharmaceutical compositions are those comprising the humanized antibody or the antibody fragment having the amino acid sequence of the variable region of any heavy chain of SEQ ID NO: 1 to 4, SEQ ID NO: 27, SEQ ID NO: 28, SEQ ID NO: 29, SEQ ID NO: 30, SEQ ID NO: 32, SEQ ID NO: 33, SEQ ID NO: 34, SEQ ID NO: 35, SEQ ID NO: 37, SEQ ID NO: 38, SEQ ID NO: 39, or SEQ ID NO: 40.
[0281] [000281] Some embodiments include isolated polynucleotides comprising the sequences encoding an antibody or antibody fragment having the amino acid sequence of the light chain variable domain of SEQ ID NO: 26, SEQ ID NO: 31, or SEQ ID NO: 36 . Particularly preferred humanized antibody compositions comprise an antibody or antibody fragment having a heavy chain variable domain and a light chain variable region comprising the amino acid sequences of SEQ ID NO: 27 and SEQ ID NO: 26, respectively; SEQ ID NO: 28 and SEQ ID NO: 26, respectively; SEQ ID NO: 29 and SEQ ID NO: 26, respectively; SEQ ID NO: 30 and SEQ ID NO: 26, respectively; SEQ ID NO: 32 and SEQ ID NO: 31, respectively; SEQ ID NO: 33 and SEQ ID NO: 31, respectively; SEQ ID NO: 34 and SEQ ID NO: 31, respectively; SEQ ID NO: 35 and SEQ ID NO: 31, respectively; SEQ ID NO: 37 and SEQ ID NO: 36, respectively; SEQ ID NO: 38 and SEQ ID NO: 36, respectively; SEQ ID NO: 39 and SEQ ID NO: 36, respectively; SEQ ID NO: 40 and SEQ ID NO: 36, respectively. Included within the present invention are isolated polynucleotides that encode any of the heavy chain sequences of SEQ ID NO: 1 to 4, SEQ ID NO: 27, SEQ ID NO: 28, SEQ ID NO: 29, SEQ ID NO: 30, SEQ ID NO: 32, SEQ ID NO: 33, SEQ ID NO: 34, SEQ ID NO: 35, SEQ ID NO: 37, SEQ ID NO: 38, SEQ ID NO: 39, SEQ ID NO: 40, SEQ ID NO: 42, SEQ ID NO: 44, SEQ ID NO: 46, SEQ ID NO: 48, SEQ ID NO: 53, SEQ ID NO: 57, SEQ ID NO: 58, SEQ ID NO: 59, SEQ ID NO: 60, SEQ ID NO: 61, SEQ ID NO: 62, SEQ ID NO: 63, SEQ ID NO: 64, SEQ ID NO: 65, SEQ ID NO: 66, SEQ ID NO: 67, SEQ ID NO: 68, SEQ ID NO: 69, SEQ ID NO: 70, SEQ ID NO: 71, SEQ ID NO: 72, or SEQ ID NO: 73. The other modalities are for isolated nucleic acids that encode a light chain sequence for any of the sequences of SEQ ID NO: 5 to SEQ ID NO: 8, SEQ ID NO: 26, SEQ ID NO: 31, SEQ ID NO: 36, SEQ ID NO: 41, SEQ ID NO: 43, SEQ ID NO: 45, SEQ ID NO: 47, SEQ ID NO: 49, SEQ ID NO: 50, SEQ ID NO: 51, SEQ ID NO: 52, SEQ ID NO: 54, SEQ ID NO: 55, SEQ ID NO: 56, SEQ ID NO: 74, SEQ ID NO: 75, or SEQ ID NO: 76.
[0282] [000282] In certain embodiments, where RA treatment is contemplated, the compositions of the invention can be used in methods to reduce signs and symptoms, induce a major clinical response and reduce the progression of structural damage in patients with moderate or severe RA. severely active that do not respond properly to MTX alone. Such current illustrative therapy is: Enbrel / Humira (Data with Humira and Enbrel were obtained from two different patient populations). The compositions of the present invention can be used instead of Enbrel / Humira therapy or in combination with Enbrel / Humira therapy for patients who do not respond to MTX alone. Preferably, in such embodiments, the compositions of the invention will be more effective than Enbrel + MTX in patients who have had an inadequate response to methotrexate, as determined, for example, by: ACR20 in 6 months> 85% for the compound plus MTX (GS: Enbrel + MTX 71% vs. Placebo + MTX 27%, Humira + MTX in 12 months 59% vs. Placebo + MTX 24%) *. Additional criteria for the superior effectiveness of the compositions of the invention may include: Inhibition of the progression of structural damage over a period of one year similar to Enbrel (after 52 weeks, mean modified Sharp score Humira + MTX 0.1 vs. Placebo + MTX 2.7) *. In yet other modalities, the compositions produce a "Main Clinical Response" superior to Enbrel in patients who have had an inadequate response to methotrexate, as measured by ACR70 (20% for Humira + MTX, 4% for Placebo + MTX) *.
[0283] [000283] In other embodiments, the compositions of the invention can be indicated to reduce signs and symptoms, induce a major clinical response and reduce the progression of structural damage in patients with moderate to severely active RA who have had an inadequate response to agents anti-TNF. The current Gold standard: biological therapy other than anti-TNF. Preferably, in such patients, the compositions of the invention have no inferior effectiveness, compared to the biological one that is not anti-TNF (for example, Orencia, Rituxan), by historical comparison in patients who have had an inadequate response to an anti-TNF agent. : ACR20 in 6 months> 50% for the compound plus DMARD (GS: Orencia + DMARD 50% vs. placebo + DMARD 20%). In still other modalities, the compositions of the invention inhibit the progression of structural damage over a period of one year evaluated, by accepted X-ray scoring methods for joint wear and narrowing of the joint space, similar to Rituxan (after 52 weeks, mean modified Sharp score Rituxan + MTX 1.0 vs. Placebo + MTX 2.31).
[0284] [000284] Several delivery systems are known and can be used to deliver the CD40 binding agent. Methods of introduction include, but are not limited to, intradermal, intramuscular, intraperitoneal, intravenous, subcutaneous, intranasal, epidural, and oral routes. The CD40 binding agent can be administered, for example, via infusion, bolus or injection, and can be administered together with other biologically active agents, such as chemotherapeutic agents. Administration can be systemic or local. In preferred embodiments, administration is by subcutaneous injection. Formulations for such injections can be prepared, for example, in pre-filled syringes that can be administered once, every other week.
[0285] [000285] The safety features of the antibodies of the invention will be determined and preferably include one or more features such as: no clinically significant adverse interactions with the other medications commonly used to treat Rheumatoid Arthritis (e.g., DMARDs, Steroids, NSAIDs); No higher discontinuation fees due to security or tolerability issues compared to Enbrel; Rate of serious infections no higher than anti-TNF agents or other commonly used biological agents; Frequency and / or severity of reactions at the injection site or infusion reaction similar to Enbrel; No development or minimal development of drug resistance (less than 5%) in repeated therapy cycles; No neutralizing antibodies or minimal neutralizing antibodies; No evidence of increased platelet aggregation / activation that could result in thromboembolic events in vivo or platelet / endothelial dysfunction that could result in bleeding.
[0286] [000286] In the specific modalities, the composition of CD40 binding agent is administered by injection, through a catheter, through a suppository, or through an implant, the implant being of a porous, non-porous material, or gelatinous, including a membrane, such as a sialastic membrane, or a fiber. Typically, when administering the composition, materials are used to which the anti-CD40 antibody or agent does not absorb.
[0287] [000287] In other embodiments, the anti-CD40 antibody or agent is released in a controlled release system. In one embodiment, a pump can be used (see, for example, Langer, 1990, Science 249: 1527-1533; Sefton, 1989, CRC Crit. Ref. Biomed. Eng. 14: 201; Buchwald et al., 1980, Surgery 88: 507; Saudek et al., 1989, N. Engl. J. Med. 321: 574). In another embodiment, polymeric materials can be used. (See, for example, Medical Applications of Controlled Release (Langer and Wise eds., CRC Press, Boca Raton, Fla., 1974); Controlled Drug Bioavailability, Drug Product Design and Performance (Smolen and Ball eds., Wiley, New York , 1984); Ranger and Peppas, 1983, Macromol. Sci. Rev. Macromol. Chem. 23:61. See also Levy et al., 1985, Science 228: 190; During et al., 1989, Ann. Neurol. 25 : 351; Howard et al., 1989, J. Neurosurg. 71: 105.) Other controlled-release systems are discussed, for example, in Langer, supra.
[0288] [000288] A CD40 binding agent (for example, an anti-CD40 antibody) can be administered as pharmaceutical compositions comprising a therapeutically effective amount of the binding agent and one or more pharmaceutically compatible ingredients.
[0289] [000289] In typical embodiments, the pharmaceutical composition is formulated, according to routine procedures, as a pharmaceutical composition adapted for intravenous or subcutaneous administration to humans. Typically, compositions for administration by injection are solutions in sterile isotonic aqueous buffer. Where necessary, the pharmaceutical substance can also include a solubilizing agent and a local anesthetic, such as lignocaine, to soothe pain at the injection site. Generally, the ingredients are provided separately or mixed together in unit dosage form, for example, as a dry or concentrated lyophilized powder without water, in an airtight container, such as an ampoule or sachet indicating the amount of active agent. Where the pharmaceutical substance is to be administered by infusion, it can be administered with an infusion bottle containing sterile, pharmaceutical grade water or saline. Where the pharmaceutical substance is administered by injection, an ampoule of sterile water for injection or saline can be provided, so that the ingredients can be mixed before administration.
[0290] [000290] Furthermore, the pharmaceutical composition can be provided as a pharmaceutical kit comprising (a) a container containing a CD40-binding agent (for example, an anti-CD40 antibody) in lyophilized form and (b) a second container containing a pharmaceutically acceptable diluent (eg, sterile water) for injection. The pharmaceutically acceptable diluent can be used for reconstitution or dilution of the anti-CD40 antibody or lyophilized agent. Optionally associated with such container (s) is a notice in the form prescribed by a government agency that regulates the manufacture, use or sale of pharmaceutical substances or biological products, a notice that reflects the approval by the manufacturing agency, of use or sale to human administration.
[0291] [000291] The amount of the CD40 binding agent (for example, the anti-CD40 antibody) that is effective in treating or preventing an immune disorder or a cancer that expresses CD40 can be determined by standard clinical techniques. In addition, in vitro assays can optionally be employed to help identify optimal dosage ranges. The exact dose to be used in the formulation will also depend on the route of administration, and the stage of the immune disorder or cancer that expresses CD40, and should be decided according to the judgment of the doctor and the circumstances of each patient. Effective doses can be extrapolated from dose response curves derived from in vitro test systems or in animal models.
[0292] [000292] For example, the toxicity and therapeutic efficacy of the anti-CD40 antibody or agent can be determined in cell cultures or experimental animals, by standard pharmaceutical procedures, to determine ED50 (the therapeutically effective dose in 50% of the population ). A CD40-binding agent (for example, an anti-CD40 antibody) which exhibits a high therapeutic index is preferred. Where the CD40-binding agent exhibits toxic side effects, a delivery system that targets the CD40-binding agent to the site of the affected tissue can be used to minimize potential damage to cells that do not express CD40 and thereby , reduce side effects.
[0293] [000293] The data obtained from cell culture assays and animal studies can be used in the formulation of a dosage range for use in humans. The dosage of the CD40 binding agent typically lies in a range of circulating concentrations that include the ED50 with little or no toxicity. The dosage can vary within this range, depending on the dosage form used and the route of administration used. For any CD40 binding agent used in the method, the therapeutically effective dose can be estimated initially from cell culture assays. A dose can be formulated in animal models to achieve a range of circulating plasma concentrations that includes the IC 50 (i.e., the concentration of the test compound that achieves a semi-maximum inhibition of symptoms), as determined in cell culture. Such information can be used to more accurately determine useful doses in humans. Plasma levels can be measured, for example, by high performance liquid chromatography, ELISA and the like.
[0294] [000294] Generally, the dosage of an anti-CD40 antibody or CD40 binding agent administered to a patient with an immune disorder or cancer that expresses CD40 is typically about 0.1 mg / kg to about 100 mg / kg weight of the patient's body. The dosage administered to a patient is about 0.1 mg / kg to about 50 mg / kg, about 1 mg / kg to about 30 mg / kg, about 1 mg / kg to about 20 mg / kg , about 1 mg / kg to about 15 mg / kg, or about 1 mg / kg to about 10 mg / kg of the patient's body weight.
[0295] [000295] Illustrative doses include, but are not limited to, from about 0.1 mg / kg to about 100 mg / kg of the patient's body weight. The dosage administered to a patient is about 0.1 mg / kg to about 50 mg / kg, about 1 mg / kg to about 30 mg / kg, about 1 mg / kg to about 20 mg / kg , about 1 mg / kg to about 15 mg / kg, or about 1 mg / kg to about 10 mg / kg of the patient's body weight. The dose can be administered, for example, daily, once a week (weekly), twice a week, three times a week, four times a week, five times a week, six times a week, biweekly or monthly, each two months, or every three months. In the specific modalities, the dose is about 0.5 mg / kg / week, about 1 mg / kg / week, about 2 mg / kg / week, about 3 mg / kg / week, about 4 mg / week. kg / week, about 5 mg / kg / week, about 6 mg / kg / week, about 7 mg / kg / week, about 8 mg / kg / week, about 9 mg / kg / week, about 10 mg / kg / week, about 11 mg / kg / week, about 12 mg / kg / week, about 13 mg / kg / week, about 14 mg / kg / week, about 15 mg / kg / week or about 16 mg / kg / week. In some embodiments, the dose ranges from about 1 mg / kg / week to about 15 mg / kg / week.
