AU2007242919B8 - Therapy of autoimmune disease in a patient with an inadequate response to a TNF-alpha inhibitor - Google Patents

Therapy of autoimmune disease in a patient with an inadequate response to a TNF-alpha inhibitor Download PDF

Info

Publication number
AU2007242919B8
AU2007242919B8 AU2007242919A AU2007242919A AU2007242919B8 AU 2007242919 B8 AU2007242919 B8 AU 2007242919B8 AU 2007242919 A AU2007242919 A AU 2007242919A AU 2007242919 A AU2007242919 A AU 2007242919A AU 2007242919 B8 AU2007242919 B8 AU 2007242919B8
Authority
AU
Australia
Prior art keywords
antibody
disease
antibodies
syndrome
autoimmune
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Expired
Application number
AU2007242919A
Other versions
AU2007242919B2 (en
AU2007242919B9 (en
AU2007242919A1 (en
Inventor
Mark Benyunes
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
F Hoffmann La Roche AG
Original Assignee
Genentech Inc
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Family has litigation
First worldwide family litigation filed litigation Critical https://patents.darts-ip.com/?family=38961380&utm_source=google_patent&utm_medium=platform_link&utm_campaign=public_patent_search&patent=AU2007242919(B8) "Global patent litigation dataset” by Darts-ip is licensed under a Creative Commons Attribution 4.0 International License.
Priority claimed from AU2004229376A external-priority patent/AU2004229376B2/en
Application filed by Genentech Inc filed Critical Genentech Inc
Priority to AU2007242919A priority Critical patent/AU2007242919B8/en
Publication of AU2007242919A1 publication Critical patent/AU2007242919A1/en
Publication of AU2007242919B2 publication Critical patent/AU2007242919B2/en
Application granted granted Critical
Publication of AU2007242919B9 publication Critical patent/AU2007242919B9/en
Publication of AU2007242919B8 publication Critical patent/AU2007242919B8/en
Assigned to F. HOFFMANN-LA ROCHE AG reassignment F. HOFFMANN-LA ROCHE AG Request for Assignment Assignors: GENENTECH, INC.
Anticipated expiration legal-status Critical
Expired legal-status Critical Current

Links

Abstract

THERAPY OF AUTOIMMUNE DISEASE IN A PATIENT WITH AN INADEQUATE RESPONSE TO A TNF-ALPHA INHIBITOR 5 The present application describes therapy with antagonists which bind to B cell surface markers, such as CD20. In particular, the application describes the use of such antagonists to treat autoimmune disease in a mammal who experiences an inadequate response to a TNFa-inhibitor.

