WO2011091138A1 - Thérapie d'association pour traiter des maladies auto-immunes - Google Patents

Thérapie d'association pour traiter des maladies auto-immunes Download PDF

Info

Publication number
WO2011091138A1
WO2011091138A1 PCT/US2011/021862 US2011021862W WO2011091138A1 WO 2011091138 A1 WO2011091138 A1 WO 2011091138A1 US 2011021862 W US2011021862 W US 2011021862W WO 2011091138 A1 WO2011091138 A1 WO 2011091138A1
Authority
WO
WIPO (PCT)
Prior art keywords
antibody
self
cell
cells
dna
Prior art date
Application number
PCT/US2011/021862
Other languages
English (en)
Inventor
Matthias Von Herrath
Hideki Garren
Nanette Solvason
Michael Leviten
Original Assignee
Bayhill Therapeutics, Inc.
La Jolla Institute For Allergy And Immunology
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
Application filed by Bayhill Therapeutics, Inc., La Jolla Institute For Allergy And Immunology filed Critical Bayhill Therapeutics, Inc.
Priority to JP2012550114A priority Critical patent/JP2013517329A/ja
Priority to EP11735172A priority patent/EP2525825A1/fr
Publication of WO2011091138A1 publication Critical patent/WO2011091138A1/fr

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K45/00Medicinal preparations containing active ingredients not provided for in groups A61K31/00 - A61K41/00
    • A61K45/06Mixtures of active ingredients without chemical characterisation, e.g. antiphlogistics and cardiaca
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P17/00Drugs for dermatological disorders
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P19/00Drugs for skeletal disorders
    • A61P19/02Drugs for skeletal disorders for joint disorders, e.g. arthritis, arthrosis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P21/00Drugs for disorders of the muscular or neuromuscular system
    • A61P21/04Drugs for disorders of the muscular or neuromuscular system for myasthenia gravis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P29/00Non-central analgesic, antipyretic or antiinflammatory agents, e.g. antirheumatic agents; Non-steroidal antiinflammatory drugs [NSAID]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P3/00Drugs for disorders of the metabolism
    • A61P3/08Drugs for disorders of the metabolism for glucose homeostasis
    • A61P3/10Drugs for disorders of the metabolism for glucose homeostasis for hyperglycaemia, e.g. antidiabetics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/02Immunomodulators
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/02Immunomodulators
    • A61P37/06Immunosuppressants, e.g. drugs for graft rejection
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00

Definitions

  • This invention relates to the treatment of autoimmune diseases with a combination of an immunosuppressive or immunomodulatory agent, for example, one that targets B cells (e.g., by arresting their growth, destroying them, deleting them or altering their differentiation) and a DNA antigen specific therapeutic agent encoding a self protein associated with the autoimmune disease. More specifically, the present invention provides, for example a combination of a immunosuppressive or immunomodulatory agent, such as an antibody and a DNA antigen specific therapeutic agent, such as a DNA plasmid vector encoding an autoantigen associated the autoimmune disease. Pharmaceutical compositions and articles of manufacture such as kits comprising the agents and combinations thereof are also provided.
  • Autoimmune disease is a disease caused by adaptive immunity that becomes misdirected at healthy cells and/or tissues of the body. Autoimmune disease affects 3% of the U.S. population and likely a similar percentage of the industrialized world population (Jacobson et al., Clin Immunol Immunopathol, 84:223-43 (1997)). Autoimmune diseases are characterized by T and B lymphocytes that aberrantly target self-proteins, polypeptides, peptides, and/or other self-molecules causing injury and or malfunction of an organ, tissue, or cell-type within the body (for example, pancreas, brain, thyroid or gastrointestinal tract) to cause the clinical
  • Autoimmune diseases include diseases that affect specific tissues as well as diseases that can affect multiple tissues. This may, in part, for some diseases depend on whether the autoimmune responses are directed to an antigen confined to a particular tissue or to an antigen that is widely distributed in the body.
  • the characteristic feature of tissue-specific autoimmunity is the selective targeting of a single tissue or individual cell type. Nevertheless, certain autoimmune diseases that target ubiquitous self-proteins can also effect specific tissues. For example, in polymyositis the autoimmune response targets the ubiquitous protein histidyl-tRNA synthetase, yet the clinical manifestations primarily involved are autoimmune destruction of muscle.
  • the immune system employs a highly complex mechanism designed to generate responses to protect mammals against a variety of foreign pathogens while at the same time preventing responses against self-antigens.
  • the immune system In addition to deciding whether to respond (antigen specificity), the immune system must also choose appropriate effector functions to deal with each pathogen (effector specificity).
  • effector specificity A cell critical in mediating and regulating these effector functions is the CD4 + T cell.
  • CD4 + T cell A cell critical in mediating and regulating these effector functions.
  • characterizing the types of cytokines made by CD4 + T cells as well as how their secretion is controlled is extremely important in understanding how the immune response is regulated.
  • cytokines such as IL-9, IL-10, were isolated from Th2 clones (Van Snick et al., J. Exp. Med., 169:363-368 (1989); Fiorentino et al., J. Exp. Med. , 170:2081 -2095 (1989)). Finally, additional cytokines, such as IL-3, granulocyte macrophage colony-stimulating factor (GM-CSF), and tumor necrosis factor- (TNF-a) were found to be secreted by both Th l and Th2 cells.
  • GM-CSF granulocyte macrophage colony-stimulating factor
  • TNF-a tumor necrosis factor-
  • SLE Systemic lupus erythematosus
  • immunosuppressive agents are used in management of the systemic autoimmune diseases.
  • Cyclophosphamide is an alkylating agent that causes profound depletion of both T- and B- lymphocytes and impairment of cell-mediated immunity. Cyclosporine, tacrolimus, and mycophenolate mofetil are natural products with specific properties of T-lymphocyte
  • MS multiple sclerosis
  • MS is also treated with immunosuppressive agents including methylprednisolone, other steroids, methotrexate, cladribine and cyclophosphamide.
  • immunosuppressive agents including methylprednisolone, other steroids, methotrexate, cladribine and cyclophosphamide. These immunosuppressive agents, as currently used, have minimal efficacy in treating MS.
  • Current therapy for RA utilizes agents that non-specifically suppress or modulate immune function such as methotrexate, sulfasalazine, hydroxychloroquine, leuflonamide, prednisone, as well as the recently developed TNFa antagonists etanercept and infliximab (Moreland et al., J Rheumatol, 28: 1431 -52 (2001 )).
  • Soluble protein antigens have been administered systemically to inhibit the subsequent immune response to that antigen.
  • Such therapies include delivery of myelin basic protein, its dominant peptide, or a mixture of myelin proteins to animals with experimental autoimmune encephalomyelitis and humans with multiple sclerosis (Brocke et al., Nature, 379:343-6 (1996); Critchfield et al., Science, 263: 1 139-43 (1994); Weiner et al., Annu Rev Immunol, 12:809-37 (1994)), administration of type II collagen or a mixture of collagen proteins to animals with collagen-induced arthritis and humans with rheumatoid arthritis
  • T cell receptor (TCR) peptide immunization See, e.g., Vandenbark, A. A. et al., Nature, 341 :541 (1989).
  • TCR T cell receptor
  • Still another approach is the induction of oral tolerance by ingestion of peptide or protein antigens. See, e.g., Weiner, H. L., Immmunol Today, 18:335 (1997).
  • T1 D various treatment modalities have been investigated with the goal of inducing long-term tolerance to one or all of the antigens targeted in T1 D. See, e.g., M. von Herrath, S. Sanda, K. Herold, Nat Rev Immunol 7, 988 (Dec, 2007) and T. Staeva-Vieira, M. Peakman, M. von Herrath, Clin Exp Immunol 148, 17 (Apr, 2007).
  • the various modalities can be broadly classified into two categories; antigen-nonspecific and antigen-specific interventions.
  • ALS anti-thymocyte globulin
  • systemic immuno-modulators like cyclosporine and anti-CD3 have all shown varying degrees of success.
  • ALS selectively depletes Teff cells while sparing or even inducing the generation of CD4 + CD25 + Foxp3 + Tregs See, e.g., M. Lopez, M. R. Clarkson, M. Albin, M. H. Sayegh, N. Najafian, J Am Soc Nephrol 17, 2844 (Oct, 2006) and K. Minamimura, W. Gao, T.
  • anti-CD3 treatment induces shift from Th l toward Th2 profile in mice (Belghith, 2003 #221 ;von Herrath, 2002 #92) and in humans (B. Bisikirska, J. Colgan, J. Luban, J. A. Bluestone, K. C. Herold, J Clin Invest 115, 2904 (Oct, 2005) and K. C. Herold et al. , J Clin Invest 111, 409 (Feb, 2003)) through a combination of activation induced cell death of Teff cells and selective inactivation of THl -cell subsets (J. A.
  • DiaPep277 (derived from Hsp60). In one study, during a period of two years, administration of DiaPep277 maintained insulin secretion in patients with new onset T1D (I. Raz et al., Lancet 358, 1749 (Nov 24, 2001)), but this was not seen in all follow-up studies, although the effect on T cells was consistent (Roep et al.). Similarly, administration of insulin or insulin peptides in mice leads to the generation of antigen-specific Treg cells that can prevent T1D (N. R. Martinez et al., J Clin Invest 111, 1365 (May, 2003), D. Homann, T. Dyrberg, J. Petersen, M. B. Oldstone, M. G.
  • Tregs selectively proliferate in the - pancreatic draining lymph nodes (PLN) and are capable of dampening autoaggressive CD8 responses (D. Homann, T. Dyrberg, J. Petersen, M. B. Oldstone, M. G. von Herrath, J Immunol 163, 1833 (Aug 15, 1999)) but incapable of reverting recent-onset T1 D (M. Larche, D. C.
  • TI D such auto-reactive T-cells, and in some cases B-cells (H. Bour-Jordan, J. A. Bluestone, J Clin Invest 117, 3642 (Dec, 2007)), infiltrate the pancreas (insulitis).
  • TI D is characterized by the early appearance of islet-specific autoantibodies (L.
  • the major autoantigens in TI D include proinsulin or insulin itself, glutamic acid decarboxylase 65 (GAD65), the islet tyrosine phosphatase (IA-2), and the islet-specific glucose-6-phosphatase catalytic subunit-related protein (M. von Herrath, S. Sanda, K. Herold, Nat Rev Immunol 7, 988 (Dec, 2007)).
  • GID65 glutamic acid decarboxylase 65
  • IA-2 islet tyrosine phosphatase
  • IA-2 islet-specific glucose-6-phosphatase catalytic subunit-related protein
  • Current evidence suggests that insulin is the primary antigen leading to islet cell destruction in mice (M. Nakayama et al, Nature 435, 220 (May 12, 2005)), and probably in humans (S. Arif et al. , J Clin Invest 113, 451 (Feb, 2004), P. F. Bougneres et al. , Diabetes 39,
  • B cells mediate autoimmune diseases by producing autoantibodies or by acting as antigen presenting cells (APCs) (F. S. Wong, L. Wen, Rev Diabet Stud 2, 121 (Fall, 2005)).
  • APCs antigen presenting cells
  • Tl D while such autoantibodies may facilitate or enhance diabetes development, they do not induce the disease (F. S. Wong, L. Wen, Rev Diabet Stud 2, 121 (Fall, 2005)).