[0296] [000296] In some embodiments, pharmaceutical compositions comprising the CD40 binding agent may additionally comprise a therapeutic agent, conjugated or non-conjugated to the binding agent. The anti-CD40 antibody or CD40 binding agent can be co-administered in combination with one or more therapeutic agents, for the treatment or prevention of immune disorders or cancers that express CD40. For example, combination therapy can include a cytostatic, cytotoxic, or immunosuppressive agent. Combination therapy may also include, for example, administration of an agent that targets a receptor or receptor complex other than CD40 on the surface of activated lymphocytes, dendritic cells or cancer cells that express CD40. An example of such an agent includes a second, non-CD40 antibody that binds to a molecule on the surface of an activated lymphocyte, dendritic cell or cancer cell that expresses CD40. Another example includes a linker that targets such a receptor or receptor complex. Typically, such an antibody or ligand binds to a cell surface receptor on activated lymphocytes, dendritic cells or the cancer cell that expresses CD40 and increases the cytotoxic or cytostatic effect of the anti-CD40 antibody by releasing a cytostatic signal or cytotoxic to the activated lymphocyte, the dendritic cell or the cancer cell that expresses CD40.
[0297] [000297] Such administration of combination therapy may have an additive or synergistic effect on the parameters of the disease (for example, the severity of a symptom, the number of symptoms, or the frequency of relapse).
[0298] [000298] Regarding therapeutic regimens for administration by combination, in a specific modality, an anti-CD40 antibody or CD40-binding agent is administered simultaneously with a therapeutic agent. In another specific embodiment, the therapeutic agent is administered prior to, or subsequent to, administration of the anti-CD40 antibody or CD40 binding agent, for at least one hour and up to several months, for example, at least one hour, five hours, 12 hours, a day, a week, a month, or three months before, or subsequent to, administration of the anti-CD40 antibody or CD40-binding agent.
[0299] [000299] Useful classes of cytotoxic or immunosuppressive agents include, for example, antitubulin agents, auristatins (for example, MMAE, or MMAF), secondary DNA groove ligands, inhibitors of replication of DNA, alkylating agents (for example, platinum complexes, such as cis-platinum, mono (platinum), bis (platinum) and trinuclear platinum complexes and carboplatin), anthracyclines, antibiotics, antifolates, antimetabolites, chemotherapy sensitizers, duocarmicins, etoposides, fluorinated pyrimidines, ionophores, lexitropsins, nitrosoureas, platinum, preforming compounds, purine antimetabolites, puromycins, radiation sensitizers , steroids, taxanes, topoisomerase inhibitors, vinca alkaloids, or the like.
[0300] [000300] Individual cytotoxic or immunosuppressive agents include, for example, an androgen, anthramycin (AMC), asparaginase, 5-azacytidine, azathioprine, bleomycin, busulfan, butionin sulfoximine, camptothecin, carboplatin, carmustine (BSNU), CD-1065, chlorambucil, cisplatin, colchicine, cyclophosphamide, cytarabine, cytidine arabinoside, cytochalasin B, dacarbazine, dactinomycin (formerly actinomycin), daunorubin, decarbazine docetaxel, doxorubicin, an estrogen, 5-fluordesoxyuridine, 5-fluorouracil, gramicidin D, hydroxyurea, idarubicin, ifosfamide, irinotecan, lomustine (CCNU), meclorethamine, melphalan, 6- mercaptopurine, methotrexate, mitramicin, mitomycin C, mitoxantrone, nitroimidazole, paclitaxel, plicamycin, procarbizine, streptozotocin, tenoposide, 6-thioguanine, thioTEPA, topotechin, vimine, vinoxastine, vim vinorelbine, VP-16 and VM-26.
[0301] [000301] In some typical embodiments, the therapeutic agent is a cytotoxic agent. Suitable cytotoxic agents include, for example, dolastatin (for example, auristatin E, AFP, MMAF, MMAE, AEB or AVB), secondary DNA groove ligands (for example, enediins and lexitropsins), duocarmicins, taxanes (eg paclitaxel and docetaxel), puromycins, vinca alkaloids, CC-1065, SN-38, topotecan, morpholino-doxorubicin, rhizoxin, cyanomorpholino-doxorubicin, echinomycin, combrestastatin, netropsin, epothilone A and B, estramustine, cryptophysins, cemadotine, maytansinoids, discodermolide, eleuterobin, or mitoxantrone.
[0302] [000302] In some embodiments, the cytotoxic agent is a conventional chemotherapy, such as, for example, doxorubicin, paclitaxel, melphalan, vinca alkaloids, methotrexate, mitomycin C or etoposide. In addition, potent agents, such as CC-1065 analogs, calicheamicin, maytansine, dolastatin 10 analogs, rhizoxin, and palitoxin, can be linked to anti-CD40 antibodies or agents thereof.
[0303] [000303] In specific embodiments, the cytotoxic or cytostatic agent is auristatin E (also known in the art as dolastatin-10) or a derivative thereof. Typically, the auristatin E derivative is, for example, an ester formed between auristatin E and an acid keto. For example, auristatin E can be reacted with para-acetyl benzoic acid or benzoylvaleric acid, to produce AEB and AEVB, respectively. The other typical auristatin derivatives include AFP, MMAF, and MMAE. The synthesis and structure of auristatin E and its derivatives are described, for example, in U.S. Patent Application Publications Nos. 2004-0157782 A1 and 2005-0238649; in International Patent Application No. PCT / US03 / 24209, in International Patent Application No. PCT / US02 / 13435, and in Pats. U.S. Nos. 6,884,869; 6,323,315; 6,239,104; 6,034,065; 5,780,588; 5,665,860; 5,663,149; 5,635,483; 5,599,902; 5,554,725; 5,530,097; 5,521,284; 5,504,191; 5,410,024; 5,138,036; 5,076,973; 4,986,988; 4,978,744; 4,879,278; 4,816,444; and 4,486,414; whose disclosures are incorporated by reference in this document.
[0304] [000304] In specific embodiments, the cytotoxic agent is a secondary DNA groove binding agent. (See, for example, U.S. Pat. No. 6,130,237.) For example, in some embodiments, the secondary groove binding agent is a CBI compound. In other embodiments, the secondary groove binding agent is an enediin (for example, calicheamicin).
[0305] [000305] Examples of antitubulin agents include, but are not limited to, taxanes (e.g., Taxol® (paclitaxel), Taxotere® (docetaxel)), T67 (Tularik), vinca alkaloids (e.g. vincristine, vinblastine, vindesine, and vinorelbine), and dolastatin (e.g., auristatin E, AFP, MMAF, MMAE, AEB, AEVB). Other anti-tubulin agents include, for example, baccatin derivatives, taxane analogs (eg epothilone A and B), nocodazole, colchicine and colcimid, estramustine, cryptophysins, cemadotine, maytansinoids, combretastatines, discodermolide, and eleuterobin.
[0306] [000306] In some embodiments, the cytotoxic agent is a maytansinoid, another group of antitubulin agents. For example, in specific embodiments, maytansinoid is maytansine or DM-1 (ImmunoGen, Inc .; see also Chari et al., 1992, Cancer Res. 52: 127-131).
[0307] [000307] In some embodiments, the therapeutic agent is not a radioisotope.
[0308] [000308] In some embodiments, the cytotoxic or immunosuppressive agent is an antimetabolite. The antimetabolite can be, for example, a purine antagonist (for example, azothioprine or mycophenolate mofetil), a dihydrofolate reductase inhibitor (for example, methotrexate), acyclovir, gangciclovir, zidovudine, vidarabine, ribavarin, azidotimidine, azidotimidine, amantidine , didesoxyuridine, iododeoxyuridine, poscamet, or trifluridine.
[0309] [000309] In other embodiments, the cytotoxic or immunosuppressive agent is tacrolimus, cyclosporine or rapamycin. In the additional modalities, the cytotoxic agent is aldesleukin, alentuzumab, alitretinoin, allopurinol, altretamine, amifostine, anastrozole, arsenic trioxide, bexarotene, bexarotene, calusterone, capecitabine, celecoxib, cladribine, dyroboxyrine, propane, dithromethaxine, dyromethaxine, dithromethaxine, dithromethaxine, drafting , Epoetin alfa, estramustine, exemestane, Filgrastim, floxuridine, fludarabine, fulvestrant, gemcitabine, gentuzumab ozogamycin, goserelin, idarrubicin, ifosfamide, imatinib mesylate, interferon alfa-2a, irinotecan, megaminoline, nitrogen, mecanine, estrogen , mesna, methotrexate, methoxsalene, mitomycin C, mitotane, nandrolone fempropionate, oprelvecine, oxaliplatin, pamidronate, pegademase, pegaspargase, pegfilgrastim, pentostatin, pipobroman, plicamycin, estrogen, quinidine, saroxine, procarbazine, procarbazine , temozolomide, teniposide, testolactone, thioguan ina, toremifene, Tositumomab, Trastuzumab, tretinoin, uracil mustard, valrubicin, vinblastine, vincristine, vinorelbine and zoledronate.
[0310] [000310] In the additional embodiments, the drug is a humanized anti-HER2 monoclonal antibody; RITUXAN (rituximab; Genentech, Inc., South San Francisco, Calif.); a chimeric anti-CD20 monoclonal antibody); OVAREX (AltaRex Corporation, MA); PANOREX (Glaxo Wellcome, NC; a mouse IgG2a antibody); Cetuximab Erbitux (Imclone Systems Inc., NY; a chimeric anti-EGFR IgG antibody); Vitaxin (MedImmune, Inc., MD); Campath I / H (Leukosite, MA; a humanized IgG1 antibody); Smart MI95 (Protein Design Labs, Inc., CA; a humanized anti-CD33 IgG antibody); LymphoCide (Immunomedics, Inc., NJ; a humanized anti-CD22 IgG antibody); Smart ID10 (Protein Design Labs, Inc., CA; a humanized anti-HLA-DR antibody); Oncolym (Techniclone, Inc., CA; a radiolabeled rat anti-HLA-Dr10 antibody); Allomune (BioTransplant, CA; a humanized anti-CD2 mAb); Avastin (Genentech, Inc., CA; a humanized anti-VEGF antibody); Epratuzamab (Immunomedics, Inc., NJ and Amgen, CA; an anti-CD22 antibody); and CEAcide (Immunomedics, NJ; a humanized anti-CEA antibody).
[0311] [000311] Other suitable antibodies include, but are not limited to, antibodies against the following antigens: CA125, CA15-3, CA19-9, L6, Lewis Y, Lewis X, alpha fetoprotein, CA 242, placental alkaline phosphatase, specific antigen prostate cancer, prostatic acid phosphatase, epidermal growth factor, MAGE-1, MAGE-2, MAGE-3, MAGE-4, anti-transferrin receptor, p97, MUC1-KLH, CEA, gp100, MART1, Prostate Specific Antigen, receptor of IL-2, CD20, CD52, CD33, CD22, human chorionic gonadotropin, CD38, mucin, P21, MPG, and Neu oncogene product.
[0312] [000312] In some embodiments, the therapeutic agent is an immunosuppressive agent. The immunosuppressive agent can be, for example, ganciclovir, etanercept, tacrolimus, cyclosporine, rapamycin, cyclophosphamide, azathioprine, mycophenolate mofetil or methotrexate. Alternatively, the immunosuppressive agent can be, for example, a glucocorticoid (for example, cortisol or aldosterone) or a glucocorticoid analogue (for example, prednisone or dexamethasone).
[0313] [000313] Suitable cycloxygenase inhibitors include meclofenamic acid, mefenamic acid, carprofen, diclofenac, diflunisal, fembufen, fenoprofen, ibuprofen, indomethacin, ketoprofen, nabumetone, naproxen, sulindac, sulindac enoxicam, tolmetin, and acetylsalicylic acid.
[0314] [000314] Suitable lipoxygenase inhibitors include redox inhibitors (eg, catechol butane derivatives, nordihydroguaiaretic acid (NDGA), masoprocol, phenidone, Ianopalene, indazolinones, nafazatrom, benzofuranol, alkylhydroxylamine ), and non-redox inhibitors (for example, hydroxythiazoles, methoxyalkylthiazoles, benzopyrans and their derivatives, methoxytetrahydropyran, bosonic acids and acetylated derivatives of bosonic acids, and quinolinamethoxyphenylacetic acids substituted with cycloalkyl radicals), and the precursors of inhibitors of inhibitors
[0315] [000315] Other suitable lipoxygenase inhibitors include antioxidants (eg, phenols, propyl gallate, flavonoids and / or naturally occurring substrates containing flavonoids, hydroxylated derivatives of flavones, flavonol, dihydroquercetin, luteolin, galangin, orobol, derivatives of calcona, 4,2 ', 4'-triidroxicalcona, orthoaminophenols, N-hydroxyureas, benzofuranols, ebselene and species that increase the activity of reducing selenzymes), iron chelating agents (for example, hydroxamic acids and their derivatives, N-hydroxyureas , 2-benzyl-1-naphthol, catechols, hydroxylamines, carnosol trolox C, catechol, naphthol, sulfasalazine, zyleuton, 5-hydroxyanthranilic acid and 4- (omega-arylalkyl) phenylalkanoic acids), the compounds containing imidazole (for example, ketoconazole and itraconazole), phenothiazines, and benzopyran derivatives.
[0316] [000316] Still other suitable lipoxygenase inhibitors include eicosanoid inhibitors (for example, octadecatetraenoic, eicosatetraenoic, docosapentaenoic, eicosaexaenoic and docosahexenic acids and their esters, PGE1 (prostaglandin E1), PGA2 (prostaglandin), prostaglandin A2 (prostaglandin) , 15-monohydroxyeicosatetraenoic acids, 15-monohydroxy-eicosatrienoic and 15-monohydroxyeicosapentaenoic acids, and leukotrienes B5, C5 and D5), compounds that interfere with calcium flows, phenothiazines, diphenylbutylamines, verapamil, fusco, curcumin, chlorogenic acid, caffeic acid, 5,8,11,14-eicosatetraienoic acid (ETYA), hydroxyphenylretinamide, lonapalene, esculin, diethylcarbamazine, phenanthroline, baicalein, proxicromil, thioethers, diallyl sulfide and di- (1-propenyl) sulfide.