Description

AUSTRALIA Patents Act 1990 COMPLETE SPECIFICATION Standard Patent Applicant: GENENTECH, INC. Invention Title: THERAPY OF AUTOIMMUNE DISEASE IN A PATIENT WITH AN INADEQUATE RESPONSE TO A TNF-ALPHA INHIBITOR The following statement is a full description of this invention, including the best method for performing it known to me/us: THERAPY OF AUTOIMMUNE DISEASE IN A PATIENT WITH AN INADEQUATE RESPONSE TO A TNF-ALPHA INHIBITOR Field of the Invention 5 The present invention concerns therapy with antagonists which bind to B cell surface markers, such as CD20. In particular, the invention concerns the use of such antagonists to treat autoimmune disease in a mammal who experiences an inadequate response to a TNFa-inhibitor. 10 Background of the Invention All references, including any patents or patent application, cited in this specification are hereby incorporated by reference. No admission is made that any reference constitutes prior art. The discussion of the references states what their authors assert, and the applicants reserve the right to challenge the accuracy and pertinency of 15 the cited documents. It will be clearly understood that, although a number of prior art publications are referred to herein, this reference does not constitute an admission that any of these documents form part of the common general knowledge in the art, in Australia or in any other country. Lymphocytes are one of many types of white blood cells produced in the bone 20 marrow during the process of hematopoiesis. There are two major populations of lymphocytes: B lymphocytes (B cells) and T lymphocytes (T cells). The lymphocytes of particular interest herein are B cells. B cells mature within the bone marrow and leave the marrow expressing an antigen-binding antibody on their cell surface. When a naive B cell first encounters the 25 antigen for which its membrane-bound antibody is specific, the cell begins to divide rapidly and its progeny differentiate into memory B cells and effector cells called "plasma cells". Memory B cells have a longer life span and continue to express membrane-bound antibody with the same specificity as the original parent cell. Plasma cells do not produce membrane-bound antibody but instead produce the antibody in a 30 form that can be secreted. Secreted antibodies are the major effector molecule of humoral immunity. The CD20 antigen (also called human B-lymphocyte-restricted differentiation antigen, Bp35) is a hydrophobic transmembrane protein with a molecular weight of approximately 35 kD located on pre-B and mature B lymphocytes (Valentine et al. J. 35 Biol. Chem. 264(19):11282-11287 (1989); and Einfeld et al. EMBO J. 7(3):711-717 (1988)). The antigen is also expressed on greater than 90% of B cell non-Hodgkin's lymphomas (NHL) (Anderson et al. Blood 63(6):1424-1433 (1984)), but is not found on -2hematopoietic stem cells, pro-B cells, normal plasma cells or other normal tissues (Tedder et al. J. Immunol. 135(2):973-979 (1985)). CD20 regulates an early step(s) in the activation process for cell cycle initiation and differentiation (Tedder et al., supra) and possibly functions as a calcium ion channel (Tedder et al. J Cell. Biochem. 5 14D:195 (1990)). Given the expression of CD20 in B cell lymphomas, this antigen can serve as a candidate for "targeting" of such lymphomas. In essence, such targeting can be generalized as follows: antibodies specific to the CD20 surface antigen of B cells are administered to a patient. These anti-CD20 antibodies specifically bind to the CD20 10 antigen of (ostensibly) both normal and malignant B cells; the antibody bound to the CD20 surface antigen may lead to the destruction and depletion of neoplastic B cells. Additionally, chemical agents or radioactive labels having the potential to destroy the tumor can be conjugated to the anti-CD20 antibody such that the agent is specifically "delivered" to the neoplastic B cells. Irrespective of the approach, a primary goal is to 15 destroy the tumor; the specific approach can be determined by the particular anti-CD20 antibody which is utilized and, thus, the available approaches to targeting the CD20 antigen can vary considerably. CD19 is another antigen that is expressed on the surface of cells of the B lineage. Like CD20, CD19 is found on cells throughout differentiation of the lineage 20 from the stem cell stage up to a point just prior to terminal differentiation into plasma cells (Nadler, L. Lymphocyte Typing 112: 3-37 and Appendix, Renling et al. eds. (1986) by Springer Verlag). Unlike CD20 however, antibody binding to CD19 causes internalization of the CD19 antigen. CD19 antigen is identified by the HD237-CD19 antibody (also called the "B4" antibody) (Kiesel et al. Leukemia Research 11, 12: 1119 25 (1987)), among others. The CD19 antigen is present on 4-8% of peripheral blood mononuclear cells and on greater than 90% of B cells isolated from peripheral blood, spleen, lymph node or tonsil. CD19 is not detected on peripheral blood T cells, monocytes or granulocytes. Virtually all non-T cell acute lymphoblastic leukemias (ALL), B cell chronic lymphocytic leukemias (CLL) and B cell lymphomas express 30 CD19 detectable by the antibody B4 (Nadler et al. J. Immunol. 131:244 (1983); and Nadler et al. in Progress in Hematology Vol. XII pp. 187-206. Brown, E. ed. (1981) by Grune & Stratton, Inc). Additional antibodies which recognize differentiation stage-specific antigens expressed by cells of the B cell lineage have been identified. Among these are the B2 35 antibody directed against the CD21 antigen; B3 antibody directed against the CD22 antigen; and the J5 antibody directed against the CD1O antigen (also called CALLA). See US Patent No. 5,595,721 issued January 21, 1997 (Kaminski et al.). -3- The rituximab (RITUXAN@) antibody is a genetically engineered chimeric murine/human monoclonal antibody directed against the CD20 antigen. Rituximab is the antibody called "C2B8" in US Patent No. 5,736,137 issued April 7, 1998 (Anderson et al.). RITUXAN@ is indicated for the treatment of patients with relapsed or refractory 5 low-grade or follicular, CD20 positive, B cell non-Hodgkin's lymphoma. In vitro mechanism of action studies have demonstrated that RITUXAN@ binds human complement and lyses lymphoid B cell lines through complement-dependent cytotoxicity (CDC) (Reff et al. Blood 83(2):435-445 (1994)). Additionally, it has significant activity in assays for antibody-dependent cellular cytotoxicity (ADCC). 10 More recently, RITUXAN@ has been shown to have anti-proliferative effects in tritiated thymidine incorporation assays and to induce apoptosis directly, while other anti-CD 19 and CD20 antibodies do not (Maloney et al. Blood 88(10):637a (1996)). Synergy between RITUXAN@ and chemotherapies and toxins has also been observed experimentally. In particular, RITUXAN@ sensitizes drug-resistant human B cell 15 lymphoma cell lines to the cytotoxic effects of doxorubicin, CDDP, VP-16, diphtheria toxin and ricin (Demidem et al. Cancer Chemotherapy & Radiopharmaceuticals 12(3):177-186 (1997)). In vivo preclinical studies have shown that RITUXAN@ depletes B cells from the peripheral blood, lymph nodes, and bone marrow of cynomolgus monkeys, presumably through complement and cell-mediated processes 20 (Reff et al. Blood 83(2):435-445 (1994)). Patents and patent publications concerning CD20 antibodies include US Patent Nos. 5,776,456, 5,736,137, 6,399,061, and 5,843,439, as well as US patent appln nos. US 2002/0197255A1 and US 2003/0021781A1 (Anderson et al.); US Patent No. 6,455,043B1 and WOOO/09160 (Grillo-Lopez, A.); W00/27428 (Grillo-Lopez and 25 White); WOOO/27433 (Grillo-Lopez and Leonard); WOOO/44788 (Braslawsky et al.); WOO1/10462 (Rastetter, W.); WOO1/10461 (Rastetter and White); WOOI/10460 (White and Grillo-Lopez); US appln no. US2002/0006404 and W002/04021 (Hanna and Hariharan); US appln no. US2002/0012665 Al and WO01/74388 (Hanna, N.); US appln no. US2002/0009444A1, and WOO1/80884 (Grillo-Lopez, A.); WO01/97858 30 (White, C.); US appln no. US2002/0128488AI and WOO2/34790 (Reff, M.);WO02/060955 (Braslawsky et al.);W02/096948 (Braslawsky et al.);WO02/079255 (Reff and Davies); US Patent No. 6,171,586B1, and W098/56418 (Lam et al.); W098/58964 (Raju, S.); W099/22764 (Raju, S.);W099/51642, US Patent No. 6,194,551B1, US Patent No. 6,242,195B1, US Patent No. 6,528,624B1 and US Patent 35 No. 6,538,124 (Idusogie et al.); WOOO/42072 (Presta, L.); WOOO/67796 (Curd et al.); WOO 1/03734 (Grillo-Lopez et al.); US appln no. US 2002/0004587AI and WO01/77342 (Miller and Presta); US appln no. US2002/0197256 (Grewal, I.); US -4- Patent Nos. 6,090,365B 1, 6,287,537B 1, 6,015,542, 5,843,398, and 5,595,721, (Kaminski et al.); US Patent Nos. 5,500,362, 5,677,180, 5,721,108, and 6,120,767 (Robinson et al.); US Pat No. 6,410,391B1 (Raubitschek et al.); US Patent No. 6,224,866B1 and W00/20864 (Barbera-Guillem, E.); WOO1/13945 (Barbera-Guillem, 5 E.); WOOO/67795 (Goldenberg); WOOO/74718 (Goldenberg and Hansen); WOOO/76542 (Golay et al.);WOO1/72333 (Wolin and Rosenblatt); US Patent No. 6,368,596B1 (Ghetie et al.); US Appln no. US2002/0041847A1, (Goldenberg, D.); US Appln no. US2003/0026801AI (Weiner and Hartmann); W002/102312 (Engleman, E.), each of which is expressly incorporated herein by reference. See, also, US Patent No. 10 5,849,898 and EP appln no. 330,191 (Seed et al.); US Patent No. 4,861,579 and EP332,865A2 (Meyer and Weiss); and W095/03770 (Bhat et al.). Publications concerning therapy with Rituximab include: Perotta and Abuel "Response of chronic relapsing ITP of 10 years duration to Rituximab" Abstract # 3360 Blood 10(l)(part 1-2): p. 88B (1998); Stashi et al. "Rituximab chimeric anti-CD20 15 monoclonal antibody treatment for adults with chronic idiopathic thrombocytopenic purpura" Blood 98(4):952-957 (2001); Matthews, R. "Medical Heretics" New Scientist (7 April, 2001); Leandro et al. "Clinical outcome in 22 patients with rheumatoid arthritis treated with B lymphocyte depletion" Ann Rheum Dis 61:833-888 (2002); Leandro et al. "Lymphocyte depletion in thrumatoid arthritis: early evidence for safety, 20 efficacy and dose response. Arthritis and Rheumatism 44(9): S370 (2001); Leandro et al. "An open study of B lymphocyte depletion in systemic lupus erythematosus", Arthritis & Rheumatism 46(1):2673-2677 (2002); Edwards and Cambridge "Sustained improvement in rheumatoid arthritis following a protocol designed to deplete B lymphocytes" Rhematology 40:205-211 (2001); Edwards et al. "B-lymphocyte 25 depletion therapy in rheumatoid arthritis and other autoimmune disorders" Biochem. Soc. Trans. 30(4):824-828 (2002); Edwards et al. "Efficacy and safety of Rituximab, a B-cell targeted chimeric monoclonal antibody: A randomized, placebo controlled trial in patients with rheumatoid arthritis. Arthritis and Rheumatism 46(9): S197 (2002); Levine and Pestronk "IgM antibody-related polyneuropathies: B-cell depletion chemotherapy 30 using Rituximab" Neurology 52: 1701-1704 (1999); DeVita et al. "Efficacy of selective B cell blockade in the treatment of rheumatoid arthritis" Arthritis & Rheum 46:2029 2033 (2002); Hidashida et al. "Treatment of DMARD-Refractory rheumatoid arthritis with rituximab." Presented at the Annual Scientific Meeting of the American College of Rheumatology; Oct 24-29; Ne Orleans, LA 2002; Tuscano, J. "Successful treatment of 35 Infliximab-refractory rheumatoid arthritis with rituximab" Presented at the Annual Scientific Meeting of the American College of Rheumatology; Oct 24-29; New Orleans, LA 2002. -5- Rheumatoid arthritis (RA) is an autoimmune disorder of unknown etiology. Most RA patients suffer a chronic course of disease that, even with therapy, may result in progressive joint destruction, deformity, disability and even premature death. More than 9 million physician visits and more than 250,000 hospitalizations per year result 5 from RA. The goals of RA therapy are to prevent or control joint damage, prevent loss of function and decrease pain. Initial therapy of RA usually involves administration of one or more of the following drugs: nonsteroidal antiinflammatory drugs (NSAIDs), glucocorticoid (via joint injection), and low-dose prednisone. See "Guidelines for the management of rheumatoid arthritis" Arthritis & Rheumatism 46(2): 328-346 (February, 10 2002). The majority of patients with newly diagnosed RA are started with disease modifying antirheumatic drug (DMARD) therapy within 3 months of diagnosis. DMARDs commonly used in RA are hydroxycloroquine, sulfasalazine, methotrexate, leflunomide, etanercept, infliximab (plus oral and subcutaneous methrotrexate), azathioprine, D-penicillamine, Gold (oral), Gold (intramuscular), minocycline, 15 cyclosporine, Staphylococcal protein A immunoadsorption. Because the body produces tumor necrosis factor alpha (TNFa) during RA, TNFa inhibitors have used for therapy of that disease. Etanercept (ENBREL@) is an injectable drug approved in the US for therapy of active RA. Etanercept binds to TNFa and serves to remove most TNFa from joints and 20 blood, thereby preventing TNFa from promoting inflammation and other symptoms of rheumatoid arthritis. Etanercept is an "immunoadhesin" fusion protein consisting of the extracellular ligand binding portion of the human 75 kD (p 7 5 ) tumor necrosis factor receptor (TNFR) linked to the Fc portion of a human IgGl. The drug has been associated with negative side effects including serious infections and sepsis, and 25 nervous system disorders such as multiple sclerosis (MS). See, e.g., www.remicade-infliximab.com/pages/enbrelembrel.html Infliximab, sold under the trade name REMICADE@, is an immune-suppressing drug prescribed to treat RA and Crohn's disease. Infliximab is a chimeric monoclonal antibody that binds to TNFa and reduces inflammation in the body by targeting and 30 binding to TNFa which produces inflammation. Infliximab has been linked to fatal reactions such as heart failure and infections including tuberculosis as well as demyelination resulting in MS. In December 2002, Abbott Laboratories received FDA approval to market adalimumab (HUMIRA T M ), previously known as D2E7. Adalimumab is a human 35 monoclonal antibody that binds to TNFa and is approved for reducing the signs and symptoms and inhibiting the progression of structural damage in adults with moderately -6to severely active RA who have had insufficient response to one or more traditional disease modifying DMARDs. Summary of the Invention 5 The present invention provides, in a first aspect, a method of treating rheumatoid arthritis in a human patient who experiences an inadequate response to a TNFa-inhibitor, comprising administering to the mammal a therapeutically effective amount of an antibody which binds to CD20, wherein the antibody is administered as two intravenous doses of 1000mg. 10 The present invention provides, in a second aspect, a method of achieving a clinical response selected from the group consisting of ACR50 response at week 24, ACR70 response at week 24 and no erosive progression at week 24 and beyond, in a human rheumatoid arthritis patient who experiences an inadequate response to a TNFa inhibitor, comprising administering to the patient rituximab and methotrexate, wherein 15 rituximab is administered as two intravenous doses of 1000mg. Detailed Description of the Preferred Embodiments I. Definitions In the claims of this application and in the description of the invention, except 20 where the context requires otherwise due to express language or necessary implication, the word "comprise" or variations such as "comprises" or "comprising" is used in an inclusive sense, i.e. to specify the presence of the stated features but not to preclude the presence or addition of further features in various embodiments of the invention. For the purposes herein, "tumor necrosis factor alpha (TNFa)" refers to a human 25 TNFa molecule comprising the amino acid sequence as described in Pennica et al., Nature, 312:721 (1984) or Aggarwal et al., JBC, 260:2345 (1985). A "TNFa inhibitor" herein is an agent that inhibits, to some extent, a biological function of TNFa, generally through binding to TNFa and neutralizing its activity. Examples of TNF inhibitors specifically contemplated herein are Etanercept 30 (ENBREL@), Infliximab (REMICADE@) and Adalimumab (HUMIRA T M ). The term "inadequate response to a TNFae-inhibitor" refers to an inadequate response to previous or current treatment with a TNFa-inhibitor because of toxicity and/or inadequate efficacy. The inadequate response can be assessed by a clinician skilled in treating the disease in question. 