  • B cells function as islet APCs for T-cells and the autoantibodies expressed on cell surface improve the capture and presentation of autoantigens (M. Falcone, J. Lee, G. Patstone, B. Yeung, N.
  • Naturally occurring CD4 + CD25 + FoxP3 + regulatory-T-cells constitute 5-10% of all CD4 + T cells in the thymus and lymph nodes. They constitutively express GITR, OX40 (CD134) and CTLA-4 (E. M. Shevach, R. S. McHugh, C. A. Piccirillo, A. M. Thornton, Immunol Rev 182, 58 (Aug, 2001 ), J. Shimizu, S. Yamazaki, T. Takahashi, Y. Ishida, S. Sakaguchi, Nat Immunol 3, 135 (Feb, 2002)).
  • CD127 expression is inversely correlated with Foxp3, and together with the other markers, identifies over 95% of the Foxp3 + cells in peripheral blood W.
  • Tregs play a central role in peripheral T-cell tolerance through the induction of IL-4, IL-10 or TGF-b (L. Chatenoud, B. Salomon, J. A. Bluestone, Immunol Rev 182, 149 (Aug, 2001)) and can promote infectious tolerance (H. Waldmann, S. Cobbold, Annu Rev Immunol 16, 619 (1998), H. Waldmann, S.
  • Tregs results in autoimmunity (E. Maggi et al, Autoimmun Rev 4, 579 (Nov, 2005), S. Sakaguchi, Nat Immunol 6, 345 (Apr, 2005)).
  • they can be used for the treatment of Tl D (S. Sakaguchi, Nat Immunol 6, 345 (Apr, 2005), S. Sakaguchi, Nat Immunol 6, 345 (Apr, 2005)).
  • B cells may vary depending on the autoimmune disease.
  • T1D autoantibodies against major T1 D antigens appear early (L. Yu et al , Proc Natl Acad Sci U S A 97, 1701 (Feb 15, 2000)) and are a predictive marker for diabetes progression in humans.
  • B-cells although not essential for the generation of diabetes, are clearly involved in the progression of diabetes, owing to their antigen presenting cell (APC) function to T cells.
  • APC antigen presenting cell
  • B cell membrane receptors evolve and change throughout the B cell life span.
  • Tumor necrosis factor receptor superfamily member 13B TACI
  • tumor necrosis factor receptor superfamily member 17 BCMA
  • B cell-activating factor BAFF-R
  • All of the foregoing receptors may be inhibited by an antibody to BLyS (Belimumab).
  • CD22 is a member of the sialic acid binding Ig-like lectins cell surface receptors and is found on B cells as is CD 19.
  • the CD20 antigen also called human B-lymphocyte-restricted differentiation antigen, Bp35, or B l
  • Bp35 human B-lymphocyte-restricted differentiation antigen
  • B l human B-lymphocyte-restricted differentiation antigen
  • 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 hematopoietic 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.
  • CD20 undergoes phosphorylation in activated B cells (Riley and Sliwkowski, Semin Oncol, 27(12): 17-24 (2000)). CD20 appears on the surface of B-lymphocytes at the pre-B-cell stage and is found on mature and memory B cells, but not plasma cells (Stashenko et al. J. Immunol, 125: 1678-1685 ( 1980); Clark and Ledbetter Adv. Cancer Res., 52:81-149 (1989)). CD20 has calcium-channel activity and may have a role in the development of B cells.
  • anti-CD20 antibodies including rituximab
  • rituximab have been shown to induce apoptosis in vitro when crosslinked by a secondary antibody or by other means (Ghetie et al. Proc Natl Acad Sci. USA, 94:7509-7514 ( 1997)).
  • this antigen can serve as a candidate for "targeting" of B cells in autoimmune disease.
  • 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 antigen of (ostensibly) both normal and autoimmune associated B cells; the antibody bound to the CD20 surface antigen may lead to the destruction and depletion of autoimmune associated B cells. Additionally, chemical agents or radioactive labels having the potential to destroy the autoimmune associated B cells can be conjugated to the anti-CD20 antibody such that the agent is specifically "delivered" to the autoimmune associated B cells. Irrespective of the approach, a primary goal is to destroy the autoimmune associated B cells; the specific approach can be determined by the particular anti- CD20 antibody that is utilized, and thus, the available approaches to targeting the CD20 antigen can vary considerably.
  • the rituximab (RITUXAN®) antibody is a genetically engineered chimeric
  • Rituximab is the antibody called "C2B8" in US 5736137 (Anderson et al). Rituximab is indicated for the treatment of patients with relapsed or refractory low-grade or follicular, CD20-positive, B-cell non-Hodgkin's lymphoma. In vitro mechanism-of-action studies have demonstrated that rituximab binds human complement and lyses lymphoid B-cell lines through CDC (Reff et al, Blood, 83(2): 435-445 (1994)). Additionally, it has significant activity in assays for ADCC.
  • rituximab has been shown to have anti-proliferative effects in tritiated thymidine- incorporation assays and to induce apoptosis directly, while other anti-CD19 and anti-CD20 antibodies do not (Maloney et al. Blood, 88(10): 637a (1996)). Synergy between rituximab and chemotherapies and toxins has also been observed experimentally.
  • rituximab sensitizes drug-resistant human B-cell 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)).
  • doxorubicin doxorubicin
  • CDDP CDDP
  • VP-16 diphtheria toxin
  • ricin diphtheria toxin
  • ricin ricin
  • Rituximab was approved in the United States for the treatment of patients with relapsed or refractory low-grade or follicular CD20 + B-cell NHL at a dose of 375 rag/m 2 weekly for four doses.
  • FDA Food and Drug Administration
  • additional claims for the treatment of low-grade NHL include re-treatment (weekly for four doses) and an additional dosing regimen (weekly for eight doses).
  • Many patients have been exposed to rituximab either as monotherapy or in combination with immunosuppressant or chemotherapeutic drugs. Patients have also been treated with rituximab as maintenance therapy for up to two years. Hainsworth et al, J. Clin.
  • Rituximab has also been approved in the United States in combination with MTX to reduce signs and symptoms in adult patients with moderately- to severely-active RA who have had an inadequate response to at least one TNF antagonist.
  • Many studies address the use of rituximab in a variety of non-malignant autoimmune disorders, including RA, in which B cells and autoantibodies appear to play a role in disease pathophysiology. Edwards et al, Biochem Soc. Trans., 30:824-828 (2002).
  • Rituximab has been reported to potentially relieve signs and symptoms of, for example, RA (Leandro et al, Ann. Rheum. Dis.
  • a Phase II study (WA 16291 ) has been conducted in patients with RA, providing 48- week follow-up data on safety and efficacy of rituximab (Emery et al, Arthritis Rheum., 48(9):S439 (2003); Szczepanski et al, Arthritis Rheum., 48(9):S 121 (2003)).
  • a total of 161 patients were evenly randomized to four treatment arms: MTX, rituximab alone, rituximab plus MTX, and rituximab plus cyclophosphamide (CTX).
  • CTX cyclophosphamide
  • the treatment regimen of rituximab was one gram administered intravenously on days 1 and 15.
  • Patents and patent publications concerning CD20 antibodies, CD20-binding molecules, and self-antigen vaccines include U.S. 5,776,456, 5,736,137, 5,843,439, 6,399,061 , and
  • 2006/0121032 (Dahiyat et al); WO 2006/064121 (Teillaud et al); US 2006/0153838 (Watkins), CN 1718587 (Chen et al); WO 2006/084264 (Adams et al); US 2006/0188495 (Barron et al); US 2004/0202658 and WO 2004/091657 (Benynes, K.); US 2005/0095243, US 2005/0163775, WO 2005/00351 , and WO 2006/068867 (Chan, A.); US 2006/0135430 and WO 2005/005462 (Chan et al); US 2005/0032130 and WO 2005/017529 (Beresini et al); US 2005/0053602 and WO 2005/023302 (Brunetta, P.); US 2006/0179501 and WO 2004/060052 (Chan et al); WO 2004/060053 (Ch
  • 2007/0014720 (Gazit-Bornstein et al); and US 2007/0020259 (Hansen et al); US
  • 2007/0020265 Goldenberg and Hansen
  • US 2007/0014797 Hazar et al
  • US 2007/0224189 Lazar et al
  • WO 2007/014238 Bruge and Bruger
  • WO 2008/003319 Parren and Baadsgaard
  • rituximab was found to be a well-tolerated, effective remission induction agent for severe ANCA-associated vasculitis, when used in a dose of 375 mg/m 2 x four along with oral prednisone at 1 mg/kg/day, which was reduced to 40 mg/day by week four, and to total discontinuation over the following 16 weeks.
  • Four patients were re-treated with rituximab alone for recurring/rising ANCA titers.
  • no additional immunosuppressive agents seem necessary for remission induction and maintenance of sustained remission (six months or longer). Keogh et al, Kidney Blood Press.
  • DNA antigen specific therapy can be used to modulate autoimmune processes.
  • plasmid DNA is taken up by, for example, by muscle cells allowing for the expression of the encoded polypeptide (Wolff et al., Hum Mol Genet..1(6): 363- 9 (1992)).
  • the effect is a shift in an ongoing immune response to suppress autoimmune destruction and is believed to include a shift in self-reactive lymphocytes from a Th l - to a Th2-type response.
  • the modulation of the immune response may not be systemic but occur only locally at the target organ under autoimmune attack.
  • the present invention provides methods and compositions useful in the treatment of autoimmune diseases.
  • the present invention further relates to the treatment of autoimmune diseases comprising the administration of an immunosuppressive or immunomodulatory agent (e.g., a B cell specific agent) in combination with a DNA antigen specific therapeutic agent encoding a self protein associated with the autoimmune disease.
  • an immunosuppressive or immunomodulatory agent e.g., a B cell specific agent
  • a DNA antigen specific therapeutic agent encoding a self protein associated with the autoimmune disease.
  • the present invention provides, for example a combination of a B cell specific agent, such as an antibody, in combination with a DNA antigen specific therapeutic agent, such as a DNA plasmid vector encoding an autoantigen associated the autoimmune disease.
  • the B cell specific agent is an antibody that targets a receptor on the B cell administered in combination with a DNA antigen specific therapeutic agent encoding a self-protein associated with the autoimmune disease.
  • the method of treating an autoimmune disease comprises the administration of a B cell specific agent that specifically binds to certain B cell surface antigens, such as, CD20, CD 19, CD22, TACI, BCMA, or BAFF-R, in combination with a DNA antigen specific therapeutic agent encoding a self protein associated with the autoimmune disease.
  • the method of treating an autoimmune disease comprises the administration of a B cell specific agent that specifically binds to a certain B cell surface antigens, such as CD20, in combination with a DNA antigen specific therapeutic agent encoding a self protein associated with the autoimmune disease, T1 D, such as proinsulin.
  • the B cell specific agent is an antibody, (e.g. a chimeric, humanized or human antibody).
  • the DNA antigen specific therapeutic agent encodes an autoantigen associated with the autoimmune disease.
  • Pharmaceutical compositions comprising the B cell specific agent and DNA antigen specific therapeutic agent and methods of using the B cell specific agent and DNA antigen specific therapeutic agent are also provided.