[0317] [000317] Leukotriene receptor antagonists include calcitriol, ontazolast, Bayer's Bay-x-1005, Ciba-Geigy's CGS-25019C, ebselene, Leo Denmark's ETH-615, LY-293111 from Lilly, Ono's ONO-4057, Terumo's TMK-688, Boehringer Ingleheim's BI-RM-270, Lilly's LY 213024, Lilly's LY 264086, Lilly's LY 292728, Ono's ONO LB457, Pfizer's 105696, Perdue Frederick's PF 10042, Rhone-Poulenc Rorer's RP 66153, SmithKline Beecham's SB-201146, SmithKline Beecham's SB-201993, SmithKline Beecham's SB-209247, SmithKline Beecham's SB-209247 Searle, Sumitamo's SM 15178, American Home Products' WAY 121006, Bayer's Bay-o-8276, Warner-Lambert's CI-987, Warner-Lambert's CI987BPC-15LY 223982, Lilly's LY 233569, Lilly's LY255283, MacroNex's MNX-160, Merck and Co.'s MK-591, Merck and Co.'s MK-886, Ono's ONO-LB-448, Purdue Frederick's PF-5901, RG 14893 from Rhone-Poulenc Rorer, RP 66364 from Rhone-Poulenc Rorer, RP 69698 from Rhone-Poul enc Rorer, Shionoogi's S-2474, Searle's SC-41930, Searle's SC-50505, Searle's SC-51146, Searle's SC-52798, SmithKline Beecham's SK and F-104493, SR -2566 from Leo Denmark, T-757 from Tanabe and TEI-1338 from Teijin. Manufacturing Items
[0318] [000318] In another aspect, a manufacturing article containing materials useful for the treatment of the disorders described above is included. The article of manufacture comprises a container and a label. Suitable containers include, for example, bottles, small vials, syringes and test tubes. Containers can be formed from a variety of materials, such as glass or plastic. The container comprises a composition that is effective in treating the condition and may have a sterile access port. For example, the container can be a bag of intravenous solution or a small vial having a cap that can be snapped off by a hypodermic injection needle. The active agent in the composition is the humanized anti-CD40 antibody. The label on, or associated with, the container indicates that the composition is used to treat the condition of choice. The article of manufacture may additionally comprise a second container comprising a pharmaceutically acceptable buffer, such as a phosphate buffered saline, a Ringer's solution, and a dextrose solution. It may additionally include other materials desirable from a commercial and user point of view, including other buffers, thinners, filters, needles, syringes, and instructions for use.
[0319] [000319] The invention is further described in the following examples, which are not intended to limit the scope of the invention. EXAMPLES Example 1: Production of Humanized Anti-CD40 Antibody
[0320] [000320] The antibodies of mice 20E2, and 2H11 are shown in Tables 1 and 2 contained herein above. The humanization of clones 20E2, and 2H11 has been completed. A library was made, where the residues of humans and rats were varied in such a way that in any given position there could be a residue from human or rat. Such a library was made for amino acids that were different between the human germline and the mouse antibody. Only clones that retain the function of the original mouse antibody were selected.
[0321] [000321] In this mode, Antibody a, Antibody B and Antibody C were humanized antibodies derived from mouse antibody 20E2 (Antibody A and Antibody B) or 2H11 (Antibody C) cloned in an IgG1-KO (KO = knockout) human / main chain cover. IgG1- KO has two mutations in the Fc region, Leu234Ala and Leu235Ala, to reduce binding to FcγR and complement.
[0322] [000322] The results of such humanization have resulted in several variable sequences of the humanized heavy and light chains, shown below: SEQ ID NO: 41 (variable light chain sequence): DIVMTQSPDSLAVSLGERVTMSCKSSQSLLNSGNQKNYLTWHQQKPGQPPKLLIYWTSTRESGVPDRFSGSGSGTDFTLTISSLQAEDVAVYYCQNDYTYPLTFGGGTKVEIK SEQ ID NO: 42 (variable heavy chain sequence): EVQLVKSGGGLVKPGGSLRLSCAASGFTFSDYGMHWVRQAPGKGLEWVAYISSGNRIIYYADTVKGRFTISRDNAKNSLYLQMNSLRAEDTALYYCARQDGYRYAMDYWGQGTLVTVSS SEQ ID NO: 43 (variable light chain sequence) DIVMTQSPDSLAVSLGERATMSCKSSQSLLNSGNQKNYLTWHQQKPGQPPKLLIYWTSTRESGVPDRFSGSGSGTDFTLTISSLQAEDVAVYYCQNDYTYPLTFGGGTKVEIK SEQ ID NO: 44 (variable heavy chain sequence) EVQLVESGGGLVKPGGSLRLSCAASGFTFSDYGMHWVRQAPGKGLEWVAYISSGNRIIYYADTVKGRFTISRDNAKNSLYLQMNSLRAEDTALYYCARQDGYRYAMDYWAQGTLVTVSS SEQ ID NO: 45 (variable light chain sequence) DIVMTQSPDSLAVSLGEKVTMNCKSSQSLLNSGNQKNYLTWHQQKPGQPPKLLIYWTSTRESGVPDRFSGSGSGTDFTLTISSLQAEDVAVYYCQNDYTYPLTFGAGTKVEIK SEQ ID NO: 46 (variable heavy chain sequence) EVQLVESGGGLVKPGGSRRLSCAASGFTFSDYGMHWVRQAPGKGLEWVAYISSGNRIIYYADTVKGRFTISRDNAKNSLYLQMNSLRAEDTALYYCARQDGYRYAMDYWGQGTLVTVSS. SEQ ID NO: 47 (variable light chain sequence) DIVMTQSPDSLAVSLGERVTMNCKSSQSLLNSGNQKNYLTWHQQKPGQPPKLLIYWTSTRESGVPDRFSGSGSGTDFTLTISSLQAEDVAVYYCQNDYTYPLTFGGGTKVEIK SEQ ID NO: 48 (variable heavy chain sequence) EVQLVESGGGLVKPGGSLRLSCAASGFTFSDYGMHWVRQAPGKGLEWVAYISSGNRIIYYADTVKGRFTISRDNAKNSLYLQMNSLRAEDTALYYCARQDGYRYAMDYWGQGTLVTVSS SEQ ID NO: 49 (variable light chain sequence) DIVMTQSPDSLAVSLGERVTMNCKSSQSLLNSGNQKNYLTWHQQKPGQPPKLLIYWTSTRESGVPDRFSGSGSGTDFTLTISSLQAEDVAVYYCQNDYTYPLTFGAGTKVEIK SEQ ID NO: 50 (variable light chain sequence) EVQLVESGGGLVKPGGSRRLSCAASGFTFSDYGMHWVRQAPGKGLEWVAYISSGNRIIYYADTVKGRFTISRDNAKNSLYLQMNSLRAEDTAVYYCARQDGYRYAMDYWGQGTLVTVSS SEQ ID NO: 51 (variable light chain sequence) DIVMTQSPDSLAVSLGEKVTMNCKSSQSLLNSGNQKNYLTWHQQKPGQPPKLLIYWTSTRESGVPDRFSGSGSGTDFTLTISSLQAEDLAVYYCQNDYTYPLTFGAGTKVEIK. SEQ ID NO: 52 (variable light chain sequence) DIVMTQSPDSLAVSLGEKVTINCKSSQSLLNSGNQKNYLTWHQQKPGQPPKLLIYWTSTRESGVPDRFSGSGSGTDFTLTISSLQAEDVAVYYCQNDYTYPLTFGGGTKVEIK SEQ ID NO: 53 (variable heavy chain sequence) EVQLVESGGGLVKPGGSLRLSCAASGFTFSDYGMHWVRQAPGKGLEWVAYISSGNRIIYYADTVKGRFTISRDNAKNSLYLQMNSLRAEDTAVYYCARQDGYRYAMDYWGQGTLVTVSS SEQ ID NO: 54 (variable light chain sequence) QIQMTQSPSSLSASVGDRVTITCSASSSVSYMLWFQQKPGKAPKLWIYSTSNLASGVPARFSGSGSGTDFTLTISSLQPEDFATYYCQQRTFYPYTFGGGTKVEIK SEQ ID NO: 55 (variable light chain sequence) DIQMTQSPSSLSASVGDRVTITCSASSSVSYMLWFQQKPGKAPKLLIYSTSNLASGVPARFSGSGSGTDFTLTISSLQPEDFATYYCQQRTFYPYTFGGGTKVEIK SEQ ID NO: 56 (variable light chain sequence) DIQMTQSPSSLSASVGDRVTITCSASSSVSYMLWFQQKPGKAPKLLIYSTSNLASGVPSRFSGSGSGTDFTLTISSLQPEDFATYYCQQRTFYPYTFGGGTKVEIK SEQ ID NO: 57 (variable heavy chain sequence) QVQLVQSGAEVKKPGASVKVSCTASGFNITDYYVHWVKQRPGQGLEWMGRIDPEDGDSKYAPKFQGKATMTADTSTSTVYMELSSLRSEDTAVYYCTTSYYVGTYGYWGQGTLVTVSS. SEQ ID NO: 58 (variable heavy chain sequence) QVQLVQSGAEVKKPGASVKVSCTASGFNIKDYYVHWVKQAPGQGLEWMGRIDPEDGDSKYAPKFQGKATMTADTSTSTVYMELSSLRSEDTAVYYCTTSYYVGTYGYWGQGTLVTVSS SEQ ID NO: 59 (variable heavy chain sequence) QVQLVQSGAEVKKPGASVKVSCTASGFNITDYYVHWVKQRPGQGLEWMGRIDPEDGDSKYAPKFQGKVTMTADTSTSTVYMELSSLRSEDTAVYYCTTSYYVGTYGYWGQGTLVTVSS. SEQ ID NO: 60 (variable heavy found string) QVQLVQSGAEVKKPGASVKVSCTASGFNIKDYYVHWVKQAPGQGLEWIGRIDPEDGDSKYAPKFQGKATMTADTSTSTVYMELSSLRSEDTAVYYCTTSYYVGTYGYWGQGTLVTVSS SEQ ID NO: 61 (variable heavy sequence) QVQLVQSGAEVKKPGASVKVSCTASGFNITDYYVHWVKQAPGQGLEWMGRIDPEDGDSKYAPKFQGKATMTADTSTSTVYMELSSLRSEDTAVYYCTTSYYVGTYGYWGQGTLVTVSSSS SEQ ID NO: 62 (variable heavy sequence): QVQLVQSGAEVKKPGASVKVSCTASGFNITDYYVHWVKQRPGQGLEWMGRIDPEDGDTKFAPKFQGKATMTADTSTSTVYMELSSLRSEDTAVYYCTTSYYVGTYGYWGQGTLVTVSS SEQ ID NO: 63 (variable heavy sequence) QVQLVQSGAEVKKPGASVKVSCTASGFNITDYYVHWVKQRPGQGLEWMGRIDPEDGDTKFAPKFQGKVTMTADTSTSTVYMELSSLRSEDTAVYYCTTSYYVGTYGYWGQGTLVTVSS SEQ ID NO: 64 (variable heavy string) QVQLVQSGAEVKKPGASVKVSCTASGFNIKDYYVHWVKQAPGQGLEWIGRIDPEDGDTKFAPKFQGKATMTADTSTSTVYMELSSLRSEDTAVYYCTTSYYVGTYGYWGQGTLVTVSS SEQ ID NO: 65 (variable heavy sequence) QVQLVQSGAEVKKPGASVKVSCTASGFNIKDYYVHWVKQAPGQGLEWMGRIDPEDGDTKFAPKFQGKATMTADTSTSTVYMELSSLRSEDTAVYYCTTSYYVGTYGYWGQGTLVTVSS SEQ ID NO: 66 (variable heavy sequence) QVQLVQSGAEVKKPGASVKVSCTASGFNITDYYVHWVKQAPGQGLEWMGRIDPEDGDTKFAPKFQGKATMTADTSTSTVYMELSSLRSEDTAVYYCTTSYYVGTYGYWGQGTLVTVSS SEQ ID NO: 67 (variable heavy sequence) EVQLVQSGAEVKKPGATVKISCKVSGFNIKDYYIHWVKQRPGKGLEWMGRIDPEDGDTKYDPKFQGRVTMTADTSTDTAYMELSSLRSEDTAVYYCTTSYYVGTYGYWGQGTTVTVSSSS SEQ ID NO: 68 (variable heavy string) EVQLVQSGAEVKKPGATVKISCTVSGFNIKDYYIHWVKQRPGKGLEWMGRIDPEDGDTKYDPKFQGRVTMTADTSTDTAYMELSSLRSEDTAVYYCTTSYYVGTYGYWGQGTTVTVSSSS SEQ ID NO: 69 (variable heavy sequence) EVQLVQSGAEVKKPGATVKISCTVSGFNIKDYYIHWVKQRPGKGLEWMGRIDPEDGDTKYDPKFQGKVTMTADTSTDTAYMELSSLRSEDTAVYYCTTSYYVGTYGYWGQGTTVTVSSSS SEQ ID NO: 70 (variable heavy string) EVQLVQSGAEVKKPGATVKISCTVSGFNIKDYYIHWVKQAPGKGLEWMGRIDPEDGDTKYDPKFQGKATMTADTSTDTAYMELSSLRSEDTAVYYCTTSYYVGTYGYWGQGTTVTVSSSS SEQ ID NO: 71 (variable heavy sequence) EVQLVQSGAEVKKPGATVKISCTVSGFNIKDYYIHWVKQRPGKGLEWMGRIDPEDGDTKYDPKFQGKATMTADTSTDTAYMELSSLRSEDTAVYYCTTSYYVGTYGYWGQGTTVTVSSSS SEQ ID NO: 72 (variable heavy sequence) EVQLVQSGAEVKKPGATVKISCTVSGFNIKDYYIHWVKQAPGKGLEWIGRIDPEDGDTKYDPKFQGKATMTADTSTDTAYMELSSLRSEDTAVYYCTTSYYVGTYGYWGQGTTVTVSSSS SEQ ID NO: 73 (variable heavy sequence) EVQLVQSGAEVKKPGATVKISCKVSGFNIKDYYIHWVQQAPGKGLEWMGRIDPEDGDTKYDPKFQGRVTMTADTSTDTAYMELSSLRSEDTAVYYCTTSYYVGTYGYWGQGTTVTVSSSS SEQ ID NO: 74 (variable light sequence) 1 from antibody 10F2Hum: DIQMTQSPSSLSASVGDRVTITCSATSSVSYILWFQQKPGKAPKLLIYSTSNLASGVPSRFSGSGSGTDFTLTISSLQPEDFATYYCQQRTFYPY TFGGGTKVEIK SEQ ID NO: 75 (variable light sequence) 2 from 10F2Hum antibody: DIQMTQSPSSLSASVGDRVTITCSATSSVSYILWFQQKPGKAPKLLIYSTSNLASGVPARFSGSGSGTDFTLTISSLQPEDFATYYCQQRTFYPYTFGGGTKVEIK SEQ ID NO: 76 (variable light sequence) QIQMTQSPSSLSASVGDRVTITCSATSSVSYILWFQQKPGKAPKLWIYSTSNLASGVPARFSGSGSGTDFTLTISSLQPEDFATYYCQQRTFYPYTFGGGTKVEIK
[0323] [000323] The humanized antibodies illustrative of the present invention are those having the heavy and light chain sequences, shown in the table below. The strings in bold underlined in the following table are the variable domains, while the normal strings, not underlined, are the constant domains:
[0324] A) um formato de IgG1-KO (nocaute) humana/capa com uma mutação dupla de Leu234Ala, Leu235Ala na região Fc, para reduzir a função efetora, tal como a ligação ao FcγR e ao complemento b) um formato de IgG4-DM (mutante duplo) humana/capa com uma mutação de Ser228Pro na região de articulação, para reduzir a ocorrência de semimoléculas de IgG4, e uma mutação de Leu235Glu para reduzir adicionalmente a ligação ao FcγR [000324] The variable regions have been subcloned into one or two different suitable IgG expression vectors: A) a human IgG1-KO (knockout) / cap format with a double mutation of Leu234Ala, Leu235Ala in the Fc region, to reduce effector function, such as binding to FcγR and complement b) a human IgG4-DM (double mutant) / cap format with a Ser228Pro mutation in the joint region, to reduce the occurrence of IgG4 semimolecules, and a Leu235Glu mutation to further reduce FcγR binding
[0325] - Aspecto de CCF (turvação) - Propriedades de filtração de CCF - Rendimento sobre rProteínaA - Turvação na eluição e na neutralização - Agregados solúveis (SEC) - Padrão de pureza / contaminação (SDS) - Padrão de carga (IEF) [000325] The two candidates, Antibody A and Antibody B, have been purified and evaluated by the following criteria: - CCF aspect (turbidity) - CCF filtration properties - Yield on rProteinA - Turbidity in elution and neutralization - Soluble aggregates (SEC) - Purity / contamination standard (SDS) - Load standard (IEF)
[0326] [000326] Antibody A, Antibody B and Antibody C were characterized together with antibodies 4D11 (Kirin / Astellas) and PG-102 (PanGenetics), which were produced based on published sequences. The data for Antibody A, Antibody B, Antibody C and 4D11 are shown below. PG-102 showed agonistic activity and only incomplete inhibition of B cell proliferation (not shown). Table 2.2 summarizes the data obtained. A more detailed description of the data follows Table 2.2. Table 2.2. Summary of in vitro data from Kirin anti-CD40 antibody Antibody A, Antibody B and Antibody C and 4D11.