35 The "CD20" antigen is a -35 kDa, non-glycosylated phosphoprotein found on the surface of greater than 90% of B cells from peripheral blood or lymphoid organs. CD20 is expressed during early pre-B cell development and remains until plasma cell 7 differentiation. CD20 is present on both normal B cells as well as malignant B cells. Other names for CD20 in the literature include "B-lymphocyte-restricted antigen" and "Bp35". The CD20 antigen is described in Clark et al. PNAS (USA) 82:1766 (1985), for example. 5 The term "antibody" herein is used in the broadest sense and specifically covers intact monoclonal antibodies, polyclonal antibodies, multispecific antibodies (e.g. bispecific antibodies) formed from at least two intact antibodies, and antibody fragments so long as they exhibit the desired biological activity. "Antibody fragments" comprise a portion of an intact antibody, preferably 10 comprising the antigen-binding or variable region thereof Examples of antibody fragments include Fab, Fab', F(ab') 2 , and Fv fragments; diabodies; linear antibodies; single-chain antibody molecules; and multispecific antibodies formed from antibody fragments. "Native antibodies" are usually heterotetrameric glycoproteins of about 150,000 15 daltons, composed of two identical light (L) chains and two identical heavy (H) chains. Each light chain is linked to a heavy chain by one covalent disulfide bond, while the number of disulfide linkages varies among the heavy chains of different immunoglobulin isotypes. Each heavy and light chain also has regularly spaced intrachain disulfide bridges. Each heavy chain has at one end a variable domain (VH) 20 followed by a number of constant domains. Each light chain has a variable domain at one end (VL) and a constant domain at its other end; the constant domain of the light chain is aligned with the first constant domain of the heavy chain, and the light-chain variable domain is aligned with the variable domain of the heavy chain. Particular amino acid residues are believed to form an interface between the light chain and heavy 25 chain variable domains. The term "variable" refers to the fact that certain portions of the variable domains differ extensively in sequence among antibodies and are used in the binding and specificity of each particular antibody for its particular antigen. However, the variability is not evenly distributed throughout the variable domains of antibodies. It is 30 concentrated in three segments called hypervariable regions both in the light chain and the heavy chain variable domains. The more highly conserved portions of variable domains are called the framework regions (FRs). The variable domains of native heavy and light chains each comprise four FRs, largely adopting a 0-sheet configuration, connected by three hypervariable regions, which form loops connecting, and in some 35 cases forming part of, the 0-sheet structure. The hypervariable regions in each chain are held together in close proximity by the FRs and, with the hypervariable regions from the other chain, contribute to the formation of the antigen-binding site of antibodies (see 8 Kabat et al., Sequences of Proteins of Immunological Interest, 5th Ed. Public Health Service, National Institutes of Health, Bethesda, MD. (1991)). The constant domains are not involved directly in binding an antibody to an antigen, but exhibit various effector functions, such as participation of the antibody in antibody dependent cellular 5 cytotoxicity (ADCC). Papain digestion of antibodies produces two identical antigen-binding fragments, called "Fab" fragments, each with a single antigen-binding site, and a residual "Fc" fragment, whose name reflects its ability to crystallize readily. Pepsin treatment yields an F(ab') 2 fragment that has two antigen-binding sites and is still 10 capable of cross-linking antigen. "Fv" is the minimum antibody fragment which contains a complete antigen recognition and antigen-binding site. This region consists of a dimer of one heavy chain and one light chain variable domain in tight, non-covalent association. It is in this configuration that the three hypervariable regions of each variable domain interact to 15 define an antigen-binding site on the surface of the VH-VL dimer. Collectively, the six hypervariable regions confer antigen-binding specificity to the antibody. However, even a single variable domain (or half of an Fv comprising only three hypervariable regions specific for an antigen) has the ability to recognize and bind antigen, although at a lower affinity than the entire binding site. 20 The Fab fragment also contains the constant domain of the light chain and the first constant domain (CHI) of the heavy chain. Fab' fragments differ from Fab fragments by the addition of a few residues at the carboxy terminus of the heavy chain CH1 domain including one or more cysteines from the antibody hinge region. Fab'-SH is the designation herein for Fab' in which the cysteine residue(s) of the constant 25 domains bear at least one free thiol group. F(ab') 2 antibody fragments originally were produced as pairs of Fab' fragments which have hinge cysteines between them. Other chemical couplings of antibody fragments are also known. The "light chains" of antibodies (immunoglobulins) from any vertebrate species can be assigned to one of two clearly distinct types, called kappa (K) and lambda (X), 30 based on the amino acid sequences of their constant domains. Depending on the amino acid sequence of the constant domain of their heavy chains, antibodies can be assigned to different classes. There are five major classes of intact antibodies: IgA, IgD, IgE, IgG, and IgM, and several of these may be further divided into subclasses (isotypes), e.g., IgGl, IgG2, IgG3, IgG4, IgA, and IgA2. The 35 heavy-chain constant domains that correspond to the different classes of antibodies are called o', 6, E, y, and y, respectively. The subunit structures and three-dimensional configurations of different classes of immunoglobulins are well known. 9 "Single-chain Fv" or "scFv" antibody fragments comprise the VH and VL domains of antibody, wherein these domains are present in a single polypeptide chain. Preferably, the Fv polypeptide further comprises a polypeptide linker between the VH and VL domains which enables the scFv to form the desired structure for antigen 5 binding. For a review of scFv see Plickthun in The Pharmacology of Monoclonal Antibodies, vol. 113, Rosenburg and Moore eds., Springer-Verlag, New York, pp. 269 315 (1994). The term "diabodies" refers to small antibody fragments with two antigen binding sites, which fragments comprise a heavy-chain variable domain (VH) connected 10 to a light-chain variable domain (VL) in the same polypeptide chain (VH - VL). By using a linker that is too short to allow pairing between the two domains on the same chain, the domains are forced to pair with the complementary domains of another chain and create two antigen-binding sites. Diabodies are described more fully in, for example, EP 404,097; WO 93/11161; and Hollinger et al., Proc. Natl. A cad. Sci. USA, 15 90:6444-6448 (1993). The term "monoclonal antibody" as used herein refers to an antibody obtained from a population of substantially homogeneous antibodies, i.e., the individual antibodies comprising the population are identical except for possible naturally occurring mutations that may be present in minor amounts. Monoclonal antibodies are 20 highly specific, being directed against a single antigenic site. Furthermore, in contrast to conventional (polyclonal) antibody preparations which typically include different antibodies directed against different determinants (epitopes), each monoclonal antibody is directed against a single determinant on the antigen. In addition to their specificity, the monoclonal antibodies are advantageous in that they are synthesized by the 25 hybridoma culture, uncontaminated by other immunoglobulins. The modifier "monoclonal" indicates the character of the antibody as being obtained from a substantially homogeneous population of antibodies, and is not to be construed as requiring production of the antibody by any particular method. For example, the monoclonal antibodies to be used in accordance with the present invention may be 30 made by the hybridoma method first described by Kohler et al., Nature, 256:495 (1975), or may be made by recombinant DNA methods (see, e.g., U.S. Patent No. 4,816,567). The "monoclonal antibodies" may also be isolated from phage antibody libraries using the techniques described in Clackson et al., Nature, 352:624-628 (1991) and Marks et al., J. Mol. Biol., 222:581-597 (1991), for example. 35 The monoclonal antibodies herein specifically include "chimeric" antibodies (immunoglobulins) in which a portion of the heavy and/or light chain is identical with or homologous to corresponding sequences in antibodies derived from a particular 10 species or belonging to a particular antibody class or subclass, while the remainder of the chain(s) is identical with or homologous to corresponding sequences in antibodies derived from another species or belonging to another antibody class or subclass, as well as fragments of such antibodies, so long as they exhibit the desired biological activity 5 (U.S. Patent No. 4,816,567; Morrison et al., Proc. Natl. A cad. Sci. USA, 81:6851-6855 (1984)). Chimeric antibodies of interest herein include "primatized" antibodies comprising variable domain antigen-binding sequences derived from a non-human primate (e.g. Old World Monkey, such as baboon, rhesus or cynomolgus monkey) and human constant region sequences (US Pat No. 5,693,780). 10 "Humanized" forms of non-human (e.g., murine) antibodies are chimeric antibodies that contain minimal sequence derived from non-human immunoglobulin. For the most part, humanized antibodies are human immunoglobulins (recipient antibody) in which residues from a hypervariable region of the recipient are replaced by residues from a hypervariable region of a non-human species (donor antibody) such as 15 mouse, rat, rabbit or nonhuman primate having the desired specificity, affinity, and capacity. In some instances, framework region (FR) residues of the human immunoglobulin are replaced by corresponding non-human residues. Furthermore, humanized antibodies may comprise residues that are not found in the recipient antibody or in the donor antibody. These modifications are made to further refine 20 antibody performance. In general, the humanized antibody will comprise substantially all of at least one, and typically two, variable domains, in which all or substantially all of the hypervariable loops correspond to those of a non-human immunoglobulin and all or substantially all of the FRs are those of a human immunoglobulin sequence. The humanized antibody optionally also will comprise at least a portion of an 25 immunoglobulin constant region (Fc), typically that of a human immunoglobulin. For further details, see Jones et al., Nature 321:522-525 (1986); Riechmann et al., Nature 332:323-329 (1988); and Presta, Curr. Op. Struct. Biol. 2:593-596 (1992). The term "hypervariable region" when used herein refers to the amino acid residues of an antibody which are responsible for antigen-binding. The hypervariable 30 region comprises amino acid residues from a "complementarity determining region" or "CDR" (e.g. residues 24-34 (LI), 50-56 (L2) and 89-97 (L3) in the light chain variable domain and 31-35 (H1), 50-65 (H2) and 95-102 (H3) in the heavy chain variable domain; Kabat et al., Sequences of Proteins of Immunological Interest, 5th Ed. Public Health Service, National Institutes of Health, Bethesda, MD. (1991)) and/or those 35 residues from a "hypervariable loop" (e.g. residues 26-32 (LI), 50-52 (L2) and 91-96 (L3) in the light chain variable domain and 26-32 (H1), 53-55 (H2) and 96-101 (H3) in the heavy chain variable domain; Chothia and Lesk J. Mol. Biol. 196:901-917 (1987)). l1 "Framework" or "FR" residues are those variable domain residues other than the hypervariable region residues as herein defined. An antagonist "which binds" an antigen of interest, e.g. a B cell surface marker, is one capable of binding that antigen with sufficient affinity and/or avidity such that 5 the antagonist is useful as a therapeutic agent for targeting a cell expressing the antigen. Examples of antibodies which bind the CD20 antigen include: "C2B8" which is now called "rituximab" ("RITUXAN@") (US Patent No. 5,736,137, expressly incorporated herein by reference); the yttrium-[90]-labeled 2B8 murine antibody designated "Y2B8" (US Patent No. 5,736,137, expressly incorporated herein by 10 reference); murine IgG2a "B 1" optionally labeled with 1'I to generate the " 3 1 1-B 1 antibody (BEXXAR
TM