  • the invention provides methods for the treatment of autoimmune diseases, such as multiple sclerosis, insulin dependent diabetes mellitus, rheumatoid arthritis, pemphigus vulgaris or myasthenia gravis by administering to the subject a therapeutically effective amount of an immunosuppressive or immunomodulatory agent (e.g., a B cell specific agent) in combination with a DNA antigen specific therapeutic agent encoding a self-protein.
  • an immunosuppressive or immunomodulatory agent e.g., a B cell specific agent
  • the invention provides for the treatment of insulin dependent diabetes mellitus comprising administering to a patient a therapeutically effective amount of an immunosuppressive or immunomodulatory agent (e.g., a B cell specific agent) in combination with a DNA antigen specific therapeutic agent encoding a self-protein.
  • an immunosuppressive or immunomodulatory agent e.g., a B cell specific agent
  • the B cell specific agent is a CD20 specific agent in combination with a DNA antigen specific therapeutic agent encoding an autoantigen associated with insulin dependent diabetes mellitus, such as insulin, proinsulin, glutamic acid decarboxylase 65 or islet cell antigen.
  • the CD20 specific agent is a monoclonal antibody such as Rituxan and the DNA antigen specific therapeutic agent encodes proinsulin.
  • compositions comprising the B cell specific agent, such as a CD20 specific agent, and DNA antigen specific therapeutic agent encoding an autoantigen associated with insulin dependent diabetes mellitus and methods of using the B cell specific agent, such as a CD20 specific agent, and DNA antigen specific therapeutic agent encoding an autoantigen associated with insulin dependent diabetes mellitus are also provided.
  • B cell specific agent such as a CD20 specific agent
  • DNA antigen specific therapeutic agent encoding an autoantigen associated with insulin dependent diabetes mellitus
  • the invention provides for the treatment of multiple sclerosis comprising administering to a patient a therapeutically effective amount of an
  • immunosuppressive or immunomodulatory agent e.g., a B cell specific agent
  • a DNA antigen specific therapeutic agent encoding a self-protein.
  • the B cell specific agent is a CD20 specific agent in combination with a DNA antigen specific therapeutic agent encoding an autoantigen associated with multiple sclerosis, such as myelin basic protein, proteolipid protein, myelin associated glycoprotein, cyclic nucleotide
  • the CD20 specific agent is a monoclonal antibody such as Rituxan and the DNA antigen specific therapeutic agent encodes myelin basic protein
  • the B cell specific agent such as a CD20 specific agent
  • DNA antigen specific therapeutic agent encodes myelin basic protein
  • compositions comprising the B cell specific agent, such as a CD20 specific agent, and DNA antigen specific therapeutic agent encoding an autoantigen associated with multiple sclerosis and methods of using the B cell specific agent, such as a CD20 specific agent, and DNA antigen specific therapeutic agent encoding an autoantigen associated with multiple sclerosis are also provided.
  • the invention provides for the treatment of an autoimmune disease comprising the administration to a subject of an immunosuppressive or immunomodulatory agent (e.g., a B cell specific agent) followed by administration of the DNA antigen specific therapeutic agent.
  • an immunosuppressive or immunomodulatory agent e.g., a B cell specific agent
  • administration of the DNA antigen specific therapeutic agent is one to several days after administration of the immunosuppressive or
  • administration of the DNA antigen specific therapeutic agent is one week after administration of the B cell specific agent.
  • a single administration of the immunosuppressive or immunomodulatory agent is followed by weekly administration of the DNA antigen specific therapeutic agent.
  • compositions comprising various combinations of the agents useful in the methods of treatment described herein.
  • the compositions may comprise, in an amount effective for the treatment of an autoimmune disease comprising: (a) an immunosuppressive or immunomodulatory agent (e.g., a B cell specific agent); (b) a DNA antigen specific therapeutic agent; and, (c) a pharmaceutically acceptable carrier.
  • an immunosuppressive or immunomodulatory agent e.g., a B cell specific agent
  • a DNA antigen specific therapeutic agent e.g., a B cell specific agent
  • a pharmaceutically acceptable carrier e.g., a B cell specific agent
  • the pharmaceutical compositions comprise an antibody that binds to CD20 and a DNA antigen specific therapeutic agent encoding a self-protein together with pharmaceutically acceptable carriers.
  • the immunosuppressive or immunomodulatory agent is an antibody.
  • the antibody is a recombinant antibody, a monoclonal antibody, a chimeric antibody, a humanized antibody, or a human antibody.
  • the antibody is isolated. In still further embodiments, the antibody is substantially pure.
  • the DNA antigen specific therapeutic agent is a DNA plasmid vector.
  • the DNA plasmid vector is isolated. In still further embodiments, the DNA plasmid vector is substantially pure.
  • kits and articles of manufacture are kits and articles of manufacture. Kits and articles of
  • manufacture preferably include:
  • the label on said container may indicate that the composition can be used for treating an autoimmune disease
  • the active agent in said first composition comprises an immunosuppressive or immunomodulatory agent
  • the active agent in said second composition comprises a DNA antigen specific therapeutic agent.
  • FIG. 1 Anti-CD20 administration alone can induce protection from diabetes in NOD mice.
  • Prediabetic 7-10 week-old NOD mice were injected with 5, 10, 50, 100 or 250 ⁇ g of anti- CD20 antibody (5D2) either only on day 1 (Fig. 1 A) or on days 1 , 2, 3, and 4 (Fig. I B).
  • Progression of NOD mice to diabetes was monitored by measuring blood glucose values every week, and the mice were considered diabetic when two consecutive BG values were >200 mg/dL. While the highest dose of lx anti-CD20 (250 ⁇ g) appears to mediate 50% protection, all the mice that received 4x anti-CD20 injections appear to be protected from diabetes onset.
  • Figure 2 Combining anti-CD20 administration with DNA self-vector encoding proinsulin administration protects NOD mice from diabetes. Prediabetic 7-10 week-old NOD mice were injected with (Fig. 2A) 10, (Fig. 2B) 50, or (Fig. 2C) 100 ⁇ g of anti-CD20 antibody (5D2) either alone or in combination with a plasmid expressing proinsulin, either l x (dl ) or 4x (dl , 8, 15, 22). Once a week, blood glucose values were determined.
  • FIG. 3 Prediabetic 7-10 week-old NOD mice were injected with either (A) anti- CD20 alone, or (B) in combination with 50 ⁇ g Proinsulin plasmid, at indicated doses. Untreated NOD mice were used as controls to monitor diabetes development. Once a week, blood glucose values were determined.
  • FIG. 4 Prediabetic 7-10 week-old NOD mice were injected with 25 ⁇ g plasmid in each thigh (total 50 ⁇ g) at weekly intervals, in combination with anti-CD20 administration (doses indicated in Figure legend) as follows; (A) single dose of anti-CD20 and Proinsulin plasmid, or (B) four doses of anti-CD20 (d l , 2, 3, 4) and single dose of Proinsulin plasmid, or (C) single dose of anti-CD20 and four doses of Proinsulin plasmid. Untreated NOD mice were used as controls to monitor diabetes development. Once a week, blood glucose values were determined.
  • Rats receiving rBHT-3034/Solu-medrol combination therapy show a statistically significant reduction in disease scores relative to the PBS treated control group and the rBHT-3034 and Solu-Medrol monotherapy groups.
  • nucleotide phosphodiesterase myelin- associated glycoprotein, myelin-associated oligodendrocytic basic protein, myelin oligodendrocyte glycoprotein, alpha-B- cry stalin
  • carboxypeptidase H insulin, proinsulin, pre- proinsulin, heat shock proteins, glima 38, islet cell antigen 69 KDa, p52, islet cell glucose transporter GLUT-2
  • Type 1 Diabetes Type 1 diabetes (T1D) (sometimes also referred to as human type I or insulin-dependent diabetes mellitus (IDDM)) is characterized by autoimmune destruction of the ⁇ cells in the pancreatic islets of Langerhans. The depletion of ⁇ cells results in an inability to regulate levels of glucose in the blood. Overt diabetes occurs when the level of glucose in the blood rises above a specific level, usually about 250 mg/dl. In humans a long presymptomatic period precedes the onset of diabetes. During this period there is a gradual loss of pancreatic beta cell function. The development of disease is implicated by the presence of autoantibodies against insulin, glutamic acid decarboxylase, and the tyrosine phosphatase IA2 (IA2).
  • IA2 tyrosine phosphatase IA2
  • Markers that may be evaluated during the presymptomatic stage are the presence of insulitis in the pancreas, the level and frequency of islet cell antibodies, islet cell surface antibodies, aberrant expression of Class II MHC molecules on pancreatic beta cells, glucose concentration in the blood, and the plasma concentration of insulin.
  • An increase in the number of T lymphocytes in the pancreas, islet cell antibodies and blood glucose is indicative of the disease, as is a decrease in insulin concentration.
  • NOD Non-Obese Diabetic
  • the presence of combinations of autoantibodies with various specificities in serum are highly sensitive and specific for human type I diabetes mellitus.
  • the presence of autoantibodies against GAD and/or IA-2 is approximately 98% sensitive and 99% specific for identifying type I diabetes mellitus from control serum.
  • the presence of autoantibodies specific for two of the three autoantigens including GAD, insulin and IA-2 conveys a positive predictive value of >90% for development of type IDM within 5 years.
  • Autoantigens targeted in human insulin dependent diabetes mellitus may include, for example, tyrosine phosphatase IA-2; ⁇ -2 ⁇ ; glutamic acid decarboxylase (GAD) both the 65 kDa and 67 kDa forms; carboxypeptidase H; insulin; proinsulin (e.g., SEQ ID NOs: 1 and 2) ; heat shock proteins (HSP); glima 38; islet cell antigen 69 KDa (ICA69); p52; two ganglioside antigens (GT3 and GM2-1 ); islet-specific glucose-6-phosphatase-related protein (IGRP); and an islet cell glucose transporter (GLUT 2).
  • GID glutamic acid decarboxylase
  • Human T1 D is currently treated by monitoring blood glucose levels to guide injection, or pump-based delivery, of recombinant insulin. Diet and exercise regimens contribute to achieving adequate blood glucose control.
  • MS Multiple sclerosis
  • MM Multiple sclerosis
  • Onset of symptoms typically occurs between 20 and 40 years of age and manifests as an acute or subacute attack of unilateral visual impairment, muscle weakness, paresthesias, ataxia, vertigo, urinary incontinence, dysarthria, or mental disturbance (in order of decreasing frequency).
  • Such symptoms result from focal lesions of demyelination which cause both negative conduction abnormalities due to slowed axonal conduction, and positive conduction abnormalities due to ectopic impulse generation (e.g. , Lhermitte's symptom).
  • Diagnosis of MS is based upon a history including at least two distinct attacks of neurologic dysfunction that are separated in time, produce objective clinical evidence of neurologic dysfunction, and involve separate areas of the CNS white matter.
  • Laboratory studies providing additional objective evidence supporting the diagnosis of MS include magnetic resonance imaging (MRI) of CNS white matter lesions, cerebral spinal fluid (CSF) oligoclonal banding of IgG, and abnormal evoked responses.