[0327] [000327] The specific binding of humanized antibodies to cellular CD40 was analyzed by flow cytometry, using HK293 cells transfected with human CD40. Concentration-dependent binding of Antibody A, Antibody B, and Antibody C was observed. The antibodies showed a similar binding profile, shown in Figure 1B. The EC50 values of the antibodies of the present invention and of the Kirin 4D11 antibody are all in the same range of ~ 1 nM, which is most likely at the sensitivity limit of the assay due to high levels of CD40 in the transfected cells. The specific binding of humanized antibodies to cellular CD40 on human Ramos cells also demonstrated concentration-dependent binding. The antibodies showed a slightly different binding profile (shown in Figure 2) and EC50 values between 0.21-1.22 nM. No binding was detected on CD40 negative cells, such as untransfected HEK293 cells or the HSB-2 T cell line, thus confirming selective binding to CD40 (data not shown).
[0328] [000328] The binding affinity of Antibody A, Antibody B and Antibody C to human CD40-Fc protein was measured using ForteBio Octet and revealed dissociation constants (KD) of <100 pM. Due to the avidity of antibody and CD40-Fc bivalence, the effects prevent Kds below 100 pM from being accurately determined. In addition, binding to CD40-Fc was analyzed in the absence and presence of 50% human serum and no significant effect of serum on binding was observed (data not shown). B. Activity of humanized antibodies in B cell activation / proliferation assays
[0329] [000329] The activity of humanized antibodies was tested in a B cell proliferation assay, in which human B cells derived from peripheral blood are stimulated with recombinant CD40L, in the presence of IL-2 and IL-4. Antibody B and Antibody C showed potent inhibition of B cell proliferation (shown in Figures 3A and 3B). The comparison with the inhibition curves and IC 50 values of the BI's antibodies and Kirin's 4D11 antibody indicates that the 4D11 antibody has a greater potency (Figures 3B and 4) when tested using multiple donors. When tested for agonistic activity in the absence of CD40L, the antibodies, Antibody B Antibody A and Antibody C, did not induce any proliferation of B cells above baseline levels at concentrations up to 10 g / ml (67 nM) (shown in Figure 4), similar to the 4D11 antibody.
[0330] [000330] The competing antibody 4D11 appeared to be slightly more potent, with an average IC50 of ~ 0.02 nM and no agonistic effects. The data for the three antibodies BI and 4D11 are summarized in Figure 4 and Table 2.2 described above. Another competing antibody, PG-102 (derived from clone 5D12), also tested in this assay, showed significant agonist effects, stimulating the proliferation of B cells in the absence of CD40L (Figure 4). Therefore, the absence of agonistic activity by our leadership candidates clearly differentiates them from the PG-102.
[0331] [000331] In a second assay, antibodies were tested for inhibition of CD86 overload in human B cells. In this situation, the assay can be performed with human whole blood or with purified B cells, both in the presence of exogenous CD40L. According to the B cell proliferation data, Antibody B, Antibody A and Antibody C tested in human whole blood showed potent inhibition of CD40-mediated CD86 uptake, as measured by flow cytometry (shown in Figure 5) . Antibody C showed similar potency to 4D11 in this assay, while the potency of Antibody B and Antibody A was somewhat weaker. The comparison of Antibody B and 4D11 on purified B cells or whole blood shows that the potency of Antibody B (IC50 and IC90 values) is relatively unchanged for purified B cells, compared to B cells in the presence of other cells or serum containing CD40, while 4D11 undergoes a dramatic change in potency in whole blood conditions (shown in Figure 6).
[0332] [000332] Similar data were developed when Antibody B, Antibody A and Antibody C were tested for inhibition of CD86 overload on B-monkey cells when performed with whole blood samples (shown in Figure 7). Antibody B, Antibody A and Antibody C, tested in whole blood of cynomolgus monkeys, showed potent inhibition of CD40-mediated CD86 overload, as measured by flow cytometry. All of these antibodies, therefore, show functional cross-reactivity to CD40 of a cynomolgus monkey, with similar potency to human CD40.
[0333] [000333] The activity of Antibody B IgG1KOb and Antibody B IgG1WT was assessed for the ability to mediate antibody-dependent cell cytotoxicity (Figure 13). In this assay, RAMOS cells were incubated with human PBMCs, in a 50: 1 effector-to-target cell ratio. Antibody B IgG1KOb and Antibody B IgG1WT were titrated from 20 µg / ml and the extent of cell death is monitored by LDH release. The data shown is from a representative experiment. The data show that IgG1Wt 20E2-12-RIgG1WT Antibody is an effective mediator of ADCC and that IgG1KOb Antibody containing mutations that eliminate effector function has no ADCC activity. Example 3: Pharmacokinetic / Pharmacodynamic Studies A. IV administration of a single dose of Antibody A and Antibody B at 1 or 10 mg / kg in Cinomolgos Monkeys
[0334] [000334] Antibody A and Antibody B were dosed, each, at 1 and 10 mg / kg IV in male cynomolgus monkeys (N = 3) / dose. Blood samples were collected from 0-504 h (3 weeks), serum was recovered, and samples were stored at -20 ° C until analysis. The samples were analyzed by fitting ELISA, as described above. The serum concentration / time profiles of both antibodies in monkeys, after both IV doses, and the pharmacokinetic parameters are summarized in Figure 8 and Tables 2.7.1 (Antibody A) and 2.7.2 (Antibody B) shown below . Both antibodies showed dose-dependent pharmacokinetics, suggesting that, at a low dose, clearance is predominantly attributable to consistent with the target-mediated disposition, while at a higher dose, the antibody is mainly cleared by catabolism. Similar dose-dependent pharmacokinetic profiles have been observed for other MAbs targeting membrane associated targets (eg, CD19, CD20, EGFR, CD146 and HER2). The clearance for Antibody A was 0.8 and 0.1 mL / h / kg for doses of 1 and 10 mg / kg, respectively. The clearance for Antibody B was 0.7 and 0.1 mL / h / kg for doses of 1 and 10 mg / kg, respectively. Similarly, the half-life of Antibody A was 1 and 13 days for the doses of 1 and 10 mg / kg, respectively, and the half-life of Antibody B was 2 and 13 days for the same respective doses. Although Antibody B had a marginally longer half-life at the lowest dose, compared to the same dose for Antibody A, this difference would not be expected to convert to more continuous exposure with chronic administration. The AUC for both compounds was supra-proportional and the volume of distribution (Vss) for both compounds approached that of the plasma volume (~ 40 mL / kg), exhibiting the limited tissue distribution typically seen for protein therapeutic substances polar, large. In all, there was no appreciable difference in pharmacokinetic parameters between the two antibodies. Table 2.7.1: Pharmacokinetic parameters of Antibody A in male cynomolgus monkeys, male (N = 3) / dose after individual IV doses of 1 and 10 mg / kg.
[0335] [000335] As part of the PK study described above, we analyzed the pharmacodynamic effects of anti-CD40 antibodies. For this purpose, whole blood samples were incubated with recombinant CD40L overnight and the increase in CD86 expression on B cells was determined by flow cytometry. The samples were analyzed on day 0 (pre-treatment), day 2, 7 and 14 after dosing. Although the increase in CD86 expression is relatively small (~ 5-20-%), a dose-dependent effect was observed (shown in Figure 9). In the group of animals dosed with 10 mg / kg of Antibody A and Antibody B, CD86 induction was completely inhibited at 2, 7 and 14 days, consistent with continuous exposure at this dose. Animals dosed at 1 mg / kg showed complete inhibition on day 2, partial inhibition on day 7 and no inhibition on day 14. Loss of pharmacodynamic effect over time correlates with faster antibody clearance in the dose group low. Example 4: Toxicity-related studies: CD40 on platelets
[0336] [000336] CD40 is constitutively expressed on human platelets (Henn, et al., 2001) and (Inwald, et al., 2003), while CD40L is rapidly and transiently expressed on the cell surface of activated platelets (Henn, et al. et al., 2001). Although anti-CD40 antibodies without binding to FcR were not expected to have effects on platelets, it is important to demonstrate directly that this is the case. Flow cytometry studies were performed to demonstrate the link of anti-CD40 leadership candidates to human and cinomolg platelets.
[0337] [000337] Before, it had been demonstrated by flow cytometry that G28.5 and anti-CD40 mAb 89 bind to human platelets at rest (Henn, et al., 2001). This was confirmed using the F28C-labeled G28.5 antibody. Serial 5-fold dilutions of G28.5 were prepared and a range of 0.5 µg / ml to 0.32 ng / ml was incubated on some 100 µl of platelets obtained from humans (2 donors) or cynomolgus monkeys ( 3 donors), for 30 minutes, at room temperature. In addition, APC-labeled anti-CD45 mAb was used to identify platelets bound to other types of CD40 + cells in order to exclude these cells from analysis. After staining the antibody, the platelets were washed and fixed with Optilyse C and flow cytometry was performed. The mean fluorescence intensity (MFI) was determined as a measure of antibody binding to CD45 platelets.
[0338] [000338] The commercially available 5c3 and selected antibodies of the anti-CD40 mouse mAb invention were labeled with FITC. Binding to Ramos cells has been confirmed. The number of FITC molecules per antibody molecule ranged from 2 to 4 FITC per antibody molecule. Serial five-fold dilutions of the commercial mAb and anti-CD40 candidate were prepared ranging from 0.5 µg / ml to 0.32 ng / ml and incubated with human platelet (3 donors) and cinomolg (2 donors) for 30 minutes at room temperature.
[0339] [000339] A representative graph demonstrating the binding of the mouse candidate anti-CD40 mAb to human platelets is shown in Figure 11. The four candidate monoclonal antibodies showed specific binding to human platelets, compared to the FITC-labeled isotype control antibody. . 10F2, 2H11, 19B10 and 20E2 showed comparable binding to platelets. A similar trend was observed for cinomolgos platelets (data not shown).