) (US Patent No. 5,595,721, expressly incorporated herein by reference); murine monoclonal antibody "1 F5" (Press et al. Blood 69(2):584-591 (1987)); "chimeric 2H7 antibody" (US Patent No. 5,677,180, expressly incorporated herein by reference); "humanized 2H7 v16" (see below); huMax-CD20 (Genmab, 15 Denmark); AME-133 (Applied Molecular Evolution); and monoclonal antibodies L27, G28-2, 93-11B3, B-Cl or NU-B2 available from the International Leukocyte Typing Workshop (Valentine et al., In: Leukocyte Typing III (McMichael, Ed., p. 440, Oxford University Press (1987)). The terms "rituximab" or "RITUXAN@" herein refer to the genetically 20 engineered chimeric murine/human monoclonal antibody directed against the CD20 antigen and designated "C2B8" in US Patent No. 5,736,137, expressly incorporated herein by reference. The antibody is an IgG kappa immunoglobulin containing murine light and heavy chain variable region sequences and human constant region sequences. Rituximab has a binding affinity for the CD20 antigen of approximately 8.OnM. 25 Purely for the purposes herein, "humanized 2H7 v16" refers to an antibody comprising the variable light and variable heavy sequences shown below . Variable light-chain domain of hu2H7 v16 DIQMTQSPSSLSASVGDRVTITCRASSSVSYMHWYQQKPGKAPKPLIYA PSNLASGVPSRFSGSGSGTDFTLTISSLQPEDFATYYCQQWSFNPPTFGQGTKVE 30 IKR (SEQ ID NO: 1) Variable heavy-chain domain of hu2H7 v16 EVQLVESGGGLVQPGGSLRLSCAASGYTFTSYNMH WVRQAPGKGLEW VGA IYPGNGDTSYNQKFKGRFTISVDKSKNTLY LQMNSLRA EDTAVYYCARV VYYSNSYWYFDVWGQGTLVTVSS (SEQ ID NO: 2) 35 Preferably humanized 2H7 v16 comprises the light chain amino acid sequence DIQMTQSPSSLSASVGDRVTITCRASSSVSYMHWYQQKPGKAPKPLIYAPSNLA SGVPSRFSGSGSGTDFTLTISSLQPEDFATYYCQQWSFNPPTFGQGTKVEIKRTV 12 AAPSVFIFPPSDEQLKSGTASVVCLLNNFYPREAKVQWKVDNALQSGNSQESV TEQDSKDSTYSLSSTLTLSKADYEKHKVYACEVTHQGLSSPVTKSFNRGEC (SEQ ID NO: 3); and heavy chain amino acid sequence 5 EVQLVESGGGLVQPGGSLRLSCAASGYTFTSYNMHWVRQAPGKGLEWVGAIY PGNGDTSYNQKFKGRFTISVDKSKNTLYLQMNSLRAEDTAVYYCARVVYYSN SYWYFDVWGQGTLVTVSSASTKGPSVFPLAPSSKSTSGGTAA LGCLVKDYFPE PVTVSWNSGALTSGVHTFPAVLQSSG LYSLS SVVTVPSSSLGTQTYICNVNHKP SNTKVDKKVEPKSCDKTHTCPPCPAPELLGGPSVFLFPPKPKDTLMISRTPEVTC 10 VVVDVSHEDPEVKFNWYVDGVEVHNAKTKPREEQYNSTYRVVSVLTVLHQD WLNGKEYKCKVSNKALPAPIEKTISKAKGQPREPQVYTLPPSREEMTKNQVSL TCLVKGFYPSDIAVEWESNGQPENNYKTTPPVLDSDGSFFLYSKLTVDKSRWQ QGNVFSCSVMHEALHNHYTQKSLSLSPGK (SEQ ID NO: 4). "Treatment" refers to both therapeutic treatment and prophylactic or 15 preventative measures. Those in need of treatment include those already with the disease or disorder as well as those in which the disease or disorder is to be prevented. Hence, the patient may have been diagnosed as having the disease or disorder or may be predisposed or susceptible to the disease. The expression "therapeutically effective amount" refers to an amount of the 20 antagonist which is effective for preventing, ameliorating or treating rheumatoid arthritis. A description follows as to exemplary techniques for the production of an antibody that binds to CD20 used in accordance with the present invention. (i) Polyclonal antibodies 25 Polyclonal antibodies are preferably raised in animals by multiple subcutaneous (sc) or intraperitoneal (ip) injections of the relevant antigen and an adjuvant. It may be useful to conjugate the relevant antigen to a protein that is immunogenic in the species to be immunized, e.g., keyhole limpet hemocyanin, serum albumin, bovine thyroglobulin, or soybean trypsin inhibitor using a bifunctional or derivatizing agent, 30 for example, maleimidobenzoyl sulfosuccinimide ester (conjugation through cysteine residues), N-hydroxysuccinimide (through lysine residues), glutaraldehyde, succinic anhydride, SOCl 2 , or R'N=C=NR, where R and R 1 are different alkyl groups. Animals are immunized against the antigen, immunogenic conjugates, or derivatives by combining, e.g., 100 tg or 5 p4g of the protein or conjugate (for rabbits or 35 mice, respectively) with 3 volumes of Freund's complete adjuvant and injecting the solution intradermally at multiple sites. One month later the animals are boosted with 1/5 to 1/10 the original amount of peptide or conjugate in Freund's complete adjuvant 13 by subcutaneous injection at multiple sites. Seven to 14 days later the animals are bled and the serum is assayed for antibody titer. Animals are boosted until the titer plateaus. Preferably, the animal is boosted with the conjugate of the same antigen, but conjugated to a different protein and/or through a different cross-linking reagent. Conjugates also 5 can be made in recombinant cell culture as protein fusions. Also, aggregating agents such as alum are suitably used to enhance the immune response. (ii) Monoclonal antibodies Monoclonal antibodies are obtained from a population of substantially homogeneous antibodies, i.e., the individual antibodies comprising the population are 10 identical except for possible naturally occurring mutations that may be present in minor amounts. Thus, the modifier "monoclonal" indicates the character of the antibody as not being a mixture of discrete antibodies. For example, the monoclonal antibodies may be made using the hybridoma method first described by Kohler et al., Nature, 256:495 (1975), or may be made by 15 recombinant DNA methods (U.S. Patent No. 4,816,567). In the hybridoma method, a mouse or other appropriate host animal, such as a hamster, is immunized as hereinabove described to elicit lymphocytes that produce or are capable of producing antibodies that will specifically bind to the protein used for immunization. Alternatively, lymphocytes may be immunized in vitro. Lymphocytes 20 then are fused with myeloma cells using a suitable fusing agent, such as polyethylene glycol, to form a hybridoma cell (Goding, Monoclonal Antibodies: Principles and Practice, pp.59-103 (Academic Press, 1986)). The hybridoma cells thus prepared are seeded and grown in a suitable culture medium that preferably contains one or more substances that inhibit the growth or 25 survival of the unfused, parental myeloma cells. For example, if the parental myeloma cells lack the enzyme hypoxanthine guanine phosphoribosyl transferase (HGPRT or HPRT), the culture medium for the hybridomas typically will include hypoxanthine, aminopterin, and thymidine (HAT medium), which substances prevent the growth of HGPRT-deficient cells. 30 Preferred myeloma cells are those that fuse efficiently, support stable high-level production of antibody by the selected antibody-producing cells, and are sensitive to a medium such as HAT medium. Among these, preferred myeloma cell lines are murine myeloma lines, such as those derived from MOPC-21 and MPC-I I mouse tumors available from the Salk Institute Cell Distribution Center, San Diego, California USA, 35 and SP-2 or X63-Ag8-653 cells available from the American Type Culture Collection, Rockville, Maryland USA. Human myeloma and mouse-human heteromyeloma cell lines also have been described for the production of human monoclonal antibodies 14 (Kozbor, J. Immunol., 133:3001 (1984); Brodeur et al., Monoclonal Antibody Production Techniques and Applications, pp. 51-63 (Marcel Dekker, Inc., New York, 1987)). Culture medium in which hybridoma cells are growing is assayed for production 5 of monoclonal antibodies directed against the antigen. Preferably, the binding specificity of monoclonal antibodies produced by hybridoma cells is determined by immunoprecipitation or by an in vitro binding assay, such as radioimmunoassay (RIA) or enzyme-linked immunoabsorbent assay (ELISA). The binding affinity of the monoclonal antibody can, for example, be 10 determined by the Scatchard analysis of Munson et al., Anal. Biochem., 107:220 (1980). After hybridoma cells are identified that produce antibodies of the desired specificity, affinity, and/or activity, the clones may be subcloned by limiting dilution procedures and grown by standard methods (Goding, Monoclonal Antibodies: Principles and Practice, pp.59-103 (Academic Press, 1986)). Suitable culture media 15 for this purpose include, for example, D-MEM or RPMI-1640 medium. In addition, the hybridoma cells may be grown in vivo as ascites tumors in an animal. The monoclonal antibodies secreted by the subclones are suitably separated from the culture medium, ascites fluid, or serum by conventional immunoglobulin purification procedures such as, for example, protein A-Sepharose, hydroxylapatite 20 chromatography, gel electrophoresis, dialysis, or affinity chromatography. DNA encoding the monoclonal antibodies is readily isolated and sequenced using conventional procedures (e.g., by using oligonucleotide probes that are capable of binding specifically to genes encoding the heavy and light chains of murine antibodies). The hybridoma cells serve as a preferred source of such DNA. Once isolated, the DNA 25 may be placed into expression vectors, which are then transfected into host cells such as E. coli cells, simian COS cells, Chinese Hamster Ovary (CHO) cells, or myeloma cells that do not otherwise produce immunoglobulin protein, to obtain the synthesis of monoclonal antibodies in the recombinant host cells. Review articles on recombinant expression in bacteria of DNA encoding the antibody include Skerra et al., Curr. 30 Opinion in Immunol., 5:256-262 (1993) and Plickthun, Immunol. Revs., 130:151-188 (1992). In a further embodiment, antibodies or antibody fragments can be isolated from antibody phage libraries generated using the techniques described in McCafferty et al., Nature, 348:552-554 (1990). Clackson et al., Nature, 352:624-628 (1991) and Marks et 35 al., J. Mol. Biol., 222:581-597 (1991) describe the isolation of murine and human antibodies, respectively, using phage libraries. Subsequent publications describe the production of high affinity (nM range) human antibodies by chain shuffling (Marks et 15 al., BiolTechnology, 10:779-783 (1992)), as well as combinatorial infection and in vivo recombination as a strategy for constructing very large phage libraries (Waterhouse et al., Nuc. Acids. Res., 21:2265-2266 (1993)). Thus, these techniques are viable alternatives to traditional monoclonal antibody hybridoma techniques for isolation of 5 monoclonal antibodies. The DNA also may be modified, for example, by substituting the coding sequence for human heavy- and light-chain constant domains in place of the homologous murine sequences (U.S. Patent No. 4,816,567; Morrison, et al., Proc. Natl Acad. Sci. USA, 81:6851 (1984)), or by covalently joining to the immunoglobulin 10 coding sequence all or part of the coding sequence for a non-immunoglobulin polypeptide. Typically such non-immunoglobulin polypeptides are substituted for the constant domains of an antibody, or they are substituted for the variable domains of one antigen-combining site of an antibody to create a chimeric bivalent antibody comprising 15 one antigen-combining site having specificity for an antigen and another antigen combining site having specificity for a different antigen. (iii) Humanized antibodies Methods for humanizing non-human antibodies have been described in the art. Preferably, a humanized antibody has one or more amino acid residues introduced into 20 it from a source which is non-human. These non-human amino acid residues are often referred to as "import" residues, which are typically taken from an "import" variable domain. Humanization can be essentially performed following the method of Winter and co-workers (Jones et al., Nature, 321:522-525 (1986); Riechmann et al., Nature, 332:323-327 (1988); Verhoeyen et al., Science, 239:1534-1536 (1988)), by substituting 25 hypervariable region sequences for the corresponding sequences of a human antibody. Accordingly, such "humanized" antibodies are chimeric antibodies (U.S. Patent No. 4,816,567) wherein substantially less than an intact human variable domain has been substituted by the corresponding sequence from a non-human species. In practice, humanized antibodies are typically human antibodies in which some hypervariable 30 region residues and possibly some FR residues are substituted by residues from analogous sites in rodent antibodies. The choice of human variable domains, both light and heavy, to be used in making the humanized antibodies is very important to reduce antigenicity. According to the so-called "best-fit" method, the sequence of the variable domain of a rodent 35 antibody is screened against the entire library of known human variable-domain sequences. The human sequence which is closest to that of the rodent is then accepted as the human framework region (FR) for the humanized antibody (Sims et al., J. 16 Immunol., 151:2296 (1993); Chothia et al., J. Mol. Biol., 196:901 (1987)). Another method uses a particular framework region derived from the consensus sequence of all human antibodies of a particular subgroup of light or heavy chains. The same framework may be used for several different humanized antibodies (Carter et al., Proc. 5 Natl. Acad. Sci. USA, 89:4285 (1992); Presta et al., J. Immunol., 151:2623 (1993)). It is further important that antibodies be humanized with retention of high affinity for the antigen and other favorable biological properties. To achieve this goal, according to a preferred method, humanized antibodies are prepared by a process of analysis of the parental sequences and various conceptual humanized products using 10 three-dimensional models of the parental and humanized sequences. Three-dimensional immunoglobulin models are commonly available and are familiar to those skilled in the art. Computer programs are available which illustrate and display probable three dimensional conformational structures of selected candidate immunoglobulin sequences. Inspection of these displays permits analysis of the likely role of the 15 residues in the functioning of the candidate immunoglobulin sequence, i.e., the analysis of residues that influence the ability of the candidate immunoglobulin to bind its antigen. In this way, FR residues can be selected and combined from the recipient and import sequences so that the desired antibody characteristic, such as increased affinity for the target antigen(s), is achieved. In general, the hypervariable region residues are 20 directly and most substantially involved in influencing antigen binding. (iv) Human antibodies As an alternative to humanization, human antibodies can be generated. For example, it is now possible to produce transgenic animals (e.g., mice) that are capable, upon immunization, of producing a full repertoire of human antibodies in the absence of 25 endogenous immunoglobulin production. For example, it has been described that the homozygous deletion of the antibody heavy-chain joining region (JH) gene in chimeric and germ-line mutant mice results in complete inhibition of endogenous antibody production. Transfer of the human germ-line immunoglobulin gene array in such germ line mutant mice will result in the production of human antibodies upon antigen 30 challenge. See, e.g., Jakobovits et al., Proc. Natl. A cad. Sci. USA, 90:2551 (1993); Jakobovits et al., Nature, 362:255-258 (1993); Bruggermann et al., Year in Immuno., 7:33 (1993); and US Patent Nos. 5,591,669, 5,589,369 and 5,545,807. Alternatively, phage display technology (McCafferty et al., Nature 348:552-553 (1990)) can be used to produce human antibodies and antibody fragments in vitro, from 35 immunoglobulin variable (V) domain gene repertoires from unimmunized donors. According to this technique, antibody V domain genes are cloned in-frame into either a major or minor coat protein gene of a filamentous bacteriophage, such as M13 or fd, 17 and displayed as functional antibody fragments on the surface of the phage particle. Because the filamentous particle contains a single-stranded DNA copy of the phage genome, selections based on the functional properties of the antibody also result in selection of the gene encoding the antibody exhibiting those properties. Thus, the 5 phage mimics some of the properties of the B cell. Phage display can be performed in a variety of formats; for their review see, e.g., Johnson, Kevin S. and Chiswell, David J., Current Opinion in Structural Biology 3:564-571 (1993). Several sources of V-gene segments can be used for phage display. Clackson et al., Nature, 352:624-628 (1991) isolated a diverse array of anti-oxazolone antibodies from a small random combinatorial 10 library of V genes derived from the spleens of immunized mice. A repertoire of V genes from unimmunized human donors can be constructed and antibodies to a diverse array of antigens (including self-antigens) can be isolated essentially following the techniques described by Marks et al., J. Mol. Biol. 222:581-597 (1991), or Griffith et al., EMBO J. 12:725-734 (1993). See, also, US Patent Nos. 5,565,332 and 5,573,905. 