  • MRI magnetic resonance imaging
  • CSF cerebral spinal fluid
  • IgG cerebral spinal fluid
  • the autoantigen targets of the autoimmune response in autoimmune demyelinating diseases may comprise epitopes from proteolipid protein (PLP); myelin basic protein (MBP); myelin oligodendrocyte glycoprotein (MOG); cyclic nucleotide phosphodiesterase (CNPase); myelin- associated glycoprotein (MAG), and myelin-associated oligodendrocytic basic protein (MBOP); alpha-B-crystalin (a heat shock protein); viral and bacterial mimicry peptides, e.g., influenza, herpes viruses, hepatitis B virus, etc.; OSP (oligodendrocyte specific-protein); citrulline- modified MBP (the C8 isoform of MBP in which 6 arginines have been de-imminated to citrulline), etc.
  • the integral membrane protein PLP is a dominant autoantigen of myelin basic protein (MBP); myelin oligodendrocyte glycoprotein (
  • Determinants of PLP antigenicity have been identified in several mouse strains, and include residues 139-151 , 103-1 16, 215-232, 43-64 and 178-191 . At least 26 MBP epitopes have been reported (Meinl et al., J Clin Invest 92, 2633-43, 1993). Notable are residues 1 -1 1 , 59-76 and 87-99. Immunodominant MOG epitopes that have been identified in several mouse strains include residues 1 -22, 35-55, 64-96.
  • MOG glycoprotein
  • MOG 34-56 MOG 34-56
  • MOG 64-96 erlero de Rosbo et al , Eur J Immunol 27, 3059-69, 1997
  • T and B cell (brain lesion-eluted Ab) response focused on MBP 87-99 (Oksenberg et al, Nature 362, 68-70, 1993).
  • Rheumatoid Arthritis Rheumatoid arthritis (RA) is a chronic autoimmune autoimmune autoimmune autoimmune autoimmune autoimmune autoimmune autoimmune autoimmune autoimmune autoimmune autoimmune autoimmune autoimmune autoimmune autoimmune autoimmune autoimmune autoimmune autoimmune autoimmune autoimmune autoimmune autoimmune autoimmune autoimmune autoimmune autoimmune autoimmune autoimmune autoimmune autoimmune autoimmune autoimmune autoimmune autoimmune autoimmune autoimmune autoimmune autoimmune autoimmune autoimmune autoimmune erythematolism, RA, RA, RA, RA, RA, RA, RA, rheumatoid arthritis (RA) is a chronic autoimmune autoimmune autoimmune autoimmune autoimmune autoimmune autoimmune autoimmune autoimmune autoimmune autoimmune autoimmune autoimmune autoimmune autoimmune autoimmune autoimmune autoimmune autoimmune autoimmune autoimmune autoimmune autoimmune autoimmune autoimmune autoimmune autoimmune autoimmune autoimmune autoimmune autoimmune autoimmune autoimmune autoimmune autoimmune autoimmune autoimmune autoimmune autoimmune autoimmune autoimmune autoimmune autoimmune autoimmune autoimmune autoimmune autoimmune autoimmune
  • RA inflammatory synovitis affecting 0.8% of the world population. It is characterized by chronic inflammatory synovitis that causes erosive joint destruction. RA is mediated by T cells, B cells and macrophages.
  • T cells play a critical role in RA includes the (1) predominance of CD4 + . T cells infiltrating the synovium, (2) clinical improvement associated with suppression of T cell function with drugs such as cyclosporine, and (3) the association of RA with certain HLA-DR alleles.
  • the HLA-DR alleles associated with RA contain a similar sequence of amino acids at positions 67-74 in the third hypervariable region of the ⁇ chain that are involved in peptide binding and presentation to T cells.
  • RA is mediated by autoreactive T cells that recognize a self- protein, or modified self-protein, present in synovial joints. Autoantigens that are targeted in RA comprise, e.g., epitopes from type II collagen; hnRNP; A2/RA33; Sa; filaggrin; keratin;
  • citrulline cartilage proteins including gp39; collagens type I, III, IV, V, IX, XI; HSP-65/60; IgM (rheumatoid factor); RNA polymerase; hnRNP-B l ; hnRNP-D; cardiolipin; aldolase A; citrulline- modified filaggrin and fibrin.
  • Autoantibodies that recognize filaggrin peptides containing a modified arginine residue (de-iminated to form citrulline) have been identified in the serum of a high proportion of RA patients.
  • Autoreactive T and B cell responses are both directed against the same immunodominant type II collagen (CII) peptide 257-270 in some patients.
  • Autoimmune Uveitis is an autoimmune disease of the eye that is estimated to affect 400,000 people, with an incidence of 43,000 new cases per year in the U.S. Autoimmune uveitis is currently treated with steroids, immunosuppressive agents such as methotrexate and cyclosporin, intravenous immunoglobulin, and TNFa-antagonists.
  • EAU Experimental autoimmune uveitis
  • CFA Complete Freund's Adjuvant
  • Autoantigens targeted by the autoimmune response in human autoimmune uveitis may include S-antigen, interphotoreceptor retinoid binding protein (IRBP), rhodopsin, and recoverin.
  • IRBP interphotoreceptor retinoid binding protein
  • PBC Primary Biliary Cirrhosis
  • IBEC intrahepatic biliary epithelial cells
  • M2 represents multiple autoantigenic subunits of enzymes of the 2-oxoacid dehydrogenase complex (2-OADC) and is another example of the self-protein, -polypeptide, or -peptide of the instant invention.
  • 2-OADC 2-oxoacid dehydrogenase complex
  • PDC pyruvate dehydrogenase complex
  • E2 74 kDa subunit belonging to the PDC-E2.
  • PDC-E2 The most frequent reactivity in 95% of cases of PBC is the E2 74 kDa subunit, belonging to the PDC-E2.
  • OGDC 2-oxoglutarate dehydrogenase complex
  • BC branched-chain
  • E3BP E-3 Binding protein
  • the E2 polypeptide contains two tandemly repeated lipoyl domains, while E3BP has a single lipoyl domain.
  • the lipoyl domain is found in a number of autoantigen targets of PBC and is referred to herein as the "PBC lipoyl domain.”
  • PBC is treated with glucocorticoids and immunosuppressive agents including methotrexate and cyclosporin A.
  • a murine model of experimental autoimmune cholangitis uses intraperitoneal (i.p.) sensitization with mammalian PDC in female SJL/J mice, inducing non-suppurative destructive cholangitis (NSDC) and production of AMA (Jones, J Clin Pathol 53:813-21 , 2000).
  • Autoanti gens for myasthenia gravis may include epitopes within the acetylcholine receptor.
  • Autoantigens targeted in pemphigus vulgaris may include desmoglein-3.
  • Sjogren's syndrome antigens may include SSA (Ro); SSB (La); and fodrin.
  • the dominant autoantigen for pemphigus vulgaris may include desmoglein-3.
  • Panels for myositis may include tRNA synthetases ⁇ e.g., threonyl, histidyl, alanyl, isoleucyl, and glycyl); Ku; Scl; SSA; Ul Sn ribonuclear protein; Mi-1 ; Mi- 1 ; Jo-1 ; Ku; and SRP.
  • Panels for scleroderma may include Scl-70; centromere; U l ribonuclear proteins; and fibrillarin.
  • Panels for pernicious anemia may include intrinsic factor; and glycoprotein beta subunit of gastric H/K ATPase.
  • Epitope Antigens for systemic lupus erythematosus may include DNA; phospholipids; nuclear antigens; Ro; La; Ul ribonucleoprotein; Ro60 (SS-A); Ro52 (SS-A); La (SS-B); calreticulin; Grp78; Scl-70; histone; Sm protein; and chromatin, etc.
  • SLE systemic lupus erythematosus
  • Epitope Antigens for systemic lupus erythematosus may include DNA; phospholipids; nuclear antigens; Ro; La; Ul ribonucleoprotein; Ro60 (SS-A); Ro52 (SS-A); La (SS-B); calreticulin; Grp78; Scl-70; histone; Sm protein; and chromatin, etc.
  • Grave's disease epitopes may include the Na+/I- symporter; thyrotropin receptor; Tg; and TPO.
  • Graft Versus Host Disease One of the greatest limitations of tissue and organ transplantation in humans is rejection of the tissue transplant by the recipient's immune system. It is well established that the greater the matching of the MHC class I and II (HLA-A, HLA-B, and HLA-DR) alleles between donor and recipient the better the graft survival. Graft versus host disease (GVHD) causes significant morbidity and mortality in patients receiving transplants containing allogeneic hematopoietic cells. Hematopoietic cells are present in bone-marrow transplants, stem cell transplants, and other transplants.
  • T lymphocytes and other immune cell in the donor graft attack the recipients' cells that express polypeptides variations in their amino acid sequences, particularly variations in proteins encoded in the major histocompatibility complex (MHC) gene complex on chromosome 6 in humans.
  • MHC major histocompatibility complex
  • the most influential proteins for GVHD in transplants involving allogeneic hematopoietic cells are the highly polymorphic (extensive amino acid variation between people) class I proteins (HLA-A, -B, and -C) and the class II proteins (DRB 1 , DQB 1 , and DPB 1 ) (Appelbaum, Nature 41 1 :385-389, 2001 ). Even when the MHC class I alleles are serologically 'matched' between donor and recipient, DNA sequencing reveals there are allele-level mismatches in 30% of cases providing a basis for class I-directed GVHD even in matched donor-recipient pairs (Appelbaum, Nature 41 1 , 385-389, 2001).
  • GVHD minor histocompatibility self-antigens GVHD frequently causes damage to the skin, intestine, liver, lung, and pancreas.
  • GVHD is treated with glucocorticoids, cyclosporine, methotrexate, fludarabine, and OKT3.
  • Tissue Transplant Rejection Immune rejection of tissue transplants, including lung, heart, liver, kidney, pancreas, and other organs and tissues, is mediated by immune responses in the transplant recipient directed against the transplanted organ. Allogeneic transplanted organs contain proteins with variations in their amino acid sequences when compared to the amino acid sequences of the transplant recipient. Because the amino acid sequences of the transplanted organ differ from those of the transplant recipient they frequently elicit an immune response in the recipient against the transplanted organ. Rejection of transplanted organs is a major complication and limitation of tissue transplant, and can cause failure of the transplanted organ in the recipient. The chronic inflammation that results from rejection frequently leads to dysfunction in the transplanted organ. Transplant recipients are currently treated with a variety of immunosuppressive agents to prevent and suppress rejection. These agents include glucocorticoids, cyclosporin A, Cellcept, FK-506, and OKT3.
  • an "immunsuppressive or immunomodulatory agent” is an agent that generally or specifically suppresses or modulates a mammalian immune response. Such agents may be, for example, a small molecule or a biological therapeutic ⁇ e.g. an inhibitory nucleic acid or a protein, such as a soluble receptor or antibody).
  • exemplary immunosuppressive agents include glucocorticoids ⁇ e.g., methylprednisolone), cytotoxic agents, corticosteroids ⁇ e.g., prednisone), cyclophosphamide, cyclosporine, tacrolimus, methotrexate, cladribine, mycophenolate mofetil, and the like.
  • Immunomodulatory agents include antibodies or other proteins that affect the action of lymphocytes (B cells or T cells) or cytokines, such as TNFa ⁇ e.g., etanercept and infliximab).
  • B cells or T cells lymphocytes
  • cytokines such as TNFa ⁇ e.g., etanercept and infliximab.