[0340] [000340] In addition to these studies, Antibody B directly labeled and 4D11 were compared for their ability to bind platelets and B cells in samples of human whole blood and cynomolgus monkeys (shown in Figure 12) 4D11 showed a similar link (as exemplified by EC50) for both B cells and platelets in human blood samples and cynomolgus monkeys. Antibody B showed a similar pattern, but with much weaker binding power. Example 5: In vivo pharmacology studies in the NSG mouse model
[0341] [000341] The efficacy of the humanized antibodies, Antibody A, was evaluated in an antibody production model, where human PBMC's were injected into immunodeficient NSG mice to generate a graft versus response. host. Significant production of human IgM (hIgM) and IgG (hIgG) can be detected starting 2 weeks after grafting. Treatment with Antibody A at doses of 5 and 1 mg / kg significantly inhibited the response of hIgG and hIgM at weeks 2 and 3 after grafting. A comparator antibody (4D11) was evaluated in a single dose of 5 mg / kg and also demonstrated nullification of the response. In a second study, all antibodies, Antibody A, Antibody B and Antibody C, were tested in a single dose of 1 mg / kg and showed complete inhibition of the IgM and IgG response at week 2 (Figure 10). Example 6: Analysis of the biomarker Receptor overload
[0342] [000342] Receptor overload induced by CD40L can be measured by flow cytometry. Human whole blood can be stimulated with optimized concentration of soluble CD40L and the total percentage of CD20 + Receptor + cells can be measured by flow cytometry. The change in the percentage of CD86 expression on CD20 positive cells was measured in parallel with the cyanomologous pk study evaluating Antibodies A and B (Figure 9). The data shows the inhibition of CD86 overload at time points consistent with the exposure of the antibodies. Targeted Proteomics:
[0343] [000343] Increased protein secretion with stimulation of CD40 in whole blood can be used as a potential biomarker (s). An optimized concentration of soluble CD40L and the stimulation time were established using a multiplex Luminex blood cell platform that detects MDC / CCL22 and several other secreted proteins. Clinical samples will be evaluated from human whole blood in the total dose range of anti-CD40 mAb. Receiver occupation:
[0344] [000344] CD40 receptor occupation can be determined in an in vitro or ex vivo assay based on cytometric analysis of B cell flow in human whole blood. Current candidates, Antibody of the present invention and non-competing anti-CD40 antibody 5C3, will be used to quantify the receptor occupation assay. Example 7: Anti-Tumor Activity of the Humanized Anti-CD40 Antibody
[0345] [000345] In some situations, it may be desirable to determine the anti-tumor properties of the antibodies of the present invention. Such determination can be made by testing the anti-tumor activity of the humanized anti-CD40 antibody in a SCID mouse lymphoma xenograft model. Such a SCID model can be injected with cancer cells to present a tumor, for example, 5 x 106 million tumor cells can be injected subcutaneously into SCID mice (10 / group), thirteen days before starting treatment with the drug. The anti-CD40 antibodies of rats of the present invention or a comparison (for example, a humanized control antibody or the other) is given intraperitoneally 3 times a week (4 mg / kg / dose), with 8 or 5 doses administered. The development and growth of tumors are monitored in the mouse and the tumor volume can be measured weekly during the selected study period, for example, a 14-day study period. Preferably, the results will show an increase of 2, 3, 4, 5, 6, 7, 8, 9, 10 or more times in the growth of tumors in the control mice, compared to the mice treated with the antibodies of the present invention. . Preferably, during the treatment period, tumor growth in mice treated with the antibodies of the invention will be insignificant. Such data can confirm that the humanized antibody being tested is effective in suppressing tumor growth in this model of lymphoma xenograft b. Example 8: Prolonged survival by the Humanized Anti-CD40 Antibody
[0346] [000346] The efficacy of the humanized anti-CD40 antibody on the survival of mice containing tumor, such as those described above, can be tested in a SCID mouse lymphoma xenograft model. SCID mice (10 / group) are inoculated intravenously with 1 x 106 million tumor cells, three days before treatment with the antibody. The mice are then treated with the rat or humanized anti-CD40 antibodies of the present invention or an Ig control, administered intraperitoneally, twice a week (4 mg / kg / dose), for a total of five doses. The housings of the mice can then be examined daily for mortality to determine the level of effectiveness of the antibodies in prolonging the survival of a patient with cancer.
[0347] [000347] Various references, including patent applications, patents, and scientific publications, are cited in this document, the disclosures of which are incorporated herein by reference in their entirety. The citation or identification of any reference in this document will not be interpreted as an admission that such reference is available as a prior art to the present invention.
[0348] [000348] The preferred aspects of the present invention can be described according to the modalities in the following paragraphs:
[0349] [000349] Paragraph 1. A humanized monoclonal antibody, where said antibody specifically binds to human CD40 having an IC50 antagonistic activity of less than 1 nM and has no agonism up to 100 µg / ml in B cell proliferation and where the said antibody is further characterized by the fact that the antibody has an in vivo half-life in non-human primates that is at least 10 days.
[0350] [000350] Paragraph 2. The humanized monoclonal antibody of Paragraph 1, where said antibody has a half-life in monkeys monkeys of more than 8 days, at a dose of less than 30 mg / kg.
[0351] [000351] Paragraph 3. The antibody of Paragraph 1, where the antibody comprises a sequence of the heavy chain selected from the group consisting of any of SEQ ID NO: 1 through SEQ ID NO: 4 and a sequence of the selected light chain a from the group consisting of any of SEQ ID NO: 5 through SEQ ID NO: 8.
[0352] [000352] Paragraph 4. The antibody of Paragraph 1, wherein said antibody is a humanized antibody or antigen binding fragment of an antibody having the amino acid sequence of the variable region of any heavy chain of SEQ ID NO: 1 to 4 , SEQ ID NO: 27, SEQ ID NO: 28, SEQ ID NO: 29, SEQ ID NO: 30, SEQ ID NO: 32, SEQ ID NO: 33, SEQ ID NO: 34, SEQ ID NO: 35, SEQ ID NO: 37, SEQ ID NO: 38, SEQ ID NO: 39, SEQ ID NO: 40, SEQ ID NO: 42, SEQ ID NO: 44, SEQ ID NO: 46, SEQ ID NO: 48, SEQ ID NO . 50 SEQ ID NO: 53, SEQ ID NO: 57, SEQ ID NO: 58, SEQ ID NO: 59, SEQ ID NO: 60, SEQ ID NO: 61, SEQ ID NO: 62, SEQ ID NO: 63, SEQ ID NO: 64, SEQ ID NO: 65, SEQ ID NO: 66, SEQ ID NO: 67, SEQ ID NO: 68, SEQ ID NO: 69, SEQ ID NO: 70, SEQ ID NO: 71, SEQ ID NO : 72, or SEQ ID NO: 73.
[0353] [000353] Paragraph 5. The antibody of Paragraph 1, wherein said antibody is a humanized antibody or antigen-binding fragment of an antibody comprising an amino acid sequence of the light chain variable domain of SEQ ID NO: 5 through SEQ ID NO: 8, SEQ ID NO: 26, SEQ ID NO: 31, SEQ ID NO: 36, SEQ ID NO: 41, SEQ ID NO: 43, SEQ ID NO: 45, SEQ ID NO: 47, SEQ ID NO: 49, SEQ ID NO: 50, SEQ ID NO: 51, SEQ ID NO: 52, SEQ ID NO: 54, SEQ ID NO: 55, SEQ ID NO: 56, SEQ ID NO: 74, SEQ ID NO: 75, or SEQ ID NO: 76.
[0354] [000354] Paragraph 6. The monoclonal antibody of Paragraph 1, wherein said antibody comprises a heavy chain and a light chain, where the CDR1 sequence of the heavy chain selected from the group consisting of SEQ ID NO: 9 through SEQ ID NO: 11, a CDR2 sequence of the heavy chain selected from the group consisting of SEQ ID NO: 12 through SEQ ID NO: 15 and a CDR3 sequence of the heavy chain selected from the group consisting of SEQ ID NO: 16 through SEQ ID NO: 17; and where the light chain CDR1 sequence has a sequence selected from the group consisting of SEQ ID NO: 18 through SEQ ID NO: 21, a CDR2 sequence from the light chain from SEQ ID NO: 22 to SEQ ID NO: 23 and a CDR3 sequence of the light chain selected from the group consisting of SEQ ID NO: 24 through SEQ ID NO: 25.
[0355] [000355] Paragraph 7. The monoclonal antibody of Paragraph 1, wherein said antibody comprises a sequence of CDR1 of the heavy chain of SEQ ID NO: 10, a sequence of CDR2 of the heavy chain of SEQ ID NO: 13 and a sequence of CDR3 of the heavy chain of SEQ ID NO: 16 and where said antibody comprises a sequence of CDR1 of the light chain of SEQ ID NO: 19, a sequence of CDR2 of the light chain of SEQ ID NO: 22 and a sequence of CDR3 of the light chain of SEQ ID NO: 24.
[0356] [000356] Paragraph 8. The monoclonal antibody of Paragraph 1, wherein said antibody comprises a sequence of CDR1 of the heavy chain of SEQ ID NO: 9, a sequence of CDR2 of the heavy chain of SEQ ID NO: 14 and a sequence of CDR3 of the heavy chain of SEQ ID NO: 16 and where said antibody comprises a sequence of CDR1 of the light chain of SEQ ID NO: 20, a sequence of CDR2 of the light chain of SEQ ID NO: 22 and a sequence of CDR3 of the light chain of SEQ ID NO: 24.
[0357] [000357] Paragraph 9. An anti-CD40 antibody comprising a heavy chain variable domain sequence of any one of SEQ ID NOs: 1 through 4.
[0358] [000358] Paragraph 10. An anti-CD40 antibody comprising a light chain variable domain sequence of any one of SEQ ID NO: 5 through SEQ ID NO: 8.
[0359] [000359] Paragraph 12. A humanized antibody or antibody fragment having a heavy chain variable domain and a light chain variable region comprising the amino acid sequences of SEQ ID NO: 27 and SEQ ID NO: 26, respectively; SEQ ID NO: 28 and SEQ ID NO: 26, respectively; SEQ ID NO: 29 and SEQ ID NO: 26, respectively; SEQ ID NO: 30 and SEQ ID NO: 26, respectively; SEQ ID NO: 32 and SEQ ID NO: 31, respectively; SEQ ID NO: 33 and SEQ ID NO: 31, respectively; SEQ ID NO: 34 and SEQ ID NO: 31, respectively; SEQ ID NO: 35 and SEQ ID NO: 31, respectively; SEQ ID NO: 37 and SEQ ID NO: 36, respectively; SEQ ID NO: 38 and SEQ ID NO: 36, respectively; SEQ ID NO: 39 and SEQ ID NO: 36, respectively; SEQ ID NO: 40 and SEQ ID NO: 36, respectively.
[0360] [000360] Paragraph 13. An isolated antibody or antigen-binding fragment that specifically binds to human CD40, comprising a humanized heavy chain variable domain comprising a framework region having an amino acid sequence at least 90% identical to the amino acid sequence of the heavy chain amino acid sequence structure region of the human variable domain of SEQ ID NO: 27, SEQ ID NO: 28, SEQ ID NO: 29 or SEQ ID NO: 30, and comprising a light chain amino acid sequence by minus 90% identical to a variable domain of the corresponding light chain of SEQ ID NO: 26.
[0361] [000361] Paragraph 14. An isolated antibody or antigen-binding fragment that specifically binds to human CD40, comprising a humanized heavy chain variable domain comprising a framework region having an amino acid sequence at least 90% identical to the amino acid sequence the heavy chain amino acid sequence structure region of the human variable domain of SEQ ID NO: 32, SEQ ID NO: 33, SEQ ID NO: 34 or SEQ ID NO: 35, and comprising a light chain amino acid sequence by minus 90% identical to a corresponding light chain variable of SEQ ID NO: 31.
[0362] [000362] Paragraph 15. An isolated antibody or antigen-binding fragment that specifically binds to human CD40, comprising a humanized heavy chain variable domain comprising a framework region having an amino acid sequence at least 90% identical to the amino acid sequence the heavy chain amino acid sequence structure region of the human variable domain of SEQ ID NO: 37, SEQ ID NO: 38, SEQ ID NO: 39 or SEQ ID NO: 40, and comprising a light chain amino acid sequence by minus 90% identical to a corresponding light chain of SEQ ID NO: 36.
[0363] [000363] Paragraph 16. The antibody of Paragraph 1, where said antibodies are unable to stimulate the production of cytokines from B cells in this absence of CD40L.
[0364] [000364] Paragraph 17. The antibody of Paragraph 1, where said antibodies bind to human CD40 in the presence of 50% human serum with a reduction in the rate of less than twice.
[0365] [000365] Paragraph 18. The antibody of Paragraph 1, where said antibody produces inhibition of the production of IgM and IgG in a mammal at a concentration of 1 mg / kg.
[0366] [000366] Paragraph 19. A method of blocking the function of human CD40 in a mammal, comprising administering to said mammal a composition comprising an antibody of Paragraph 1 in an amount sufficient to block a CD40-mediated immune response in said mammal.
[0367] [000367] Paragraph 20. A method of treating or ameliorating graft vs. disease. host in a mammal, comprising administering to said mammal a composition comprising an antibody of Paragraph 1, in an amount sufficient to lessen one or more of the symptoms of graft vs. disease. host in said animal.
[0368] [000368] Paragraph 21. The method of Paragraph 20, where said mammal has an autoimmune or inflammatory disease selected from the group consisting of rheumatoid arthritis, multiple sclerosis, lupus proliferative glomerulonephritis, inflammatory bowel disease (IBD), psoriasis , idiopathic thrombocytopenic purpura (ITP), Crohn's disease and systemic lupus erythematosus (SLE), Hashimoto's thyroiditis, primary myxoedema, thyrotoxicosis / Graves' disease, pernicious anemia, autoimmune atrophic gastritis, autoimmune carditis, Addison's disease, menopause, premature, menopausal type 1 melito, Good pasture syndrome, myasthenia gravis, autoimmune hemolytic anemia, idiopathic leukopenia, primary biliary cirrhosis, chronic active hepatitis (HBs Ag negative), cryptogenic cirrhosis, Sjogren's syndrome, dermatomyositis, scleroderma, mixed connective tissue disease, lupus discoid erythematosus, and systemic vasculitis.