1 5 Human antibodies may also be generated by in vitro activated B cells (see US Patents 5,567,610 and 5,229,275). (v) Antibodyfragments Various techniques have been developed for the production of antibody fragments. Traditionally, these fragments were derived via proteolytic digestion of 20 intact antibodies (see, e.g., Morimoto et al., Journal of Biochemical and Biophysical Methods 24:107-117 (1992) and Brennan et al., Science, 229:81 (1985)). However, these fragments can now be produced directly by recombinant host cells. For example, the antibody fragments can be isolated from the antibody phage libraries discussed above. Alternatively, Fab'-SH fragments can be directly recovered from E. coli and 25 chemically coupled to form F(ab') 2 fragments (Carter el al., Bio/Technology 10:163 167 (1992)). According to another approach, F(ab') 2 fragments can be isolated directly from recombinant host cell culture. Other techniques for the production of antibody fragments will be apparent to the skilled practitioner. In other embodiments, the antibody of choice is a single chain Fv fragment (scFv). See WO 93/16185; US Patent 30 No. 5,571,894; and US Patent No. 5,587,458. The antibody fragment may also be a "linear antibody", e.g., as described in US Patent 5,641,870 for example. Such linear antibody fragments may be monospecific or bispecific. (vi) Bispecific antibodies Bispecific antibodies are antibodies that have binding specificities for at least 35 two different epitopes. Exemplary bispecific antibodies may bind to two different epitopes of the B cell surface marker. Other such antibodies may bind a first B cell marker and further bind a second B cell surface marker. Alternatively, an anti-B cell 18 marker binding arm may be combined with an arm which binds to a triggering molecule on a leukocyte such as a T-cell receptor molecule (e.g. CD2 or CD3), or Fc receptors for IgG (FcyR), such as FcyRI (CD64), Fc'yRIl (CD32) and Fc'yRIII (CD16) so as to focus cellular defense mechanisms to the B cell. Bispecific antibodies may also be used 5 to localize cytotoxic agents to the B cell. These antibodies possess a B cell marker binding arm and an arm which binds the cytotoxic agent (e.g. saporin, anti-interferon-o' vinca alkaloid, ricin A chain, methotrexate or radioactive isotope hapten). Bispecific antibodies can be prepared as full length antibodies or antibody fragments (e.g. F(ab') 2 bispecific antibodies). 10 Methods for making bispecific antibodies are known in the art. Traditional production of full length bispecific antibodies is based on the coexpression of two immunoglobulin heavy chain-light chain pairs, where the two chains have different specificities (Millstein et al., Nature, 305:537-539 (1983)). Because of the random assortment of immunoglobulin heavy and light chains, these hybridomas (quadromas) 15 produce a potential mixture of 10 different antibody molecules, of which only one has the correct bispecific structure. Purification of the correct molecule, which is usually done by affinity chromatography steps, is rather cumbersome, and the product yields are low. Similar procedures are disclosed in WO 93/08829, and in Traunecker et al., EMBOJ., 10:3655-3659 (1991). 20 According to a different approach, antibody variable domains with the desired binding specificities (antibody-antigen combining sites) are fused to immunoglobulin constant domain sequences. The fusion preferably is with an immunoglobulin heavy chain constant domain, comprising at least part of the hinge, CH2, and CH3 regions. It is preferred to have the first heavy-chain constant region (CHI) containing the site 25 necessary for light chain binding, present in at least one of the fusions. DNAs encoding the immunoglobulin heavy chain fusions and, if desired, the immunoglobulin light chain, are inserted into separate expression vectors, and are co-transfected into a suitable host organism. This provides for great flexibility in adjusting the mutual proportions of the three polypeptide fragments in embodiments when unequal ratios of 30 the three polypeptide chains used in the construction provide the optimum yields. It is, however, possible to insert the coding sequences for two or all three polypeptide chains in one expression vector when the expression of at least two polypeptide chains in equal ratios results in high yields or when the ratios are of no particular significance. In a preferred embodiment of this approach, the bispecific antibodies are 35 composed of a hybrid immunoglobulin heavy chain with a first binding specificity in one arm, and a hybrid immunoglobulin heavy chain-light chain pair (providing a second binding specificity) in the other arm. It was found that this asymmetric structure 19 facilitates the separation of the desired bispecific compound from unwanted immunoglobulin chain combinations, as the presence of an immunoglobulin light chain in only one half of the bispecific molecule provides for a facile way of separation. This approach is disclosed in WO 94/04690. For further details of generating bispecific 5 antibodies see, for example, Suresh et al., Methods in Enzymology, 121:210 (1986). According to another approach described in US Patent No. 5,731,168, the interface between a pair of antibody molecules can be engineered to maximize the percentage of heterodimers which are recovered from recombinant cell culture. The preferred interface comprises at least a part of the CH 3 domain of an antibody constant 10 domain. In this method, one or more small amino acid side chains from the interface of the first antibody molecule are replaced with larger side chains (e.g. tyrosine or tryptophan). Compensatory "cavities" of identical or similar size to the large side chain(s) are created on the interface of the second antibody molecule by replacing large amino acid side chains with smaller ones (e.g. alanine or threonine). This provides a 15 mechanism for increasing the yield of the heterodimer over other unwanted end products such as homodimers. Bispecific antibodies include cross-linked or "heteroconjugate" antibodies. For example, one of the antibodies in the heteroconjugate can be coupled to avidin, the other to biotin. Such antibodies have, for example, been proposed to target immune 20 system cells to unwanted cells (US Patent No. 4,676,980), and for treatment of HIV infection (WO 91/00360, WO 92/200373, and EP 03089). Heteroconjugate antibodies may be made using any convenient cross-linking methods. Suitable cross-linking agents are well known in the art, and are disclosed in US Patent No. 4,676,980, along with a number of cross-linking techniques. 25 Techniques for generating bispecific antibodies from antibody fragments have also been described in the literature. For example, bispecific antibodies can be prepared using chemical linkage. Brennan et al., Science, 229: 81 (1985) describe a procedure wherein intact antibodies are proteolytically cleaved to generate F(ab') 2 fragments. These fragments are reduced in the presence of the dithiol complexing agent sodium 30 arsenite to stabilize vicinal dithiols and prevent intermolecular disulfide formation. The Fab' fragments generated are then converted to thionitrobenzoate (TNB) derivatives. One of the Fab'-TNB derivatives is then reconverted to the Fab'-thiol by reduction with mercaptoethylamine and is mixed with an equimolar amount of the other Fab'-TNB derivative to form the bispecific antibody. The bispecific antibodies produced can be 35 used as agents for the selective immobilization of enzymes. Recent progress has facilitated the direct recovery of Fab'-SH fragments from E. coli, which can be chemically coupled to form bispecific antibodies. Shalaby et al., J 20 Exp. Med., 175: 217-225 (1992) describe the production of a fully humanized bispecific antibody F(ab') 2 molecule. Each Fab' fragment was separately secreted from E. coli and subjected to directed chemical coupling in vitro to form the bispecific antibody. The bispecific antibody thus formed was able to bind to cells overexpressing the ErbB2 5 receptor and normal human T cells, as well as trigger the lytic activity of human cytotoxic lymphocytes against human breast tumor targets. Various techniques for making and isolating bispecific antibody fragments directly from recombinant cell culture have also been described. For example, bispecific antibodies have been produced using leucine zippers. Kostelny et al., J. 10 Immunol., 148(5):1547-1553 (1992). The leucine zipper peptides from the Fos and Jun proteins were linked to the Fab' portions of two different antibodies by gene fusion. The antibody homodimers were reduced at the hinge region to form monomers and then re-oxidized to form the antibody heterodimers. This method can also be utilized for the production of antibody homodimers. The "diabody" technology described by Hollinger 15 et al., Proc. Natl. A cad. Sci. USA, 90:6444-6448 (1993) has provided an alternative mechanism for making bispecific antibody fragments. The fragments comprise a heavy-chain variable domain (VH) connected to a light-chain variable domain (VL) by a linker which is too short to allow pairing between the two domains on the same chain. Accordingly, the VH and VL domains of one fragment are forced to pair with the 20 complementary VL and VH domains of another fragment, thereby forming two antigen binding sites. Another strategy for making bispecific antibody fragments by the use of single-chain Fv (sFv) dimers has also been reported. See Gruber et al., J. Immunol., 152:5368 (1994). Antibodies with more than two valencies are contemplated. For example, 25 trispecific antibodies can be prepared. Tutt et al. J. Immunol. 147: 60 (1991). II. Pharmaceutical Formulations Therapeutic formulations of the antibodies that bind CD20 used in accordance with the present invention are prepared for storage by mixing an antagonist having the desired degree of purity with optional pharmaceutically acceptable carriers, excipients 30 or stabilizers (Remington's Pharmaceutical Sciences 16th edition, Osol, A. Ed. (1980)), in the form of lyophilized formulations or aqueous solutions. Acceptable carriers, excipients, or stabilizers are nontoxic to recipients at the dosages and concentrations employed, and include buffers such as phosphate, citrate, and other organic acids; antioxidants including ascorbic acid and methionine; preservatives (such as 35 octadecyldimethylbenzyl ammonium chloride; hexamethonium chloride; benzalkonium chloride, benzethonium chloride; phenol, butyl or benzyl alcohol; alkyl parabens such as methyl or propyl paraben; catechol; resorcinol; cyclohexanol; 3-pentanol; and m 21 cresol); low molecular weight (less than about 10 residues) polypeptides; proteins, such as serum albumin, gelatin, or immunoglobulins; hydrophilic polymers such as polyvinylpyrrolidone; amino acids such as glycine, glutamine, asparagine, histidine, arginine, or lysine; monosaccharides, disaccharides, and other carbohydrates including 5 glucose, mannose, or dextrins; chelating agents such as EDTA; sugars such as sucrose, mannitol, trehalose or sorbitol; salt-forming counter-ions such as sodium; metal complexes (e.g. Zn-protein complexes); and/or non-ionic surfactants such as
TWEEN