  • immunomodulatory agent is a B cell specific agent.
  • a "B cell” is a lymphocyte that matures within the bone marrow, and includes a naive B cell, memory B cell, or effector B cell (plasma cells).
  • the B cell herein is a normal or non- malignant B cell.
  • a "B-cell surface marker” or "B-cell surface antigen” herein is an antigen expressed on the surface of a B cell that can be targeted with an antagonist that binds thereto.
  • Exemplary B- cell surface markers include the CD 10, CD 19, CD20, CD21 , CD22, CD23, CD24, CD37, CD40, CD53, CD72, CD73, CD74, CDw75, CDw76, CD77, CDw78, CD79a, CD79b, CD80, CD81 , CD82, CD83, CDw84, CD85 and CD86 leukocyte surface markers (for descriptions, see The Leukocyte Antigen Facts Book, 2 nd Edition. 1997, ed. Barclay et al.
  • B-cell surface markers include RP 105, FcRH2, B-cell CR2, CCR6, P2X5, HLA-DOB, CXCR5, FCER2, BR3, BAFF, BLyS, Btig, NAG 14, SLGC 16270, FcRHl , IRTA2, ATWD578, FcRH3, IRTA 1 , FcRH6, BCMA, and 239287.
  • the B-cell surface marker of particular interest is preferentially expressed on B cells compared to other non-B-cell tissues of a mammal and may be expressed on both precursor B cells and mature B cells.
  • the preferred B-cell surface markers herein are CD20, CD22, CD23, CD40, BR3, BLyS, and BAFF.
  • CD20 antigen is an about 35-kDa, non-glycosylated phosphoprotein found on the surface of greater than 90% of B cells from peripheral blood or lymphoid organs. CD20 is present on normal B cells and is not expressed on stem cells. Other names for CD20 in the literature include "B-lymphocyte-restricted antigen” and "Bp35". The CD20 antigen is described in Clark et al., Proc. Natl. Acad. Sci. (USA), 82: 1766 (1985), for example.
  • CD22 antigen also known as BL-CAM or Lyb8
  • BL-CAM BL-CAM
  • Lyb8 is a type 1 integral membrane glycoprotein with molecular weight of about 130 (reduced) to 140kD (unreduced). It is expressed in both the cytoplasm and cell membrane of B-lymphocytes.
  • CD22 antigen appears early in B-cell lymphocyte differentiation at approximately the same stage as the CD19 antigen. Unlike other B-cell markers, CD22 membrane expression is limited to the late differentiation stages comprised between mature B cells (CD22+) and plasma cells (CD22-).
  • the CD22 antigen is described, for example, in Wilson et al, J. Exp. Med., 173: 137 (1991) and Wilson et al, J. Immunol. 150:5013 (1993).
  • B cell specific agent and "an agent that targets B cells” refer to an immunomodulatory agent that specifically targets a B cell by modifying, altering, reducing or inhibiting B cell function, e.g. by reducing or preventing a humoral response elicited by the B cell.
  • the B cell specific agent preferably is able to deplete B cells ⁇ i.e. reduce circulating B-cell levels) in a mammal treated therewith. Such depletion may be achieved via various mechanisms such as ADCC and/or CDC, inhibition of B-cell proliferation and/or induction of B-cell death ⁇ e.g. via apoptosis) or inhibiting or interfering with B cell
  • an agent of the invention may be described as “an agent that arrests the growth of B cells” "a B cell growth inhibitory agent,” “an agent that arrests B cell
  • the agent used is not critical to the invention and can be, for example, a polypeptide ⁇ e.g., antibodies or antibody fragments), a nucleic acid ⁇ e.g., inhibitory nucleic acids such as siRNA) or a small molecule.
  • the B cells targeted by the invention are B cells expressing CD20.
  • B cell specific agent(s) can be screened by various methods known in the art for apoptosis and other measurements for the depletion, and retardation or stopping of proliferation and growth of B cells or survival of B cells.
  • Exemplary antibodies include rituximab, ocrelizumab, ofatumumab, and veltuzumab, all of which are described in the prior art.
  • a method of screening can be employed as described in Sundberg et al, Cancer Research 66, 1775-1782 (2006) wherein a compound was screened for inhibition of B- cell proliferation by targeting c-myc protein for rapid and specific degradation. See also Mackay et al., Annual Review of Immunology, 21 : 231 -264 (2003) regarding BAFF, APRIL, and a tutorial on B-cell survival and screening, and Thangarajh et al., Scandinavian J. Immunol, 65(1 ):92 (2007) on B-cell proliferation and APRIL. In addition, Sakurai et al, European J.
  • B cell specific agents included within the scope of the present invention include antibodies, synthetic or native-sequence peptides, immunoadhesins, and small-molecule antagonists that bind to a B-cell surface marker or a B-cell specific survival or proliferation factor, optionally conjugated with or fused to another molecule.
  • the preferred B cell specific agent comprises an antibody or immunoadhesin. It includes BLyS antagonists such as immunoadhesins, and is preferably anti-CD23 (e.g., lumiliximab), anti-CD20, anti-CD22, or anti-BR3 antibodies, APRIL antagonists, and/or BLyS immunoadhesins.
  • the BLyS include antibodies, synthetic or native-sequence peptides, immunoadhesins, and small-molecule antagonists that bind to a B-cell surface marker or a B-cell specific survival or proliferation factor, optionally conjugated with or fused to another molecule.
  • the preferred B cell specific agent
  • immunoadhesin preferably is selected from the group consisting of BR3 immunoadhesin comprising the extracellular domain of BR3, TACI immunoadhesin comprising the extracellular domain of TACI, and BCMA immunoadhesin comprising the extracellular domain of BCMA.
  • the most preferred BR3 immunoadhesin is hBR3-Fc of SEQ ID NO:2 of WO 2005/00351 and US 2005/0095243. See also US 2005/0163775 and WO 2006/068867.
  • Another preferred BLyS B cell specific agent is an anti-BLyS antibody, more preferably wherein the anti-BLyS antibody binds BLyS within a region of BLyS comprising residues 162-275, or an anti-BR3 antibody, more preferably wherein the anti-BR3 antibody binds BR3 in a region comprising residues 23-38 of human BR3.
  • Especially preferred immunoadhesins herein are TACI-Ig, or atacicept, and BR3-Ig.
  • a preferred set of B cell specific agents are to CD20, CD22, BAFF, or APRIL.
  • the B cell specific agent may be, in one aspect, an antibody or TACI-Ig.
  • antibody 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.
  • An "isolated" antibody is one which has been identified and separated and/or recovered from a component of its natural environment. Contaminant components of its natural environment are materials which would interfere with research, diagnostic or therapeutic uses for the antibody, and may include enzymes, hormones, and other proteinaceous or nonproteinaceous solutes.
  • an antibody is purified (1 ) to greater than 95% by weight of antibody as determined by, for example, the Lowry method, and in some embodiments, to greater than 99% by weight; (2) to a degree sufficient to obtain at least 15 residues of N-terminal or internal amino acid sequence by use of, for example, a spinning cup sequenator, or (3) to homogeneity by SDS-PAGE under reducing or nonreducing conditions using, for example, Coomassie blue or silver stain.
  • Isolated antibody includes the antibody in situ within recombinant cells since at least one component of the antibody's natural environment will not be present. Ordinarily, however, isolated antibody will be prepared by at least one purification step.
  • “Native antibodies” and “native immunoglobulins” are usually heterotetrameric glycoproteins of about 150,000 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) followed by a number of constant domains.
  • VH variable domain
  • 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- and heavy-chain variable domains.
  • an "antibody B cell specific agent” or “antibody antagonist” herein is an antibody that, upon binding to a B-cell surface marker on B cells, destroys or depletes B cells in a mammal and/or interferes with one or more B-cell functions, e.g., by reducing or preventing a humoral response elicited by the B cell.
  • the antibody antagonist preferably is able to deplete B cells (i.e., reduce circulating B-cell levels) in a mammal treated therewith. Such depletion may be achieved via various mechanisms such as ADCC and/or CDC, inhibition of B-cell proliferation and/or induction of B-cell death (e.g., via apoptosis).
  • an "antibody that binds to a B-cell surface marker” or “antibody to a B-cell surface marker” is a molecule that, upon binding to a B-cell surface marker, destroys or depletes B cells in a mammal and/or interferes with one or more B-cell functions, e.g. by reducing or preventing a humoral response elicited by the B cell.
  • the antibody preferably is able to deplete B cells (i.e. reduce circulating B-cell levels) in a mammal treated therewith.
  • the antibody that binds to a B-cell surface marker may be designated as follows: an antibody that binds to CD20 or CD22 is an "anti-CD20 antibody” or "anti-CD22 antibody,” respectively.
  • the antibody is an anti-CD20, anti- CD22, anti-CD23, or anti-BR3 antibody.
  • Another embodiment typically used is an antibody that is an anti-CD20, anti-CD22, or anti-BR3 antibody.
  • Another typically used embodiment is an anti-CD20 or anti-CD22 antibody, and more, frequently used embodiment is the antibody that is an anti-CD20 antibody.
  • anti-CD20 antibodies examples include: “C2B8,” which is now called “rituximab” (“RITUXAN®/MABTHERA®”) (U.S. 5,736,137); the yttrium-[90]-labelled 2B8 murine antibody designated “Y2B8” or “Ibritumomab Tiuxetan” (ZEVALIN®) commercially available from Biogen pout Inc. (e.g., U.S. 5,736,137; 2B8 deposited with ATCC under accession no.
  • a 2H7 antibody e.g., WO 2004/056312 (Lowman et al.) and as set forth below
  • HUMAX-CD20TM fully human, high-affinity antibody targeted at the CD20 molecule in the cell membrane of B-cells (Genmab, Denmark; see, for example, Glennie and van de Winkel, Drug Discovery Today 8: 503-510 (2003) and Cragg et al., Blood 101 : 1045-1052 (2003))
  • the human monoclonal antibodies set forth in WO 2004/035607 and WO 2005/103081 Teeling et al, GenMab/Medarex
  • the antibodies having complex N-glycoside-linked sugar chains bound to the Fc region described in US 2004/0093621 hitara et al.
  • a chimerized or humanized monoclonal antibody having a high binding affinity to an extracellular epitope of a CD20 antigen described in WO
  • CD20-binding molecules such as the A E series of antibodies, e.g., AME-133TM antibodies as set forth, for example, in WO 2004/103404; US 2005/0025764; and US 2006/0251652 (Watkins et al, Applied Molecular Evolution, Inc.) and the anti-CD20 antibodies with Fc mutations as set forth, for example, in WO 2005/070963 (Allan et al, Applied Molecular Evolution, Inc.); CD20-binding molecules such as those described in WO 2005/016969 and US 2005/0069545 (Carr et al); bispecific antibodies as set forth, for example,
  • Amgen/AstraZeneca antibodies against CD20 as described, for example, in WO 2006/126069 (Morawala, Avestha Gengraine Technologies Pvt Ltd.); chimeric or humanized B-Lyl antibodies to CD20 ⁇ e.g., GA-101 ) as described, for example, in WO 2005/044859; US 2005/0123546; US 2004/0072290; and US 2003/0175884 (Umana et al; GlycArt Biotechnology AG); A20 antibody or variants thereof such as chimeric or humanized A20 antibody (cA20, hA20, respectively) and IMMUN-106 ⁇ e.g., US 2003/0219433, Immunomedics); and monoclonal antibodies L27, G28-2, 93-1 B3, B-C l or NU-B2 available from the International Leukocyte Typing Workshop ⁇ e.g., Valentine et al., In: Leukocyte Typing III (McMichael, Ed.,
  • the preferred anti-CD20 antibodies herein are chimeric, humanized, or human anti-CD20 antibodies, more preferably rituximab, a 2H7 antibody, chimeric or humanized A20 antibody (Immunomedics), and HUMAX-CD20TM human anti- CD20 antibody (Genmab).