[0369] [000369] Paragraph 22. The method of Paragraph 19, where said mammal has rheumatoid arthritis.
[0370] [000370] Paragraph 23. The method of Paragraph 20, additionally comprising administering a second therapeutic agent selected from the group consisting of a TNF antagonist, a disease modifying antirheumatic drug, a CTLA4 antagonist, an IL- antireceptor mAb 6 and an anti-CD20 mAb.
[0371] [000371] Paragraph 24. The method according to Paragraph 20, where said inflammatory disease or autoimmune disease is an inflammatory disease or autoimmune disease that is associated with cells that express both CD40 and CD20.
[0372] [000372] Paragraph 25. The method of Paragraph 19, where said anti-CD40 antibody is administered by a parenteral route of administration.
[0373] [000373] Paragraph 26. The method of Paragraph 19, where said anti-CD40 antibody is administered intravenously or subcutaneously.
[0374] [000374] Paragraph 27. A method of inhibiting the production of antibodies by B cells in a human patient, comprising administering to said human patient an effective amount of an anti-CD40 antibody of Paragraph 1.
[0375] [000375] Paragraph 28. The method of Paragraph 27, where said human patient has an inflammatory disease or autoimmune disease that is associated with cells that express CD40.
[0376] [000376] Paragraph 29. The method of Paragraph 27, where said human patient is suffering from an autoimmune disease selected from the group consisting of autoimmune or inflammatory disease, selected from the group consisting of rheumatoid arthritis, multiple sclerosis, proliferative lupus glomerulonephritis, inflammatory bowel disease (IBD), psoriasis, idiopathic thrombocytopenic purpura (ITP), Crohn's disease and systemic lupus erythematosus (SLE), Hashimoto's thyroiditis, primary myxedema, thyrotoxicosis / gastric disease, Grave disease, severe gastric disease atrophic autoimmune, autoimmune carditis, Addison's disease, premature menopause, type 1 diabetes mellitus, Good pasture syndrome, myasthenia gravis, autoimmune hemolytic anemia, idiopathic leukopenia, primary biliary cirrhosis, active chronic hepatitis (HBs Ag negative), cryptogenic cirrhosis, syndrome de Sjogren, dermatomyositis, scleroderma, mixed connective tissue disease, discoid lupus erythematosus, and systemic vasculitis.
[0377] [000377] Paragraph 30. A method for inhibiting the growth of cells expressing the human CD40 antigen, comprising administering the antibody or antigen-binding fragment of Paragraph 1 to the cells, antibody or antigen-binding fragment which specifically binds to the CD40 antigen on the human cell surface, where the binding of the antibody or antigen-binding fragment to the CD40 antigen inhibits cell growth or differentiation.
[0378] [000378] Paragraph 31. A method for treating a patient having a CD40-associated disorder, comprising administering to the patient the antibody or antigen-binding fragment of Paragraph 1, antibody or antigen-binding fragment which specifically binds to CD40 where the binding of the antibody or antigen-binding fragment to CD40 inhibits the growth or differentiation of cells from the CD40-associated disorder.
[0379] [000379] Paragraph 32. The method of Paragraph 31, where the cells of the disorder associated with CD40 are B lymphoblastoid cells, pancreatic cells, lung cells, breast cells, ovarian cells, colon cells, cells prostate cells, skin cells, head and neck cells, bladder cells, bone cells or kidney cells.
[0380] [000380] Paragraph 33. The method of Paragraph 31, where the disorder associated with CD40 is chronic lymphocytic leukemia, Burkitt's lymphoma, multiple myeloma, T-cell lymphoma, Non-Hodgkin's lymphoma, Hodgkin's disease , Waldenstrom's macroglobulinemia or Kaposi's sarcoma.
[0381] [000381] Paragraph 34. A method of inducing depletion of peripheral B cells, comprising administering to the cells the antibody or antigen-binding fragment of Paragraph 1, antibody or antigen-binding fragment which specifically binds to a CD40 antigen from the human cell surface, where the binding of the antibody or antigen-binding fragment to the CD40 antigen induces cell depletion.
[0382] [000382] Paragraph 35. The method of Paragraph 34, where the antibody or antigen binding fragment is administered to a patient having an immune disorder.
[0383] [000383] Paragraph 36. The method of Paragraph 34, where the immune disorder is rheumatoid arthritis or systemic lupus erythematosus.
[0384] [000384] Paragraph 37. A method of treating rheumatoid arthritis in a patient, comprising administering to said patient an antibody of Paragraph 1, wherein said antibody is an antagonistic antibody that blocks the function of CD40 in said patient.
[0385] [000385] Paragraph 38. The method of Paragraph 37, where said antibody is administered in an effective amount to inhibit B cell differentiation and antibody isotype exchange in said patient.
[0386] [000386] Paragraph 39. The method of Paragraph 37, wherein said antibody is administered in an effective amount to inhibit the production of cytokine and chemokine and the suppression of adhesion molecules in T cells and macrophages in said patient.
[0387] [000387] Paragraph 40. The method of Paragraph 37, where said antibody is administered in an effective amount to inhibit the activation of dendritic cells in said patient.
[0388] [000388] Paragraph 41. The method of Paragraph 37, wherein said antibody is administered in an effective amount to inhibit the production of proinflammatory cytokines, chemokines, matrix metalloproteinases, prostaglandins, and to unregulate the adhesion molecules in non-immune cells in said patient.
[0389] [000389] Paragraph 42. The method of Paragraph 37, wherein said antibody is administered in combination with a regimen comprising the administration of methotrexate and / or the administration of Enbrel / Humira.
[0390] [000390] Paragraph 43. The method of Paragraph 37, where said patient is a patient who has rheumatoid arthritis and has been unresponsive to treatment with methotrexate alone.
[0391] [000391] Paragraph 44. The method of Paragraph 43, wherein said method comprises treating said patient with a regimen comprising the administration of methotrexate and / or the administration of Enbrel / Humira.
[0392] [000392] Paragraph 45. The method of Paragraph 37, where the treatment of said patient with said antagonistic anti-CD40 antibody is more effective than treatment with methotrexate alone, Enbrel alone, a combination of Enbrel + methotrexate.
[0393] [000393] Paragraph 46. The method of Paragraph 43, where the treatment of said patient with said antagonistic anti-CD40 antibody is more effective than treatment with Enbrel + MTX in patients who have had an inadequate response to methotrexate.
[0394] [000394] Paragraph 47. The method of Paragraph 37, wherein said antibody is administered in combination with a regimen comprising an anti-TNF agent.
[0395] [000395] Paragraph 48. The method of Paragraph 37, where said patient is a patient who has rheumatoid arthritis and has been unresponsive to treatment with an anti-TNF agent alone.
[0396] [000396] Paragraph 49. The method of Paragraph 48, wherein said method comprises treating said patient with a regimen comprising treatment with an anti-TNF agent in combination with said antagonistic anti-CD40 antibody.
[0397] [000397] Paragraph 50. The method of Paragraph 37, where the treatment of said patient with said antagonistic anti-CD40 antibody is more effective than treatment with an anti-TNF agent.
[0398] [000398] Paragraph 51. The method of Paragraph 48, where the treatment of said patient with said antagonistic anti-CD40 antibody is more effective than treatment with Orencia or Rituxan, in patients who have had an inadequate response to an anti- TNF alone.
[0399] [000399] Paragraph 52. A pharmaceutical composition comprising: (i) the antibody or antigen-binding fragment of Paragraph 8; and (ii) a pharmaceutically acceptable excipient.
[0400] [000400] Paragraph 53. A pharmaceutical composition of Paragraph 52, wherein said antibody or its antigen-binding fragment is conjugated to a second agent.
[0401] [000401] Paragraph 54. The pharmaceutical composition of Paragraph 53, wherein said second agent is a cytotoxic agent, a PEG vehicle, an enzyme or a marker.
[0402] [000402] Paragraph 55. An isolated polynucleotide encoding an amino acid sequence from the variable region of any heavy chain of SEQ ID NO: 1 to 4, SEQ ID NO: 27, SEQ ID NO: 28, SEQ ID NO: 29, SEQ ID NO: 30, SEQ ID NO: 32, SEQ ID NO: 33, SEQ ID NO: 34, SEQ ID NO: 35, SEQ ID NO: 37, SEQ ID NO: 38, SEQ ID NO: 39, SEQ ID NO : 40, SEQ ID NO: 42, SEQ ID NO: 44, SEQ ID NO: 46, SEQ ID NO: 48, SEQ ID. AT THE. 50, SEQ ID NO: 53, SEQ ID NO: 57, SEQ ID NO: 58, SEQ ID NO: 59, SEQ ID NO: 60, SEQ ID NO: 61, SEQ ID NO: 62, SEQ ID NO: 63, SEQ ID NO: 64, SEQ ID NO: 65, SEQ ID NO: 66, SEQ ID NO: 67, SEQ ID NO: 68, SEQ ID NO: 69, SEQ ID NO: 70, SEQ ID NO: 71, SEQ ID NO: 72, or SEQ ID NO: 73.
[0403] [000403] Paragraph 56. An isolated polynucleotide encoding an amino acid sequence from the variable region of the light chain of any of SEQ ID NO: 5 to SEQ ID NO: 8, SEQ ID NO: 26, SEQ ID NO: 31, SEQ ID NO : 36, SEQ ID NO: 41, SEQ ID NO: 43, SEQ ID NO: 45, SEQ ID NO: 47, SEQ ID NO: 49, SEQ ID NO: 50, SEQ ID NO: 51, SEQ ID NO: 52 , SEQ ID NO: 54, SEQ ID NO: 55, SEQ ID NO: 56, SEQ ID NO: 74, SEQ ID NO: 75, or SEQ ID NO: 76.
[0404] [000404] Paragraph 57. A use of an antibody of Paragraph 1 in the manufacture of a medicine to block the function of human CD40 in a mammal, where the medicine blocks a CD40-mediated immune response in said mammal.
[0405] [000405] Paragraph 58. A use of an antibody of Paragraph 1 for the manufacture of a medicament to treat or ameliorate graft vs. disease. host in a mammal.
[0406] [000406] Paragraph 59. The use of Paragraph 58, where said medication is manufactured for the treatment of an autoimmune or inflammatory disease selected from the group consisting of rheumatoid arthritis, multiple sclerosis, lupus proliferative glomerulonephritis, inflammatory bowel disease (IBD), psoriasis, idiopathic thrombocytopenic purpura (ITP), Crohn's disease and systemic lupus erythematosus (SLE), Hashimoto's thyroiditis, primary myxoedema, thyrotoxicosis / Graves' disease, pernicious anemia, autoimmune atrophic gastritis, autoimmune disease, autoimmune carditis , premature menopause, type 1 diabetes mellitus, Good pasture syndrome, myasthenia gravis, autoimmune hemolytic anemia, idiopathic leukopenia, primary biliary cirrhosis, chronic active hepatitis (HBs Ag negative), cryptogenic cirrhosis, Sjogren's syndrome, dermatomyositis, scleroderma, disease mixed connective tissue, discoid lupus erythematosus, and systemic vasculitis.
[0407] [000407] Paragraph 60. A use according to Paragraph 58, wherein said drug additionally comprises a second therapeutic agent selected from the group consisting of a TNF antagonist, a disease modifying antirheumatic drug, a CTLA4 antagonist, an IL-6 anti-receptor mAb and an anti-CD20 mAb.
[0408] [000408] Paragraph 61. A use in accordance with Paragraph 57, where said medication is manufactured for use in a parenteral route of administration.
[0409] [000409] Paragraph 62. A use in accordance with Paragraph 57, where said medication is manufactured for use intravenously or subcutaneously.
[0410] [000410] Paragraph 63. A use of an antibody of Paragraph 1 in the manufacture of a medicament for inhibiting the production of antibodies by B cells in a human patient.
[0411] [000411] Paragraph 64. A use of an antibody of Paragraph 1 for the manufacture of a medicament for inhibiting the growth and / or differentiation of cells expressing the human CD40 antigen.
[0412] [000412] Paragraph 65. A use of an antibody of Paragraph 1 for the manufacture of a medicine for the treatment of a patient having a disorder associated with CD40, where the binding of the antibody or the antigen binding fragment in said medicine to CD40 inhibits the growth or differentiation of cells from the disorder associated with CD40.
[0413] [000413] Paragraph 66. A use according to Paragraph 65, where the drug is used for the treatment of cells of a CD40-associated disorder selected from B lymphoblastoid cells, pancreatic cells, lung cells, breast cells, cells ovarian cells, colon cells, prostate cells, skin cells, head and neck cells, bladder cells, bone cells or kidney cells.
[0414] [000414] Paragraph 67. The use according to Paragraph 65, where the drug is used for the treatment of chronic lymphocytic leukemia, Burkitt's lymphoma, multiple myeloma, T-cell lymphoma, Non-Hodgkin's lymphoma, Hodgkin's disease , Waldenstrom's macroglobulinemia or Kaposi's sarcoma.
[0415] [000415] Paragraph 68. A use of an antibody of Paragraph 1 in the manufacture of a medicament to induce depletion of peripheral B cells, where the antibody or antigen-binding fragment of the medicament specifically binds to a CD40 antigen on the cell surface human, where the binding of the antibody or antigen-binding fragment to the CD40 antigen induces cell depletion.
[0416] [000416] Paragraph 69. The use according to Paragraph 68, where the medication is for the treatment of a patient having an immune disorder.
[0417] [000417] Paragraph 70. The use according to Paragraph 68, where the drug is for the treatment of rheumatoid arthritis or systemic lupus erythematosus.