T M , PLURONICS T M or polyethylene glycol (PEG). Exemplary anti-CD20 antibody formulations are described in W098/56418, 10 expressly incorporated herein by reference. This publication describes a liquid multidose formulation comprising 40 mg/mL rituximab, 25 mM acetate, 150 mM trehalose, 0.9% benzyl alcohol, 0.02% polysorbate 20 at pH 5.0 that has a minimum shelf life of two years storage at 2-8*C. Another anti-CD20 formulation of interest comprises 10mg/mL rituximab in 9.0 mg/mL sodium chloride, 7.35 mg/mL sodium 15 citrate dihydrate, 0.7mg/mL polysorbate 80, and Sterile Water for Injection, pH 6.5. Lyophilized formulations adapted for subcutaneous administration are described in W097/04801. Such lyophilized formulations may be reconstituted with a suitable diluent to a high protein concentration and the reconstituted formulation may be administered subcutaneously to the mammal to be treated herein. 20 The formulation herein may also contain more than one active compound as necessary for the particular indication being treated, preferably those with complementary activities that do not adversely affect each other. For example, it may be desirable to further provide a cytotoxic agent, chemotherapeutic agent, cytokine or immunosuppressive agent (e.g. one which acts on T cells, such as cyclosporin or an 25 antibody that binds T cells, e.g. one which binds LFA-1). The effective amount of such other agents depends on the amount of antagonist present in the formulation, the type of disease or disorder or treatment, and other factors discussed above. These are generally used in the same dosages and with administration routes as used hereinbefore or about from I to 99% of the heretofore employed dosages. 30 The active ingredients may also be entrapped in microcapsules prepared, for example, by coacervation techniques or by interfacial polymerization, for example, hydroxymethylcellulose or gelatin-microcapsules and poly-(methylmethacylate) microcapsules, respectively, in colloidal drug delivery systems (for example, liposomes, albumin microspheres, microemulsions, nano-particles and nanocapsules) or in 35 macroemulsions. Such techniques are disclosed in Remington's Pharmaceutical Sciences 16th edition, Osol, A. Ed. (1980). Sustained-release preparations may be prepared. Suitable examples of 22 sustained-release preparations include semipermeable matrices of solid hydrophobic polymers containing the antagonist, which matrices are in the form of shaped articles, e.g. films, or microcapsules. Examples of sustained-release matrices include polyesters, hydrogels (for example, poly(2-hydroxyethyl-methacrylate), or poly(vinylalcohol)), 5 polylactides (U.S. Pat. No. 3,773,919), copolymers of L-glutamic acid and y ethyl-L glutamate, non-degradable ethylene-vinyl acetate, degradable lactic acid-glycolic acid copolymers such as the LUPRON DEPOT T M (injectable microspheres composed of lactic acid-glycolic acid copolymer and leuprolide acetate), and poly-D-(-)-3 hydroxybutyric acid. 10 The formulations to be used for in vivo administration must be sterile. This is readily accomplished by filtration through sterile filtration membranes. III. Treatment The present invention concerns therapy of humans, with, or susceptible to, rheumatoid arthritis, who experience an inadequate response to previous or current 15 treatment with a TNFae-inhibitor. Generally, the patient to be treated herein will be identified following therapy with one or more treatments with one or more TNFa inhibitor(s) such as Etanercept (ENBREL@), Infliximab (REMICADE@) or Adalimumab (HUMIRATM), as experiencing an inadequate response to previous or current treatment with a TNFae-inhibitor because of toxicity and/or inadequate efficacy. 20 However, the invention is not limited to a prior therapy step with such a TNFa inhibitor; for instance, the patient may be considered to be prone to experience a toxicity, e.g. cardiac toxicity, with a TNFa-inhibitor before therapy therewith has begun, or the patient may be determined to be one who is unlikely to respond to therapy with a TNFc-inhibitor. 25 Generally, the patient treated herein will not be suffering from a B-cell malignancy. According to one embodiment of the invention contemplated herein, the therapeutic approach will reduce negative side effects (such as infections, heart failure and demyelination) associated with therapy with a TNFa-inhibitor. 30 The composition comprising an antibody that binds to CD20 will be formulated, dosed, and administered in a fashion consistent with good medical practice. Factors for consideration in this context include the particular disease or disorder being treated, the particular mammal being treated, the clinical condition of the individual patient, the cause of the disease or disorder, the site of delivery of the agent, the method of 35 administration, the scheduling of administration, and other factors known to medical practitioners. The therapeutically effective amount of the antagonist to be administered will be governed by such considerations. 23 As a general proposition, the therapeutically effective amount of the antibody administered parenterally per dose will be in the range of about 0.1 to 20 mg/kg of patient body weight per day, with the typical initial range of antagonist used being in the range of about 2 to 10 mg/kg. 5 The preferred antibody is RITUXAN@, which is not conjugated to a cytotoxic agent. The key factor in selecting an appropriate dose and scheduling is the result obtained, as indicated above. For example, relatively higher doses may be needed initially for the treatment of ongoing and acute diseases. To obtain the most efficacious results, depending on the disease or disorder, the antibody is administered as close to 10 the first sign, diagnosis, appearance, or occurrence of the disease or disorder as possible or during remissions of the disease or disorder. One may administer other compounds, such as cytotoxic agents, chemotherapeutic agents, immunosuppressive agents and/or cytokines with the antagonists herein. The combined administration includes coadministration, using 15 separate formulations or a single pharmaceutical formulation, and consecutive administration in either order, wherein preferably there is a time period while both (or all) active agents simultaneously exert their biological activities. For RA, CD20 antibody may be combined with any one or more of disease-modifying antirheumatic drugs (DMARDs) such as hydroxycloroquine, sulfasalazine, methotrexate, leflunomide, 20 azathioprine, D-penicillamine, Gold (oral), Gold (intramuscular), minocycline, cyclosporine, Staphylococcal protein A immunoadsorption; intravenous immunoglobulin (IVIG); nonsteroidal antiinflammatory drugs (NSAIDs); glucocorticoid (e.g. via joint injection); corticosteroid (e.g. methylprednisolone and/or prednisone); folate etc. Preferably, a TNFa-inhibitor is not administered to the 25 mammal during the period of treatment with the CD20 antagonist. Aside from administration of antibodies to the patient the present application contemplates administration of antagonists by gene therapy. Such administration of nucleic acid encoding the antagonist is encompassed by the expression "administering a therapeutically effective amount of an antagonist". See, for example, W096/07321 30 published March 14, 1996 concerning the use of gene therapy to generate intracellular antibodies. There are two major approaches to getting the nucleic acid (optionally contained in a vector) into the patient's cells; in vivo and ex vivo. For in vivo delivery the nucleic acid is injected directly into the patient, usually at the site where the antagonist is 35 required. For ex vivo treatment, the patient's cells are removed, the nucleic acid is introduced into these isolated cells and the modified cells are administered to the patient either directly or, for example, encapsulated within porous membranes which are 24 implanted into the patient (see, e.g. U.S. Patent Nos. 4,892,538 and 5,283,187). There are a variety of techniques available for introducing nucleic acids into viable cells. The techniques vary depending upon whether the nucleic acid is transferred into cultured cells in vitro, or in vivo in the cells of the intended host. Techniques suitable for the 5 transfer of nucleic acid into mammalian cells in vitro include the use of liposomes, electroporation, microinjection, cell fusion, DEAE-dextran, the calcium phosphate precipitation method, etc. A commonly used vector for ex vivo delivery of the gene is a retrovirus. The currently preferred in vivo nucleic acid transfer techniques include 10 transfection with viral vectors (such as adenovirus, Herpes simplex I virus, or adeno associated virus) and lipid-based systems (useful lipids for lipid-mediated transfer of the gene are DOTMA, DOPE and DC-Chol, for example). In some situations it is desirable to provide the nucleic acid source with an agent that targets the target cells, such as an antibody specific for a cell surface membrane protein or the target cell, a 15 ligand for a receptor on the target cell, etc. Where liposomes are employed, proteins which bind to a cell surface membrane protein associated with endocytosis may be used for targeting and/or to facilitate uptake, e.g. capsid proteins or fragments thereof tropic for a particular cell type, antibodies for proteins which undergo internalization in cycling, and proteins that target intracellular localization and enhance intracellular half 20 life. The technique of receptor-mediated endocytosis is described, for example, by Wu et al., J Biol. Chem. 262:4429-4432 (1987); and Wagner et al., Proc. NatL. Acad. Sci. USA 87:3410-3414 (1990). For review of the currently known gene marking and gene therapy protocols see Anderson et al., Science 256:808-813 (1992). See also WO 93/25673 and the references cited therein. 25 Further details of the invention are illustrated by the following non-limiting Examples. The disclosures of all citations in the specification are expressly incorporated herein by reference. Example 1 30 A patient with active rheumatoid arthritis who has an inadequate response to one or more TNFae-inhibitor therapies is treated with an antibody that binds the B-cell surface antigen, CD20. Candidates for therapy according to this example include those who have experienced an inadequate response to previous or current treatment with etanercept, 35 infliximab and/or adalimumab because of toxicity or inadequate efficacy (etanercept for > 3 months at 25 mg twice a week or at least 4 infusions of infliximab at 3 mg/kg). Patients may have swollen joint count (SJC) 8 (66 joint count), and tender 25 joint count (TJC) 8 (68 joint count) at screening and randomization; either CRP 1.5 mg/dl (15 mg/L) or ESR 28 mm/h; and/or radiographic evidence of at least one joint with definite erosion attributable to rheumatoid arthritis, as determined by the central reading site (any joint of the hands, wrists or feet can be considered with the exception 5 of the DIP joints of the hands). The CD20 antibody used for therapy may be Rituximab (commercially available from Genentech, Inc.) or humanized 2H7 v16. Patients are treated with a therapeutically effective dose of the CD20 antibody, for instance, 1000mg i.v. on Days 1 and 15, or 375mg/m2 i.v. weekly x 4. 10 Patients may also receive concomitant MTX (10-25 mg/week per oral (p.o.) or parenteral), together with a corticosteroid regimen consisting of methylprednisolone 100 mg i.v. 30 minutes prior to infusions of the CD20 antibody and prednisone 60 mg p.o. on Days 2-7, 30 mg p.o. Days 8-14, returning to baseline dose by Day 16. Patients may also receive folate (5 mg/week) given as either a single dose or as divided daily 15 doses. Patients optionally continue to receive any background corticosteroid (51 Omg/d prednisone or equivalent) throughout the treatment period. The primary endpoint may be the proportion of patients with an ACR20 response at Week 24 using a Cochran-Mantel-Haenszel (CMH) test for comparing group differences, adjusted for rheumatoid factor and region. 20 Potential secondary endpoints include: 1. Proportion of patients with ACR50 and 70 responses at Week 24. These may be analyzed as specified for the primary endpoint. 2. Change in Disease Activity Score (DAS) from screening to Week 24. These may be assessed using an ANOVA model with baseline DAS, rheumatoid factor, and 25 treatment as terms in the model. 3. Categorical DAS responders (EULAR response) at Week 24. These may be assessed using a CMH test adjusted for rheumatoid factor. 4. Changes from screening in ACR core set (SJC, TJC, patient's and physician's global assessments, HAQ, pain, CRP, and ESR). Descriptive statistics may be reported 30 for these parameters. 5. Changes from screening in SF-36. Descriptive statistics may be reported for the 8 domain scores and the mental and physical component scores. In addition, the mental and physical component scores may be further categorized and analyzed. 6. Change in modified Sharp radiographic total score, erosion score, and joint 35 space narrowing score. These may be analyzed using continuous or categorical methodology, as appropriate. 26 Exploratory endpoints and analysis may involve: ACR(20/50/70 and ACR n) and change in DAS responses over Weeks 8, 12, 16, 20, 24 and beyond will be assessed using a binary or continuous repeated measures model, as appropriate. Exploratory radiographic analyses including proportion of 5 patients with no erosive progression may be assessed at weeks 24 and beyond. Further exploratory endpoints (for example complete clinical response, disease free period) will be analyzed descriptively as part of the extended observation period. Changes from Screen in FACIT-F fatigue will be analyzed with descriptive statistics. 10 Therapy of RA with the CD20 antibody in patients with an inadequate response to TNFa inhibitor therapy as described above will result in a beneficial clinical response according to any one or more of the endpoints noted above. The entire disclosure in the complete specification of our Australian Patent Application No. 2004229376 is by this cross-reference incorporated into the present 15 specification. 27 THE CLAIMS DEFINING THE INVENTION ARE AS FOLLOWS: 1. A method of treating rheumatoid arthritis in a human patient who experiences an inadequate response to a TNFa-inhibitor, comprising administering to the patient an 5 antibody that binds to CD20, wherein the antibody is administered as two intravenous does of 1000mg. 2. The method of claim 1, wherein the antibody comprises rituximab. 10 3. The method of claim 1 or claim 2, wherein the patient is further treated with concomitant methotrexate (MTX). 4. The method of claim 3, wherein the patient is further treated with a corticosteroid regimen. 15 5. The method of claim 4, wherein the corticosteroid regimen consists of methylprednisolone and prednisone. 6. The method of any preceding claim, wherein the CD20 antibody is the only B 20 cell surface marker antibody administered to the patient. 7. The method of any preceding claim, wherein the patient has no erosive progression at weeks 24 and beyond. 25 8. A method of achieving a clinical response selected from the group consisting of ACR50 response at week 24, ACR70 response at week 24 and no erosive progression at week 24 and beyond, in a human rheumatoid arthritis patient who experiences an inadequate response to a TNFa-inhibitor, comprising administering to the patient rituximab and methotrexate, wherein rituximab is administered as two intravenous 30 doses of 1000mg. 9. Use of an antibody that binds CD20 in the manufacture of a medicament for treating rheumatoid arthritis in a human patient who experiences an inadequate response to a TNFa-inhibtior, wherein the antibody is for administering as two 35 intravenous doses of 1000mg. 28 10. Use of rituxan in the manufacture of a medicament for treating rheumatoid arthritis in a patient who experiences an inadequate response to a TNFc-inhibitor, wherein the rituxan is for administering as two intravenous doses of 1000mg. 5 11. The method or use of any preceding claim, substantially as hereinbefore described, with reference to the example. 29