  • anti-CD22 antibodies include those described in EP 1 ,476,120 (Tedder and Italiano), EP 1 ,485,130 (Tedder), and EP 1 ,504,035 (Popplewell et al), as well as those described in US 2004/0258682 (Leung et al), U.S. 5,484,892 (Dana-Farber), U.S. 6, 183,744 (Immunomedics, epratuzumab), and U.S. 7,074,403 (Goldenberg and Hansen).
  • antibodies to B-cell surface markers include rituximab, a 2H7 antibody and variants thereof as defined herein, 2F2 (HUMAX-CD20TM) (ofatumumab) human anti-CD20 antibody (an IgG l ⁇ human MAb that binds to a different CD20 epitope than rituximab), humanized A20 antibody veltuzumab (IMMUN-106TM or hA20), a humanized engineered antibody with complementarity-determining regions (CDRs) of murine origin and with 90% of the human framework regions identical to epratuzumab (a humanized anti-CD22 IgG l antibody); a small, modular immunopharmaceutical (SMIP) (herein called
  • variable region refers to the amino-terminal domains of the heavy or light chain of the antibody.
  • variable domain of the heavy chain may be referred to as "VH.”
  • variable domain of the light chain may be referred to as "VL.”
  • 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 concentrated in three segments called complementarity determining regions (CDRs) or 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 region (FR).
  • CDRs complementarity determining regions
  • FR framework region
  • the variable domains of native heavy and light chains each comprise four FR regions, largely adopting a ⁇ -sheet configuration, connected by three CDRs, which form loops connecting, and in some cases forming part of, the ⁇ -sheet structure.
  • the CDRs in each chain are held together in close proximity by the FRs and, with the CDRs from the other chain, contribute to the formation of the antigen-binding site of antibodies (see Kabat et al., N1H Publ. No.91 -3242, Vol. I, 647-669 [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 cytotoxicity.
  • the "light chains" of antibodies (immunoglobulins) from any vertebrate species can be assigned to one of two clearly distinct types, called kappa ( ⁇ ) and lambda ( ⁇ ), based on the amino acid sequences of their constant domains.
  • immunoglobulins can be assigned to different classes. There are five major classes of immunoglobulins: IgA, IgD, IgE, IgG, and IgM, and several of these may be further divided into subclasses (isotypes), e.g., IgG l , IgG2, IgG3, IgG4, IgA, and IgA2.
  • the heavy-chain constant domains that correspond to the different classes of immunoglobulins are called ⁇ , ⁇ , ⁇ , ⁇ and ⁇ , respectively.
  • the subunit structures and three-dimensional configurations of different classes of immunoglobulins are well known.
  • ADCC antigen-dependent cell-mediated cyotoxicity
  • CDC complement dependent cytotoxicity
  • This may be achieved by introducing one or more amino acid substitutions in an Fc region of the antibody.
  • cysteine residue(s) may be introduced in the Fc region, thereby allowing interchain disulfide bond formation in this region.
  • the homodimeric antibody thus generated may have improved internalization capability and/or increased complement-mediated cell killing and antibody-dependent cellular cytotoxicity (ADCC). See Caron et al., J. Exp Med.
  • Homodimeric antibodies with enhanced anti-tumor activity may also be prepared using heterobifunctional cross-linkers as described in Wolff et al. Cancer Research 53:2560-2565 ( 1993).
  • an antibody can be engineered which has dual Fc regions and may thereby have enhanced complement lysis and ADCC capabilities. See Stevenson et al. Anti-Cancer Drug Design
  • the agents which arrests the growth of, destroys or causes the deletion of cells expressing CD20 are antibodies.
  • the CD20 antigen used for production of antibodies may be, e.g., a soluble form of the extracellular domain of CD20 or a portion thereof, containing the desired epitope.
  • cells expressing CD20 at their cell surface can be used to generate antibodies.
  • Other forms of antigen useful for generating antibodies will be apparent to those skilled in the art.
  • 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, for example, maleimidobenzoyl
  • Animals are immunized against the antigen, immunogenic conjugates, or derivatives by combining, e.g., 100 ⁇ g or 5 ⁇ g of the protein or conjugate (for rabbits or mice, respectively) with 3 volumes of Freund's complete adjuvant and injecting the solution intradermally at multiple sites.
  • the animals are boosted with 1/5 to 1 /10 the original amount of peptide or conjugate in Freund's complete adjuvant 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.
  • 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 can be made in recombinant cell culture as protein fusions.
  • aggregating agents such as alum are suitably used to enhance the immune response.
  • 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 highly specific, being directed against a single antigenic site. Furthermore, in contrast to conventional (polyclonal) antibody preparations that 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 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.
  • the monoclonal antibodies to be used in accordance with the present invention may be 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 a!., J. Mol. Biol, 222:581 -597 (1991 ), for example.
  • 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.
  • the monoclonal antibodies to be used in accordance with the present invention may be made by a variety of techniques, including, for example, the hybridoma method (e.g., Kohler and Milstein., Nature, 256:495-97 (1975); Hongo et al, Hybridoma, 14(3):253-260 (1995), Harlow et al, Antibodies: A Laboratory Manual, (Cold Spring Harbor Laboratory Press, 2 nd ed. 1988); Hammerling et al , in: Monoclonal Antibodies and T-Cell Hybridomas, 563-681 (Elsevier, N.Y., 1981 )), recombinant DNA methods (see, e.g., U.S.
  • phage-display technologies see, e.g., Clackson et al., Nature, 352: 624-628 (1991 ); Marks et al, J. Mol. Biol, 222:581 -597 (1992); Sidhu et al, J. Mol. Biol, 338(2):299-310 (2004); Lee et al, J. Mol. Biol, 340(5): 1073- 1093 (2004); Fellouse, Proc. Natl. Acad. Sci. USA, 101 (34): 12467- 12472 (2004); and Lee et al, J.
  • 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 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 (U.S. Patent No. 4,816,567; Morrison et al., Proc. Natl. Acad. Sci. USA, 81 :6851 -6855 [ 1984]).
  • 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 recombinant DNA methods (U.S. Patent No. 4,816,567).
  • 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.
  • lymphocytes may be immunized in vitro. Lymphocytes 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)).
  • a suitable fusing agent such as polyethylene glycol
  • 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 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.
  • 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.
  • preferred myeloma cell lines are murine myeloma lines, such as those derived from MOPC-21 and MPC-1 1 mouse tumors available from the Salk Institute Cell Distribution Center, San Diego, California USA, 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 (Kozbor, J. Immunol., 133:3001 (1984); and 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 of monoclonal antibodies directed against the antigen.
  • 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).
  • RIA radioimmunoassay
  • ELISA enzyme-linked immunoabsorbent assay
  • the binding affinity of the monoclonal antibody can, for example, be determined by the Scatchard analysis of Munson et al., Anal. Biochem., 107:220 (1980).
  • 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 for this purpose include, for example, D-MEM or RPM1-1640 medium.
  • 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 antibody purification procedures such as, for example, protein A-Sepharose, hydroxylapatite 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.
  • the DNA 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 antibody protein, to obtain the synthesis of monoclonal antibodies in the recombinant host cells.
  • monoclonal 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 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 al., Bio/Technology,
  • the DNA also may be modified, for example, by substituting the coding sequence for human heavy chain and light chain constant domains in place of the homologous murine sequences (U.S. Patent No. 4,816,567; and Morrison, et al., Proc. Natl Acad. Sci. USA, 81 :6851 (1984)), or by covalently joining to the immunoglobulin coding sequence all or part of the coding sequence for a non-immunoglobulin polypeptide.
  • 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 one antigen-combining site having specificity for an antigen and another antigen-combining site having specificity for a different antigen.
  • Humanized forms of non-human (e.g., murine) antibodies are chimeric
  • immunoglobulins immunoglobulin chains or fragments thereof (such as Fv, Fab, Fab', F(ab') 2 or other antigen-binding subsequences of antibodies) which contain minimal sequence derived from non-human immunoglobulin.
  • humanized antibodies are human
  • immunoglobulins in which residues from a complementarity determining region (CDR) of the recipient are replaced by residues from a CDR of a non-human species (donor antibody) such as mouse, rat or rabbit having the desired specificity, affinity, and capacity.
  • CDR complementarity determining region
  • donor antibody non-human species
  • framework region (FR) residues of the human immunoglobulin are replaced by corresponding non-human residues.
  • humanized antibodies may comprise residues that are found neither in the recipient antibody nor in the imported CDR or framework sequences.
  • the humanized antibody will comprise substantially all of at least one, and typically two, variable domains, in which all or substantially all of the CDRs 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 optimally also will comprise at least a portion of an immunoglobulin constant region (Fc), typically that of a human immunoglobulin.
  • Fc immunoglobulin constant region
  • the humanized antibody includes a PRIMATIZED ® antibody wherein the antigen-binding region of the antibody is derived from an antibody produced by immunizing macaque monkeys with the antigen of interest. [0107] Methods for humanizing non-human antibodies have been described in the art.
  • a humanized antibody has one or more amino acid residues introduced into it from a source that 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 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.
  • humanized antibodies are typically human antibodies in which some hypervariable region residues and possibly some FR residues are substituted by residues from analogous sites in rodent antibodies.
  • variable domains both light and heavy
  • sequence of the variable domain of a rodent 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. 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. Natl. Acad. Sci. USA, 89:4285 (1992); Presta et al., J. Immunol., 151 :2623 (1993)).
  • humanized antibodies are prepared by a process of analysis of the parental sequences and various conceptual humanized products using 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 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.
  • 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.
  • the hypervariable region residues are directly and most substantially involved in influencing antigen binding.
  • the humanized or affinity matured antibody may be an antibody fragment, such as a Fab, which is optionally conjugated with one or more cytotoxic agent(s) in order to generate an immunoconjugate.
  • the humanized or affinity matured antibody may be an intact antibody, such as an intact IgG l antibody.
  • a "human antibody” is one which possesses an amino-acid sequence which
  • 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 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.
  • JH antibody heavy-chain joining region
  • phage display technology can be used to produce human antibodies and antibody fragments in vitro, from immunoglobulin variable (V) domain gene repertoires from unimmunized donors.
  • V domain genes are cloned in-frame into either a major or minor coat protein gene of a filamentous bacteriophage, such as M l 3 or fd, 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.
  • the 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).
  • 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 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, U.S. Patent Nos. 5,565,332 and 5,573,905.
  • Antibody fragments comprise a portion of an intact antibody, preferably the antigen binding or variable region of the intact antibody.
  • antibody fragments include Fab, Fab', F(ab')2, and Fv fragments; diabodies; linear antibodies (Zapata et al., Protein Eng.