[0418] [000418] Paragraph 71. A use of an antibody of Paragraph 1 for the manufacture of a medicament for the treatment of rheumatoid arthritis in a patient.
[0419] [000419] Paragraph 72. The use according to Paragraph 71, where the drug is for inhibiting the differentiation of B cells and the exchange of antibody isotype in said patient.
[0420] [000420] Paragraph 73. The use in accordance with Paragraph 71, where the drug is for inhibiting the production of cytokine and chemokine and the overloading of adhesion molecules in T cells and macrophages in said patient.
[0421] [000421] Paragraph 74. The use according to Paragraph 71, where the drug is for inhibiting the activation of dendritic cells in said patient.
[0422] [000422] Paragraph 75. The use in accordance with Paragraph 71, where the drug is for the inhibition of the production of proinflammatory cytokines, chemokines, matrix metalloproteinases, prostaglandins, and infra-regulation of adhesion molecules in non-immune cells in said patient.
[0423] [000423] Paragraph 76. The use in accordance with Paragraph 71, where the medicine is a combination medicine to be administered in combination with a regimen comprising the administration of methotrexate and / or the administration of Enbrel / Humira.
[0424] [000424] Paragraph 77. The use according to Paragraph 71, where the drug additionally comprises an anti-TNF agent.
[0425] [000425] The application of the teachings disclosed in this document is not to be limited in scope by the specific modalities described here. In reality, several modifications will be within the capabilities of someone having common skill in the technique, considering the teachings contained in this document and the accompanying examples. Such modifications are intended to fall within the scope of the appended claims.
权利要求:
Claims (7)
[0001]
Humanized antibody, characterized by the fact that it has a heavy chain and a light chain comprising the amino acid sequences of SEQ ID NO: 35 and SEQ ID NO: 31, respectively.
[0002]
Use of an antibody, as defined in claim 1, characterized by the fact that it is for the preparation of a composition to treat or ameliorate a disease or disorder in a mammal.
[0003]
Use, according to claim 2, characterized by the fact that the disease or disorder is selected from the group consisting of: graft vs. disease. host, autoimmune or inflammatory disease, and cancer that expresses CD40.
[0004]
Use according to claim 3, characterized by the fact that autoimmune or inflammatory disease is selected from the group consisting of rheumatoid arthritis, multiple sclerosis, lupus proliferative glomerulonephritis, inflammatory bowel disease (IBD), psoriasis, idiopathic thrombocytopenic purpura (ITP ), Crohn's disease and systemic lupus erythematosus (SLE), Hashimoto's thyroiditis, primary myxoedema, thyrotoxicosis / Graves' disease, pernicious anemia, autoimmune atrophic gastritis, autoimmune carditis, Addison's disease, premature menopause, type 1 diabetes mellitus, syndrome 1 Good pasture, myasthenia gravis, autoimmune hemolytic anemia, idiopathic leukopenia, primary biliary cirrhosis, chronic active hepatitis (HBsAg negative), cryptogenic cirrhosis, Sjogren's syndrome, dermatomyositis, scleroderma, mixed connective tissue disease, discoid lupus erythematosus, and systemic vasculitis .
[0005]
Use according to any one of claims 2 to 4, characterized in that it additionally comprises a second selected therapeutic agent.
[0006]
Use according to any one of claims 2 to 5, characterized in that the composition comprising the antibody is administered parenterally, intravenously or subcutaneously.
[0007]
Pharmaceutical composition, characterized by the fact that it comprises: (i) an antibody as defined in claim 1; and (ii) a pharmaceutically acceptable excipient; the antibody or antigen-binding fragment thereof being optionally conjugated to a second agent.
类似技术:
公开号 | 公开日 | 专利标题
US11242394B2|2022-02-08|Anti-CD40 antibodies
US8492531B2|2013-07-23|Nucleic acids encoding humanized anti-CD40 antibodies
US20210261678A1|2021-08-26|Anti-cd40 antibodies for use in treating autoimmune disease
同族专利:
公开号 | 公开日
IL221639A|2016-07-31|
IL236740D0|2015-02-26|
CN102918063A|2013-02-06|
ES2807217T3|2021-02-22|
US20170058039A1|2017-03-02|
AU2011235214A1|2012-09-13|
TN2012000462A1|2014-01-30|
PT2796467T|2018-05-02|
IL236740A|2016-11-30|
WO2011123489A3|2011-12-01|
SI2796467T1|2018-05-31|
WO2011123489A2|2011-10-06|
EP3178851A1|2017-06-14|
CA2794332A1|2011-10-06|
MY155950A|2015-12-31|
HRP20180542T1|2018-05-04|
EP2796467A1|2014-10-29|
CA2794332C|2019-05-14|
ES2665592T3|2018-04-26|
HUE038788T2|2018-11-28|
DK2796467T3|2018-05-07|
DK3178851T3|2020-07-27|
HUE049849T2|2020-10-28|
US20190169301A1|2019-06-06|
MA34091B1|2013-03-05|
NZ602020A|2014-05-30|
TW201201836A|2012-01-16|
JP2015145383A|2015-08-13|
CY1120181T1|2018-12-12|
AP4082A|2017-03-29|
PT3178851T|2020-07-17|
US20150315282A1|2015-11-05|
US20160222124A1|2016-08-04|
EP2552965A2|2013-02-06|
JP5752865B1|2015-07-22|
US11242394B2|2022-02-08|
CO6612271A2|2013-02-01|
PL2796467T3|2018-07-31|
UA107827C2|2015-02-25|
AP2012006457A0|2012-08-31|
KR20130064723A|2013-06-18|
NO2796467T3|2018-08-25|
BR112012024713A2|2017-01-10|
MX2012011112A|2012-10-15|
ECSP12012221A|2012-11-30|
US20110243932A1|2011-10-06|
US9090696B2|2015-07-28|
EA031872B1|2019-03-29|
EA201201357A1|2013-04-30|
SG183947A1|2012-10-30|
PE20130159A1|2013-03-10|
HRP20201088T1|2020-10-30|
LT3178851T|2020-08-10|
KR101760242B1|2017-07-21|
GEP20166486B|2016-06-10|
JP2013523130A|2013-06-17|
US8591900B2|2013-11-26|
AR081750A1|2012-10-17|
CN102918063B|2016-06-29|
AU2011235214B2|2015-04-23|
LT2796467T|2018-05-10|
SI3178851T1|2020-09-30|
RS60612B1|2020-08-31|
EP2796467B1|2018-03-28|
PL3178851T3|2020-10-19|
EP3178851B1|2020-04-29|
UY33306A|2011-10-31|
CL2012002737A1|2013-02-15|
RS57114B1|2018-06-29|
MX340140B|2016-06-28|
TWI494126B|2015-08-01|
US20140179907A1|2014-06-26|
引用文献:
公开号 | 申请日 | 公开日 | 申请人 | 专利标题

US4179337A|1973-07-20|1979-12-18|Davis Frank F|Non-immunogenic polypeptides|
US4318980A|1978-04-10|1982-03-09|Miles Laboratories, Inc.|Heterogenous specific binding assay employing a cycling reactant as label|
US4275149A|1978-11-24|1981-06-23|Syva Company|Macromolecular environment control in specific receptor assays|
JPS6023084B2|1979-07-11|1985-06-05|Ajinomoto Kk|
WO1981001145A1|1979-10-18|1981-04-30|Univ Illinois|Hydrolytic enzyme-activatible pro-drugs|
US4419446A|1980-12-31|1983-12-06|The United States Of America As Represented By The Department Of Health And Human Services|Recombinant DNA process utilizing a papilloma virus DNA as a vector|
US4485045A|1981-07-06|1984-11-27|Research Corporation|Synthetic phosphatidyl cholines useful in forming liposomes|
NZ201705A|1981-08-31|1986-03-14|Genentech Inc|Recombinant dna method for production of hepatitis b surface antigen in yeast|
US4640835A|1981-10-30|1987-02-03|Nippon Chemiphar Company, Ltd.|Plasminogen activator derivatives|
US4601978A|1982-11-24|1986-07-22|The Regents Of The University Of California|Mammalian metallothionein promoter system|
US4560655A|1982-12-16|1985-12-24|Immunex Corporation|Serum-free cell culture medium and process for making same|
US4657866A|1982-12-21|1987-04-14|Sudhir Kumar|Serum-free, synthetic, completely chemically defined tissue culture media|
US4486414A|1983-03-21|1984-12-04|Arizona Board Of Reagents|Dolastatins A and B cell growth inhibitory substances|
US4816567A|1983-04-08|1989-03-28|Genentech, Inc.|Recombinant immunoglobin preparations|
DD266710A3|1983-06-06|1989-04-12|Ve Forschungszentrum Biotechnologie|Process for the biotechnical production of alkaline phosphatase|
US4544545A|1983-06-20|1985-10-01|Trustees University Of Massachusetts|Liposomes containing modified cholesterol for organ targeting|
US4767704A|1983-10-07|1988-08-30|Columbia University In The City Of New York|Protein-free culture medium|
US4496689A|1983-12-27|1985-01-29|Miles Laboratories, Inc.|Covalently attached complex of alpha-1-proteinase inhibitor with a water soluble polymer|
US4965199A|1984-04-20|1990-10-23|Genentech, Inc.|Preparation of functional human factor VIII in mammalian cells using methotrexate based selection|
US4879231A|1984-10-30|1989-11-07|Phillips Petroleum Company|Transformation of yeasts of the genus pichia|
US4737456A|1985-05-09|1988-04-12|Syntex Inc.|Reducing interference in ligand-receptor binding assays|
EP0206448B1|1985-06-19|1990-11-14|Ajinomoto Co., Inc.|Hemoglobin combined with a poly|
GB8516415D0|1985-06-28|1985-07-31|Celltech Ltd|Culture of animal cells|
US4927762A|1986-04-01|1990-05-22|Cell Enterprises, Inc.|Cell culture medium with antioxidant|
GB8610600D0|1986-04-30|1986-06-04|Novo Industri As|Transformation of trichoderma|
US4791192A|1986-06-26|1988-12-13|Takeda Chemical Industries, Ltd.|Chemically modified protein with polyethyleneglycol|
GB8705477D0|1987-03-09|1987-04-15|Carlton Med Prod|Drug delivery systems|
US4816444A|1987-07-10|1989-03-28|Arizona Board Of Regents, Arizona State University|Cell growth inhibitory substance|
US4975278A|1988-02-26|1990-12-04|Bristol-Myers Company|Antibody-enzyme conjugates in combination with prodrugs for the delivery of cytotoxic agents to tumor cells|
JP2783294B2|1988-09-23|1998-08-06|カイロンコーポレーション|Cell culture media for enhanced cell growth, culture life and product expression|
US5076973A|1988-10-24|1991-12-31|Arizona Board Of Regents|Synthesis of dolastatin 3|
US4978744A|1989-01-27|1990-12-18|Arizona Board Of Regents|Synthesis of dolastatin 10|
FR2646437B1|1989-04-28|1991-08-30|Transgene Sa|NOVEL DNA SEQUENCES, THEIR APPLICATION AS A SEQUENCE ENCODING A SIGNAL PEPTIDE FOR THE SECRETION OF MATURE PROTEINS BY RECOMBINANT YEASTS, EXPRESSION CASSETTES, PROCESSED YEASTS AND PROCESS FOR PREPARING THE SAME|
US4879278A|1989-05-16|1989-11-07|Arizona Board Of Regents|Isolation and structural elucidation of the cytostatic linear depsipeptide dolastatin 15|
US4986988A|1989-05-18|1991-01-22|Arizona Board Of Regents|Isolation and structural elucidation of the cytostatic linear depsipeptides dolastatin 13 and dehydrodolastatin 13|
EP0402226A1|1989-06-06|1990-12-12|Institut National De La Recherche Agronomique|Transformation vectors for yeast yarrowia|
US5013556A|1989-10-20|1991-05-07|Liposome Technology, Inc.|Liposomes with enhanced circulation time|
US5138036A|1989-11-13|1992-08-11|Arizona Board Of Regents Acting On Behalf Of Arizona State University|Isolation and structural elucidation of the cytostatic cyclodepsipeptide dolastatin 14|
US5122469A|1990-10-03|1992-06-16|Genentech, Inc.|Method for culturing Chinese hamster ovary cells to improve production of recombinant proteins|
WO1994004679A1|1991-06-14|1994-03-03|Genentech, Inc.|Method for making humanized antibodies|
DE69233254T2|1991-06-14|2004-09-16|Genentech, Inc., South San Francisco|Humanized Heregulin antibody|
DK0656789T3|1992-08-21|1998-08-24|Genentech Inc|Method of treating an LFA-1-mediated disorder.|
DE69329503T2|1992-11-13|2001-05-03|Idec Pharma Corp|Therapeutic use of chimeric and labeled antibodies directed against a differentiation antigen, the expression of which is restricted to human B lymphocyte, for the treatment of B cell lymphoma|
US5635483A|1992-12-03|1997-06-03|Arizona Board Of Regents Acting On Behalf Of Arizona State University|Tumor inhibiting tetrapeptide bearing modified phenethyl amides|
US6034065A|1992-12-03|2000-03-07|Arizona Board Of Regents|Elucidation and synthesis of antineoplastic tetrapeptide phenethylamides of dolastatin 10|
US5410024A|1993-01-21|1995-04-25|Arizona Board Of Regents Acting On Behalf Of Arizona State University|Human cancer inhibitory pentapeptide amides|