Claims (13)

1. A method of treating an autoimmune disease in a mammal who experiences an inadequate response to a TNFa-inhibitor, comprising administering to the mammal a 5 therapeutically effective amount of an antagonist which binds to a B cell surface marker.
2. The method of claim I wherein the B cell surface marker is selected from the group consisting of CD10, CD19, CD20, CD21, CD22, CD23, CD24, CD37, CD40, 10 CD53, CD72, CD73, CD74, CDw75, CDw76, CD77, CDw78, CD79a, CD79b, CD80, CD81, CD82, CD83, CDw84, CD85 and CD86.
3. The method of claim I wherein the antagonist comprises an antibody. 15
4. The method of claim 3 wherein the antibody binds CD20.
5. The method of claim I wherein the autoimmune disease is selected from the group consisting of arthritis, rheumatoid arthritis, juvenile rheumatoid arthritis, osteoarthritis, psoriatic arthritis, psoriasis, dermatitis, polymyositis/dermatomyositis, 20 toxic epidermal necrolysis, systemic scleroderma and sclerosis, responses associated with inflammatory bowel disease, Crohn's disease, ulcerative colitis, respiratory distress syndrome, adult respiratory distress syndrome (ARDS), meningitis, encephalitis, uveitis, colitis, glomerulonephritis, allergic conditions, eczema, asthma, conditions involving infiltration of T cells and chronic inflammatory responses, atherosclerosis, 25 autoimmune myocarditis, leukocyte adhesion deficiency, systemic lupus erythematosus (SLE), juvenile onset diabetes, multiple sclerosis, allergic encephalomyelitis, immune responses associated with acute and delayed hypersensitivity mediated by cytokines and T-lymphocytes, tuberculosis, sarcoidosis, granulomatosis including Wegener's granulomatosis, agranulocytosis, vasculitis (including ANCA), aplastic anemia, 30 Diamond Blackfan anemia, immune hemolytic anemia including autoimmune hemolytic anemia (AIHA), pernicious anemia, pure red cell aplasia (PRCA), Factor VIII deficiency, hemophilia A, autoimmune neutropenia, pancytopenia, leukopenia, diseases involving leukocyte diapedesis, central nervous system (CNS) inflammatory disorders, multiple organ injury syndrome, mysathenia gravis, antigen-antibody complex mediated 35 diseases, anti-glomerular basement membrane disease, anti-phospholipid antibody syndrome, allergic neuritis, Bechet disease, Castleman's syndrome, Goodpasture's syndrome, Lambert-Eaton Myasthenic Syndrome, Reynaud's syndrome, Sjorgen's -44- syndrome, Stevens-Johnson syndrome, solid organ transplant rejection, graft versus host disease (GVHD), pemphigoid bullous, pemphigus, autoimmune polyendocrinopathies, Reiter's disease, stiff-man syndrome, giant cell arteritis, immune complex nephritis, IgA nephropathy, IgM polyneuropathies or IgM mediated neuropathy, idiopathic 5 thrombocytopenic purpura (ITP), thrombotic throbocytopenic purpura (TTP), autoimmune thrombocytopenia, autoimmune disease of the testis and ovary including autoimune orchitis and oophoritis, primary hypothyroidism; autoimmune endocrine diseases including autoimmune thyroiditis, chronic thyroiditis (Hashimoto's Thyroiditis), subacute thyroiditis, idiopathic hypothyroidism, Addison's disease, 10 Grave's disease, autoimmune polyglandular syndromes (or polyglandular endocrinopathy syndromes), Type I diabetes also referred to as insulin-dependent diabetes mellitus (IDDM) and Sheehan's syndrome; autoimmune hepatitis, lymphoid interstitial pneumonitis (HIV), bronchiolitis obliterans (non-transplant) vs NSIP, Guillain-Barre' Syndrome, large vessel vasculitis (including polymyalgia rheumatica 15 and giant cell (Takayasu's) arteritis), medium vessel vasculitis (including Kawasaki's disease and polyarteritis nodosa), ankylosing spondylitis, Berger's disease (IgA nephropathy), rapidly progressive glomerulonephritis, primary biliary cirrhosis, Celiac sprue (gluten enteropathy), cryoglobulinemia, amyotrophic lateral sclerosis (ALS), coronary artery disease. 20
6. The method of claim I wherein the mammal is human.
7. The method of claim 3 wherein the antibody is not conjugated with a cytotoxic agent. 25
8. The method of claim 4 wherein the antibody comprises rituximab.
9. The method of claim 4 wherein the antibody comprises humanized 2H7 v16 comprising the variable domains as in SEQ ID Nos. I & 2. 30
10. The method of claim 3 wherein the antibody is conjugated with a cytotoxic agent.
11. The method of claim I which consists essentially of administering the 35 antagonist to the mammal. -45-
12. A method of treating rheumatoid arthritis in a mammal who experiences an inadequate response to a TNFa-inhibitor, comprising administering to the mammal a therapeutically effective amount of an antibody that binds to CD20. 5
13. A method of reducing the risk of a negative side effect selected from the group consisting of an infection, heart failure and demyelination, comprising administering to a mammal with an autoimmune disease a therapeutically effective amount of an antagonist which binds to a B cell surface marker. -46-
AU2007242919A 2003-04-09 2007-12-12 Therapy of autoimmune disease in a patient with an inadequate response to a TNF-alpha inhibitor Expired AU2007242919B8 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
AU2007242919A AU2007242919B8 (en) 2003-04-09 2007-12-12 Therapy of autoimmune disease in a patient with an inadequate response to a TNF-alpha inhibitor