  • 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.
  • the antibody of choice is a single chain Fv fragment (scFv). See WO 93/16185; U.S. Patent No. 5,571 ,894; and U.S. Patent No. 5,587,458.
  • the antibody fragment may also be a "linear antibody", e.g., as described in U.S. Patent 5,641 ,870 for example. Such linear antibody fragments may be monospecific or bispecific.
  • 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-combining sites and is still capable of cross-linking antigen.
  • 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 CH I 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 domains bear a 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.
  • Fv is the minimum antibody fragment which contains a complete antigen-recognition and -binding site. This region consists of a dimer of one heavy- and one light-chain variable domain in tight, non-covalent association. It is in this configuration that the three CDRs of 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. However, even a single variable domain (or half of an Fv comprising only three CDRs specific for an antigen) has the ability to recognize and bind antigen, although at a lower affinity than the entire binding site.
  • Single-chain Fv or “sFv” antibody fragments comprise the VH and VL domains of antibody, wherein these domains are present in a single polypeptide chain.
  • the Fv polypeptide further comprises a polypeptide linker between the VH and VL domains which enables the sFv to form the desired structure for antigen binding.
  • diabodies refers to small antibody fragments with two antigen-binding sites, which fragments comprise a heavy-chain variable domain (VH) connected to a light-chain variable domain (VL) in the same polypeptide chain (VH - VL).
  • VH heavy-chain variable domain
  • VL light-chain variable domain
  • Bispecific antibodies are antibodies that have binding specificities for at least two different epitopes. Exemplary bispecific antibodies may bind to two different epitopes of CD40 or CD20. Other such antibodies may combine a CD20 binding site with with a binding site which binds to an autoantigen associated with an autoimmune disease of interest. Alternatively, an anti-CD20 or anti-CD40 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.
  • FcyR Fc receptors for IgG
  • FcyR Fc receptors for IgG
  • FcyR FcyRI
  • CD32 FcyRII
  • FcyRIII CD 16
  • Antibodies with more than two valencies are contemplated.
  • trispecific antibodies can be prepared. Tutt et al. J. Immunol. 147: 60 ( 1991 ).
  • Binding affinity generally refers to the strength of the sum total of noncovalent interactions between a single binding site of a molecule (e.g. , an antibody) and its binding partner (e.g. , an antigen). Unless indicated otherwise, as used herein, "binding affinity” refers to intrinsic binding affinity which reflects a 1 : 1 interaction between members of a binding pair (e.g., antibody and antigen).
  • the affinity of a molecule X for its partner Y can generally be represented by the dissociation constant ( d). Affinity can be measured by common methods known in the art, including those described herein.
  • Low-affinity antibodies generally bind antigen slowly and tend to dissociate readily, whereas high-affinity antibodies generally bind antigen faster and tend to remain bound longer.
  • a variety of methods of measuring binding affinity are known in the art, any of which can be used for purposes of the present invention. Specific illustrative and exemplary embodiments for measuring binding affinity are described in the following.
  • the "Kd" or "Kd value” according to this invention is measured by a radiolabeled antigen-binding assay (RIA) performed with the Fab version of an antibody of interest and its antigen as described by the following assay.
  • RIA radiolabeled antigen-binding assay
  • Solution-binding affinity of Fabs for antigen is measured by equilibrating Fab with a minimal concentration of ( 125 I)-labeled antigen in the presence of a titration series of unlabeled antigen, then capturing bound antigen with an anti-Fab antibody-coated plate (see, e.g., Chen et al, J. Mol. Biol, 293:865-881 (1999)).
  • microtiter plates (DYNEX Technologies, Inc.) are coated overnight with 5 ⁇ g/ml of a capturing anti-Fab antibody (Cappel Labs) in 50 mM sodium carbonate (pH 9.6), and subsequently blocked with 2% (w/v) bovine serum albumin in PBS for two to five hours at room temperature (approximately 23°C).
  • a non-adsorbent plate (Nunc #269620) 100 pM or 26 pM [ 125 I]-anti gen are mixed with serial dilutions of a Fab of interest (e.g., consistent with assessment of the anti-VEGF antibody, Fab- 12, in Presta et al., Cancer Res., 57:4593-4599 (1997)).
  • the Fab of interest is then incubated overnight; however, the incubation may continue for a longer period (e.g., about 65 hours) to ensure that equilibrium is reached. Thereafter, the mixtures are transferred to the capture plate for incubation at room temperature (e.g., for one hour).
  • the Kd or Kd value is measured by using surface- plasmon resonance assays using a BIACORE ® -2000 or a BIACORE ® -3000 instrument
  • CM5 carboxymethylated dextran biosensor chips
  • EDC N-ethyl-N'- (3-dimethylaminopropyl)-carbodiimide hydrochloride
  • NHS N- hydroxysuccinimide
  • Antigen is diluted with 10 mM sodium acetate, pH 4.8, to 5 ⁇ g/ml (-0.2 ⁇ ) before injection at a flow rate of 5 ⁇ /minute to achieve approximately ten response units (RU) of coupled protein.
  • an "on-rate,” “rate of association,” “association rate,” or “k on” can also be determined as described above using a BIACORE ® -2000 or a BIACORE ® - 3000 system (BIAcore, Inc., Piscataway, NJ).
  • the term "substantially similar” or “substantially the same,” as used herein, denotes a sufficiently high degree of similarity between two numeric values (for example, one associated with an antibody of the invention and the other associated with a reference/comparator antibody), such that one of skill in the art would consider the difference between the two values to be of little or no biological and/or statistical significance within the context of the biological characteristic measured by said values (e.g., Kd values).
  • the difference between said two values is, for example, less than about 50%, less than about 40%, less than about 30%, less than about 20%, and/or less than about 10% as a function of the reference/comparator value.
  • the phrase "substantially reduced,” or “substantially different,” as used herein, denotes a sufficiently high degree of difference between two numeric values (generally one associated with a molecule and the other associated with a reference/comparator molecule) such that one of skill in the art would consider the difference between the two values to be of statistical significance within the context of the biological characteristic measured by said values (e.g., Kd values).
  • the difference between said two values is, for example, greater than about 10%, greater than about 20%, greater than about 30%, greater than about 40%, and/or greater than about 50% as a function of the value for the reference/comparator molecule.
  • the immunosuppressive or immunomodulatory agent is an inhibitory nucleic acid that inhibits expression of a target gene in B cells or other lymphocytes and includes, for example, short interfering RNA (siRNA; e.g., double-stranded ribonucleic acid (dsRNA) or small hairpin RNA (shRNA)) and short interfering DNA/RNA (siD/R-NA; e.g. double-stranded chimera of DNA and RNA (dsD/R-NA) or small hairpin chimera of DNA and RNA (shD/R-NA)).
  • siRNA short interfering RNA
  • dsRNA double-stranded ribonucleic acid
  • shRNA small hairpin RNA
  • siD/R-NA short interfering DNA/RNA
  • siRNA refers to a double-stranded RNA molecule that prevents translation of a target B cell mRNA. Standard techniques of introducing siRNA into the cell are used, including those in which DNA is a template from which RNA is transcribed.
  • the siRNA includes a part of sense nucleic acid sequence of the target gene (also referred to as "sense strand"), a part of antisense nucleic acid sequence of the target gene (also referred to as "antisense strand") or both.
  • the siRNA may be constructed such that a single transcript has both the sense and complementary antisense nucleic acid sequences of the target gene, e.g., a hairpin.
  • the siRNA may either be a dsRNA or shRNA.
  • dsRNA refers to a construct of two RNA molecules composed of complementary sequences to one another and that have annealed together via the complementary sequences to form a double-stranded RNA molecule.
  • the nucleotide sequence of two strands may include not only the "sense” or "antisense” RNAs selected from a protein coding sequence of target gene sequence, but also RNA molecule having a nucleotide sequence selected from non-coding region of the target gene.
  • shRNA refers to an siRNA having a stem-loop structure, composed of first and second regions complementary to one another, i.e., sense and antisense strands. The degree of complementarity and orientation of the regions are sufficient such that base pairing occurs between the regions, the first and second regions are joined by a loop region, and the loop results from a lack of base pairing between nucleotides (or nucleotide analogs) within the loop region.
  • the loop region of an shRNA is a single-stranded region intervening between the sense and antisense strands and may also be referred to as "intervening single- strand".
  • the immunosuppressive or immunomodulatory agents can be administered in a variety of ways, depending upon the agent, the particular disease being treated, the patient, and the like.
  • the agents or compositions of the present invention can be used to treat and/or prevent autoimmune disease in subjects or patients including human and any other mammal including, but not limited to, mouse, rat, guinea-pig, rabbit, cat, dog, sheep, goat, pig, cattle, horse, monkey, baboon, and chimpanzee, particularly a commercially important animal or a domesticated animal.
  • the immunosuppressive or immunomodulatory agents are conveniently formulated in pharmaceutical compositions which can optionally include other therapeutic substances, in particular, the DNA antigen specific therapy agents described, below.
  • the formulations may also include other agents such as, anti-inflammatory agents, pain killers, and the like.
  • the immunosuppressive or immunomodulatory agents and DNA antigen specific therapy agents of the invention can be administered sequentially or simultaneously with the one or more other pharmacologic agents.
  • the amounts of medicament and pharmacologic agent depend, for example, on what type of pharmacologic agent(s) is/are used, the disease being treated, and the scheduling and routes of administration.
  • compositions of this invention can include other agents
  • the immunosuppressive or immunomodulatory agents and/or DNA antigen specific therapy agents can be included in articles of manufacture and kits containing materials useful for treating autoimmune diseases.
  • the article of manufacture can include a container of any of the present pharmaceutical compositions with a label. Suitable containers include bottles, vials, and test tubes. The containers can be formed from a variety of materials, such as glass or plastic.
  • the label on the container should indicate the agent is used for treating or prevention of one or more conditions of the disease.
  • the label can also indicate directions for administration and so on.
  • the immunosuppressive or immunomodulatory agents of the invention can be administered directly as a pharmaceutical composition, or if necessary, that has been formulated by conventional formulation methods.
  • pharmaceutical compositions of the invention will include pharmaceutically acceptable, carriers, excipients, and such that are ordinarily used for drugs can be included as appropriate without particular limitations. Examples of such carriers are sterilized water, physiological saline, phosphate buffer, culture fluid and such.
  • the pharmaceutical agents or compositions can contain as necessary, stabilizers, suspensions, preservatives, surfactants and such.
  • the pharmaceutical agents or compositions of this invention can be used for anticancer purposes.
  • a "medicament" is an active drug to treat an autoimmune disease or the signs or symptoms or side effects of an autoimmune disease.
  • pharmaceutical formulation refers to a sterile preparation that is in such form as to permit the biological activity of the medicament to be effective, and which contains no additional components that are unacceptably toxic to a subject to which the formulation would be administered.
  • a "sterile" formulation is aseptic or free from all living microorganisms and their spores.
  • a "package insert” is used to refer to instructions customarily included in commercial packages of therapeutic products or medicaments, that contain information about the indications, usage, dosage, administration, contraindications, other therapeutic products to be combined with the packaged product, and/or warnings concerning the use of such therapeutic products or medicaments, etc.