US5780588A|1993-01-26|1998-07-14|Arizona Board Of Regents|Elucidation and synthesis of selected pentapeptides|
US5521284A|1994-08-01|1996-05-28|Arizona Board Of Regents Acting On Behalf Of Arizona State University|Human cancer inhibitory pentapeptide amides and esters|
US5530097A|1994-08-01|1996-06-25|Arizona Board Of Regents Acting On Behalf Of Arizona State University|Human cancer inhibitory peptide amides|
US5504191A|1994-08-01|1996-04-02|Arizona Board Of Regents Acting On Behalf Of Arizona State University|Human cancer inhibitory pentapeptide methyl esters|
US5554725A|1994-09-14|1996-09-10|Arizona Board Of Regents Acting On Behalf Of Arizona State University|Synthesis of dolastatin 15|
US5599902A|1994-11-10|1997-02-04|Arizona Board Of Regents Acting On Behalf Of Arizona State University|Cancer inhibitory peptides|
US5663149A|1994-12-13|1997-09-02|Arizona Board Of Regents Acting On Behalf Of Arizona State University|Human cancer inhibitory pentapeptide heterocyclic and halophenyl amides|
US6096871A|1995-04-14|2000-08-01|Genentech, Inc.|Polypeptides altered to contain an epitope from the Fc region of an IgG molecule for increased half-life|
US6130237A|1996-09-12|2000-10-10|Cancer Research Campaign Technology Limited|Condensed N-aclyindoles as antitumor agents|
US6037454A|1996-11-27|2000-03-14|Genentech, Inc.|Humanized anti-CD11a antibodies|
JP2002513402A|1997-02-25|2002-05-08|アリゾナボードオブリーゼンツ|Isolation and structure determination of the cytostatic linear and cyclic depsipeptides dolastatin 16, dolastatin 17, and dolastatin 18|
US5888809A|1997-05-01|1999-03-30|Icos Corporation|Hamster EF-1α transcriptional regulatory DNA|
US6051228A|1998-02-19|2000-04-18|Bristol-Myers Squibb Co.|Antibodies against human CD40|
US6323315B1|1999-09-10|2001-11-27|Basf Aktiengesellschaft|Dolastatin peptides|
WO2001024823A1|1999-10-04|2001-04-12|Chiron Corporation|Cd40 antagonist for treating psoriasis|
DK1391464T3|2001-04-27|2008-01-14|Kirin Pharma Kk|Anti-CD40 monoclonal antibody|
US20030059427A1|2000-04-28|2003-03-27|Force Walker R.|Isolation and characterization of highly active anti-CD40 antibody|
US7256257B2|2001-04-30|2007-08-14|Seattle Genetics, Inc.|Pentapeptide compounds and uses related thereto|
US6884869B2|2001-04-30|2005-04-26|Seattle Genetics, Inc.|Pentapeptide compounds and uses related thereto|
AR039067A1|2001-11-09|2005-02-09|Pfizer Prod Inc|ANTIBODIES FOR CD40|
AU2003217912A1|2002-03-01|2003-09-16|Xencor|Antibody optimization|
US8277810B2|2003-11-04|2012-10-02|Novartis Vaccines & Diagnostics, Inc.|Antagonist anti-CD40 antibodies|
PT2489364E|2003-11-06|2015-04-16|Seattle Genetics Inc|Monomethylvaline compounds conjugated to antibodies|
CA2551008C|2003-12-25|2013-10-01|Kirin Beer Kabushiki Kaisha|Anti-cd40 antibody mutants|
BRPI0510317A|2004-04-27|2007-10-16|Novartis Vaccines & Diagnostic|antagonist anti-cd40 monoclonal antibodies and methods for their use|
CN101237882B|2005-05-26|2012-08-15|西雅图基因公司|Humanized anti-CD40 antibodies and their methods of use|
CN101426817B|2006-04-21|2013-07-10|诺华有限公司|Antagonist anti-cd40 antibody pharmaceutical compositions|
EP1854810A1|2006-05-09|2007-11-14|PanGenetics B.V.|Deimmunized antagonistic anti-human CD40 monoclonal antibody from the ch5D12 antibody|
MY166021A|2007-03-22|2018-05-21|Biogen Ma Inc|Binding proteins,including antibodies,antibody derivatives and antibody fragments,that specifically bind cd154 and uses thereof|
HUE049849T2|2010-03-31|2020-10-28|Boehringer Ingelheim Int|Anti-cd40 antibodies|
CN113461819A|2011-04-29|2021-10-01|埃派斯进有限公司|anti-CD 40 antibodies and methods of use thereof|
US20140004131A1|2012-05-04|2014-01-02|Novartis Ag|Antibody formulation|HUE049849T2|2010-03-31|2020-10-28|Boehringer Ingelheim Int|Anti-cd40 antibodies|
WO2012048113A2|2010-10-07|2012-04-12|The General Hospital Corporation|Biomarkers of cancer|
HUE044591T2|2011-03-11|2019-11-28|Beth Israel Deaconess Medical Ct Inc|Anti-cd40 antibodies and uses thereof|
AU2012245309C1|2011-04-21|2016-07-21|Bristol-Myers Squibb Company|Antibody polypeptides that antagonize CD40|
CN113461819A|2011-04-29|2021-10-01|埃派斯进有限公司|anti-CD 40 antibodies and methods of use thereof|
MX357231B|2011-10-13|2018-07-02|Squibb Bristol Myers Co|Antibody polypeptides that antagonize cd40l.|
EP3925977A1|2012-10-30|2021-12-22|Apexigen, Inc.|Anti-cd40 antibodies and methods of use|
WO2014200349A1|2013-06-13|2014-12-18|Fast Forward Pharmaceutical B.V.|Cd40 signalling inhibitor and a further compound, wherein the further compound is a bile acid, a bile acid derivative, an tgr5-receptor agonist, an fxr agonist or a combination thereof, for the treatment of chronic inflammation, and the prevention of gastrointestinal cancer or fibrosis.|
GB201311487D0|2013-06-27|2013-08-14|Alligator Bioscience Ab|Bispecific molecules|
GB201322583D0|2013-12-19|2014-02-05|Alligator Bioscience Ab|Antibodies|
EP3113796A1|2014-03-07|2017-01-11|Bristol-Myers Squibb Company|Method of using antibody polypeptides that antagonize cd40 to treat ibd|
US20170233485A1|2014-08-18|2017-08-17|Biogen Ma Inc.|Anti-cd40 antibodies and uses thereof|
TWI595006B|2014-12-09|2017-08-11|禮納特神經系統科學公司|Anti-pd-1 antibodies and methods of use thereof|
RU2715597C2|2015-05-29|2020-03-02|Эббви Инк.|Antibodies to cd40 and methods of use thereof|
JP2018529661A|2015-09-01|2018-10-11|ベーリンガー インゲルハイム インターナショナル ゲゼルシャフト ミット ベシュレンクテル ハフツング|Use of anti-CD40 antibodies for the treatment of lupus nephritis|
SI3307322T1|2015-09-04|2021-08-31|Primatope Therapeutics Inc.|Humanized anti-cd40 antibodies and uses thereof|
BR112018006251A2|2015-09-30|2018-10-16|Janssen Biotech Inc|antagonist antibodies that specifically bind to human cd40 and methods of use|
EP3471754A1|2016-06-20|2019-04-24|Kymab Limited|Anti-pd-l1 antibodies|
EP3493845A4|2016-08-03|2020-04-15|The Board of Trustees of the Leland Stanford Junior University|Disrupting fc receptor engagement on macrophages enhances efficacy of anti-sirpalpha antibody therapy|
BR112019005292A2|2016-09-21|2019-09-03|Nextcure Inc|siglec-15 antibodies and methods of use.|
US20200061162A1|2016-11-09|2020-02-27|The Brigham And Women`S Hospital, Inc.|Restoration of tumor suppression using mrna-based delivery system|
EP3589324A1|2017-03-03|2020-01-08|Janssen Biotech, Inc.|Co-therapy comprising a small molecule csf-1r inhibitor and an agonistic antibody that specifically binds cd40 for the treatment of cancer|
EP3600416A1|2017-03-30|2020-02-05|Progenity, Inc.|Treatment of a disease of the gastrointestinal tract with an immune modulatory agent released using an ingestible device|
AR111830A1|2017-05-25|2019-08-21|Squibb Bristol Myers Co|MONOCLONAL ANTI-BODIES ANTAGONISTS AGAINST CD40 AND ITS USES|
EP3632933A4|2017-06-01|2021-03-03|Seoul National University R & DB Foundation|Novel anti-cd40 antibodies and use thereof|
MX2019014375A|2017-06-01|2020-01-23|Jiangsu Hengrui Medicine Co|Anti-cd40 antibody, antigen binding fragment thereof and medical use thereof.|
CN108503708B|2017-09-01|2021-07-30|北京智仁美博生物科技有限公司|Anti-human CD47 antibodies and uses thereof|
TWI741216B|2017-09-19|2021-10-01|臻崴生物科技有限公司|Monoclonal antibody or antigen-binding fragment for specifically inhibiting or alleviate the binding of ptx3 and ptx3 receptor and use of the same|
AU2018361743A1|2017-11-03|2020-04-09|Novartis Ag|Anti-CD40 antibodies for use in treatment of Sjogren's syndrome|
CA3090322A1|2018-02-12|2019-08-15|Diabetes-Free, Inc.|Improved antagonistic anti-human cd40 monoclonal antibodies|
BR112020020277A2|2018-04-13|2021-01-19|Novartis Ag|ANTI-CD40 ANTIBODIES FOR USE IN PREVENTION OF GRAFT REJECTION|
MA52889A|2018-06-15|2021-04-21|Flagship Pioneering Innovations V Inc|INCREASED IMMUNE ACTIVITY BY MODULATION OF POST-CELLULAR SIGNALING FACTORS|
WO2019246317A1|2018-06-20|2019-12-26|Progenity, Inc.|Treatment of a disease or condition in a tissue originating from the endoderm|
WO2019246312A1|2018-06-20|2019-12-26|Progenity, Inc.|Treatment of a disease of the gastrointestinal tract with an immunomodulator|
EA202190094A1|2018-06-29|2021-04-21|Бёрингер Ингельхайм Интернациональ Гмбх|ANTIBODIES AGAINST CD40 FOR USE IN THE TREATMENT OF AUTOIMMUNE DISEASE|
WO2020070288A1|2018-10-05|2020-04-09|INSERM |Methods and systems for controlling the agonistic properties of antibody variable domains by light|
WO2020102454A1|2018-11-13|2020-05-22|Regents Of The University Of Minnesota|Cd40 targeted peptides and uses thereof|
EP3883636A1|2018-11-19|2021-09-29|Progenity, Inc.|Ingestible device for delivery of therapeutic agent to the gastrointestinal tract|
AR117091A1|2018-11-19|2021-07-07|Squibb Bristol Myers Co|MONOCLONAL ANTIBODIES ANTAGONISTS AGAINST CD40 AND THEIR USES|
US20220025060A1|2018-11-30|2022-01-27|Jiangsu Hengrui Medicine Co., Ltd.|Anti-cd40 antibody, antigen binding fragmentand pharmaceutical use thereof|
CN112839961A|2018-11-30|2021-05-25|江苏恒瑞医药股份有限公司|CD40 antibody pharmaceutical composition and application thereof|
KR20210114964A|2019-01-11|2021-09-24|노파르티스 아게|Anti-CD40 antibody for use in the treatment of rhinitis suppurative|
WO2020227159A2|2019-05-03|2020-11-12|Flagship Pioneering Innovations V, Inc.|Methods of modulating immune activity|
WO2020225736A1|2019-05-08|2020-11-12|Novartis Ag|Anti-cd40 antibodies for use in treatment of t1dm and insulitis|
WO2020253722A1|2019-06-17|2020-12-24|EucureBiopharma Co., Ltd|Anti-cd40 antibodies and uses thereof|
WO2021048595A1|2019-09-11|2021-03-18|Novartis Ag|A method for preventing human virus associated disorders in patients|
US20210163606A1|2019-10-03|2021-06-03|Atyr Pharma, Inc.|Compositions and methods comprising anti-nrp2 antibodies|
EP3870261A1|2019-12-13|2021-09-01|Progenity, Inc.|Ingestible device for delivery of therapeutic agent to the gastrointestinal tract|
WO2021127217A1|2019-12-17|2021-06-24|Flagship Pioneering Innovations V, Inc.|Combination anti-cancer therapies with inducers of iron-dependent cellular disassembly|
WO2021142445A1|2020-01-10|2021-07-15|Rhode Island Hospital|Antibodies to pfgarp kill plasmodium falciparum malaria parasites and protect against infection and severe disease|
WO2021236546A1|2020-05-18|2021-11-25|Bristol-Myers Squibb Company|Antibody variants with improved pharmacokinetic properties|
WO2022006179A1|2020-06-29|2022-01-06|Flagship Pioneering Innovations V, Inc.|Viruses engineered to promote thanotransmission and their use in treating cancer|
法律状态:
2018-01-23| B07D| Technical examination (opinion) related to article 229 of industrial property law [chapter 7.4 patent gazette]|
2018-04-10| B06F| Objections, documents and/or translations needed after an examination request according [chapter 6.6 patent gazette]|
2019-07-02| B07E| Notification of approval relating to section 229 industrial property law [chapter 7.5 patent gazette]|Free format text: NOTIFICACAO DE ANUENCIA RELACIONADA COM O ART 229 DA LPI |
2019-09-03| B06U| Preliminary requirement: requests with searches performed by other patent offices: procedure suspended [chapter 6.21 patent gazette]|
2020-09-29| B06A| Patent application procedure suspended [chapter 6.1 patent gazette]|
2020-12-15| B09A| Decision: intention to grant [chapter 9.1 patent gazette]|
2021-02-17| B16A| Patent or certificate of addition of invention granted [chapter 16.1 patent gazette]|Free format text: PRAZO DE VALIDADE: 20 (VINTE) ANOS CONTADOS A PARTIR DE 30/03/2011, OBSERVADAS AS CONDICOES LEGAIS. |
优先权:
申请号 | 申请日 | 专利标题
US31957410P| true| 2010-03-31|2010-03-31|
US61/319,574|2010-03-31|
PCT/US2011/030427|WO2011123489A2|2010-03-31|2011-03-30|Anti-cd40 antibodies|
[返回顶部]