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
AU2004229376A AU2004229376B2 (en) 2003-04-09 2004-04-06 Therapy of autoimmune disease in a patient with an inadequate response to a TNF-alpha inhibitor
US60/461,481 2006-04-09
AU2007242919A AU2007242919B8 (en) 2003-04-09 2007-12-12 Therapy of autoimmune disease in a patient with an inadequate response to a TNF-alpha inhibitor

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
AU2004229376A Division AU2004229376B2 (en) 2003-04-09 2004-04-06 Therapy of autoimmune disease in a patient with an inadequate response to a TNF-alpha inhibitor

Publications (4)

Publication Number Publication Date
AU2007242919A1 AU2007242919A1 (en) 2008-01-10
AU2007242919B2 AU2007242919B2 (en) 2011-07-14
AU2007242919B9 AU2007242919B9 (en) 2011-07-21
AU2007242919B8 true AU2007242919B8 (en) 2011-11-10

Family

ID=38961380

Family Applications (1)

Application Number Title Priority Date Filing Date
AU2007242919A Expired AU2007242919B8 (en) 2003-04-09 2007-12-12 Therapy of autoimmune disease in a patient with an inadequate response to a TNF-alpha inhibitor

Country Status (1)

Country Link
AU (1) AU2007242919B8 (en)

Families Citing this family (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP3845238A1 (en) * 2010-04-28 2021-07-07 Oncolmmune, Inc. Methods of use of soluble cd24 for therapy of rheumatoid arthritis

Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2000074718A1 (en) * 1999-06-09 2000-12-14 Immunomedics, Inc. Immunotherapy of autoimmune disorders using antibodies which target b-cells
WO2003029296A1 (en) * 2001-10-02 2003-04-10 Chiron Corporation Human anti-cd40 antibodies

Patent Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2000074718A1 (en) * 1999-06-09 2000-12-14 Immunomedics, Inc. Immunotherapy of autoimmune disorders using antibodies which target b-cells
WO2003029296A1 (en) * 2001-10-02 2003-04-10 Chiron Corporation Human anti-cd40 antibodies

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
De Vita S et al., Arthritis & Rheumatism, 2002, Vol. 46, pages 2029-2033 *

Also Published As

Publication number Publication date
AU2007242919B2 (en) 2011-07-14
AU2007242919B9 (en) 2011-07-21
AU2007242919A1 (en) 2008-01-10

Similar Documents

Publication Publication Date Title
AU2004229376B2 (en) Therapy of autoimmune disease in a patient with an inadequate response to a TNF-alpha inhibitor
AU2005249566B2 (en) Method for treating multiple sclerosis
AU2009201932A1 (en) Detection of CD20 in therapy of autoimmune diseases
AU2004264601A1 (en) Assay for human anti CD20 antibodies and uses therefor
MXPA06002134A (en) Anti-cd20 therapy of ocular disorders.
AU2007242919B2 (en) Therapy of autoimmune disease in a patient with an inadequate response to a TNF-alpha inhibitor
RU2489166C2 (en) Using antibodies for treating autoimmune diseases in patient with inadequate response to tnf-alpha inhibitor

Legal Events

Date Code Title Description
SREP Specification republished
FGA Letters patent sealed or granted (standard patent)
TH Corrigenda

Free format text: IN VOL 25, NO 27, PAGE(S) 3480 UNDER THE HEADING APPLICATIONS ACCEPTED - NAME INDEX UNDER THE NAME GENETECH, INC., APPLICATION NO. 2007242919, UNDER INID (71) CORRECT THE APPLICANT NAME TO GENENTECH, INC.

PC Assignment registered

Owner name: F. HOFFMANN-LA ROCHE AG

Free format text: FORMER OWNER(S): GENENTECH, INC.