  • a "kit” is any manufacture (e.g a package or container) comprising at least one reagent, e.g., a medicament for treatment of an autoimmune disease, or a probe for specifically detecting a biomarker gene or protein of the invention.
  • the manufacture is preferably promoted, distributed, or sold as a unit for performing the methods of the present invention.
  • a "target audience” is a group of people or an institution to whom or to which a particular medicament is being promoted or intended to be promoted, as by marketing or advertising, especially for particular uses, treatments, or indications, such as individual patients, patient populations, readers of newspapers, medical literature, and magazines, television or internet viewers, radio or internet listeners, physicians, drug companies, etc.
  • sample shall generally mean any biological sample obtained from an individual, body fluid, body tissue, cell line, tissue culture, or other source.
  • Body fluids are, e.g., lymph, sera, whole fresh blood, peripheral blood mononuclear cells, frozen whole blood, plasma (including fresh or frozen), urine, saliva, semen, synovial fluid and spinal fluid. Samples also include synovial tissue, skin, hair follicle, and bone marrow. Methods for obtaining tissue biopsies and body fluids from mammals are well known in the art. If the term “sample” is used alone, it shall still mean that the "sample” is a "biological sample", i.e., the terms are used interchangeably.
  • sample shall generally mean any serum sample obtained from an individual. Methods for obtaining sera from mammals are well known in the art.
  • medication(s) includes the situations in which a subject or patient has progressed while receiving the medicament(s) that he or she is being given, and in which a subject or patient has progressed within 12 months (for example, within six months) after completing a regimen involving the medicament(s) to which he or she is no longer responsive.
  • the non-responsiveness to one or more medicaments thus includes subjects who continue to have active disease following previous or current treatment therewith. For instance, a patient may have active disease activity after about one to three months of therapy with the medicament(s) to which they are non-responsive. Such responsiveness may be assessed by a clinician skilled in treating the autoimmune disorder in question.
  • a subject who experiences "a clinically unacceptably high level of toxicity" from previous or current treatment with one or more medicaments experiences one or more negative side-effects or adverse events associated therewith that are considered by an experienced clinician to be significant, such as, for example, serious infections, congestive heart failure, demyelination (leading to multiple sclerosis), significant hypersensitivity, neuropathological events, high degrees of autoimmunity, a cancer such as endometrial cancer, non-Hodgkin's lymphoma, breast cancer, prostate cancer, lung cancer, ovarian cancer, or melanoma, tuberculosis (TB), etc.
  • a cancer such as endometrial cancer, non-Hodgkin's lymphoma, breast cancer, prostate cancer, lung cancer, ovarian cancer, or melanoma, tuberculosis (TB), etc.
  • reducing the risk of a negative side effect is meant reducing the risk of a side effect resulting from treatment with the antagonist herein to a lower extent than the risk observed resulting from treatment of the same patient or another patient with a previously administered medicament.
  • side effects include those set forth above regarding toxicity, and are preferably infection, cancer, heart failure, or demyelination.
  • correlate or “correlating” is meant comparing, in any way, the performance and/or results of a first analysis or protocol with the performance and/or results of a second analysis or protocol. For example, one may use the results of a first analysis or protocol in carrying out a second protocols and/or one may use the results of a first analysis or protocol to determine whether a second analysis or protocol should be performed. With respect to various
  • Each agent comprising the combination therapy will be formulated, dosed, and administered in a fashion consistent with good medical practice.
  • Factors for consideration in this context include the particular type of autoimmune disease being treated, the particular mammal being treated, the clinical condition of the individual patient, the cause of the autoimmune disease, the site of delivery of the agent, possible side-effects, the type of agent, i.e.
  • the effective amount of the agent to be administered will be governed by such considerations.
  • the term "therapeutically effective amount” is used to refer to an amount of an active immunosuppressive or immunomodulatory agent, such as a B cell specific agent having a growth arrest effect, differentition inhibitory effect or causes the deletion of the B cell.
  • the therapeutically effective amount refers to a target serum concentration that has been shown to be effective in, for example, slowing disease progression. Efficacy can be measured in conventional ways, depending on the condition to be treated. A physician having ordinary skill in the art can readily determine and prescribe the effective amount of the pharmaceutical composition of each agent required, depending on such factors as the particular agent, i.e. B cell specific agent or DNA antigen specific therapeutic agent, and safety profile.
  • the physician could start with doses of B cell specific agent, such as an anti-CD20 or anti-CD22 antibody or immunoadhesin, employed in the pharmaceutical composition at levels lower than that required to achieve the desired therapeutic effect to assess safety, and gradually increase the dosage until the desired effect (without compromising safety) is achieved with subsequent administration of the DNA antigen specific therapeutic agent.
  • B cell specific agent such as an anti-CD20 or anti-CD22 antibody or immunoadhesin
  • the effectiveness of a given dose or treatment regimen of the agent can be determined, for example, by assessing signs and symptoms in the patient using the standard measures of efficacy for a particular autoimmune disease.
  • the effective amount of the B cell specific agent administered parenterally per dose will be in the range of about 20 mg to about 5000 mg, by one or more dosages.
  • Exemplary dosage regimens for intact antibodies include 375 mg/m 2 weekly x 4 (e.g., on days 1 , 8, 15, and 22); or 500 mg x 2 (e.g., on days 1 and 15), or 1000 mg x 2 (e.g., on days 1 and 15); or 1 gram x 3 (e.g., on days 1 , 15, and 21 ); or 200 mg x 1 -4; or 300 mg x 1 -4, or 400 mg x 1 -4; or 500 mg x 3-4; or 1 gram x 4.
  • the B cell specific agent is administered in a dose of about 0.2 to 4 grams, more preferably about 0.2 to 3.5 grams, more preferably about 0.4 to 2.5 grams, more preferably about 0.5 to 1.5 grams, and even more preferably about 0.7 to 1 .1 gram. More preferably, such doses apply to antagonists that are antibodies or immunoadhesins.
  • the B cell specific agent is an anti-CD20 antibody administered at a dose of about 1000 mg x 2 on days 1 and 15 intravenously at the start of the treatment.
  • the anti-CD20 antibody is administered as a single dose or as two infusions, with each dose at about 200 mg to 1.2 g, more preferably about 200 mg to 1.1 g, and still more preferably about 200 mg to 900 mg.
  • the B-cell specific agent is an anti-CD20 antibody administered at a dose of about 1000 mg x 2 on days 1 and 1 5 intravenously at the start of the treatment.
  • the anti-CD20 antibody is administered as a single dose or as two infusions, with each dose at about 200 mg to 600 mg.
  • the B cell specific agent is administered at a frequency of one to four doses within a period of about one month.
  • the antagonist is preferably administered in two to three doses.
  • the antagonist is preferably administered within a period of about two to three weeks.
  • intravenous infusion refers to introduction of an agent into the vein of an animal or human patient over a period of time greater than approximately 15 minutes, generally between approximately 30 to 90 minutes.
  • intravenous bolus or “intravenous push” refers to drug administration into a vein of an animal or human such that the body receives the drug in approximately 15 minutes or less, generally 5 minutes or less.
  • subcutaneous administration refers to introduction of an agent under the skin of an animal or human patient, preferable within a pocket between the skin and underlying tissue, by relatively slow, sustained delivery from a drug receptacle.
  • the pocket may be created by pinching or drawing the skin up and away from underlying tissue.
  • mammal for purposes of treatment refers to any animal classified as a mammal, including humans, domestic and farm animals, and zoo, sports, or pet animals, such as dogs, horses, cats and cows to name but a few.
  • the term "package insert” is used to refer to instructions customarily included in commercial packages of therapeutic products, that contain information about the indications, usage, dosage, administration, contraindications and/or warnings concerning the use of such therapeutic products.
  • the term "subcutaneous infusion” refers to introduction of a drug under the skin of an animal or human patient, preferably within a pocket between the skin and underlying tissue, by relatively slow, sustained delivery from a drug receptacle for a period of time including, but not limited to, 30 minutes or less, or 90 minutes or less.
  • the infusion may be made by subcutaneous implantation of a drug delivery pump implanted under the skin of the animal or human patient, wherein the pump delivers a predetermined amount of drug for a predetermined period of time, such as 30 minutes, 90 minutes, or a time period spanning the length of the treatment regimen.
  • a drug delivery pump implanted under the skin of the animal or human patient, wherein the pump delivers a predetermined amount of drug for a predetermined period of time, such as 30 minutes, 90 minutes, or a time period spanning the length of the treatment regimen.
  • subcutaneous bolus refers to drug administration beneath the skin of an animal or human patient, where bolus drug delivery is preferably less than approximately 15 minutes, more preferably less than 5 minutes, and most preferably less than 60 seconds.
  • Administration is preferably within a pocket between the skin and underlying tissue, where the pocket.
  • DNA antigen specific therapeutic agent refers to a polynucleotide that is administered to a subject for the purpose of modulating an immune response.
  • DNA tolerization refers to the administration of polynucleotides encoding one or more self-polypeptides that include one or more autoantigenic epitopes associated with a disease.
  • the "DNA tolerization” or “DNA vaccination” serves the purpose of modulating an ongoing immune response to suppress autoimmune destruction for the treatment or prevention of an autoimmune disease. Modulation of an immune response in reaction to "DNA vaccination” may include shifting self-reactive lymphocytes from a Th l - to a Th2-type response. The modulation of the immune response may occur systemically or only locally at the target organ under autoimmune attack.
  • polynucleotide and nucleic acid refer to a polymer composed of a multiplicity of nucleotide units (ribonucleotide or deoxyribonucleotide or related structural variants) linked via phosphodiester bonds.
  • a polynucleotide or nucleic acid can be of substantially any length, typically from about six (6) nucleotides to about 109 nucleotides to about 4000 nucleotides or larger.
  • Polynucleotides and nucleic acids include RNA, DNA, synthetic forms, and mixed polymers, both sense and antisense strands, double- or single- stranded, and can also be chemically or biochemically modified or can contain non-natural or derivatized nucleotide bases, as will be readily appreciated by the skilled artisan.
  • Such modifications include, for example, labels, methylation, substitution of one or more of the naturally occurring nucleotides with an analog, intemucleotide modifications such as uncharged linkages (e.g., methyl phosphonates, phosphotriesters, phosphoamidates, carbamates, and the like), charged linkages (e.g., phosphorothioates, phosphorodithioates, and the like), pendent moieties (e.g., polypeptides), intercalators (e.g., acridine, psoralen, and the like), chelators, alkylators, and modified linkages (e.g., alpha anomeric nucleic acids, and the like).
  • uncharged linkages e.g., methyl phosphonates, phosphotriesters, phosphoamidates, carbamates, and the like
  • charged linkages e.g., phosphorothioates, phosphorodithioates, and the like
  • synthetic molecules that mimic polynucleotides in their ability to bind to a designated sequence via hydrogen bonding and other chemical interactions.
  • Such molecules are known in the art and include, for example, those in which peptide linkages substitute for phosphate linkages in the backbone of the molecule.
  • promoter is used here to refer to the polynucleotide region recognized by RNA polymerases for the initiation of RNA synthesis, or “transcription”. Promoters are one of the functional elements of self-vectors that regulate the efficiency of transcription and thus the level of protein expression of a self-polypeptide encoded by a self-vector. Promoters can be “constitutive”, allowing for continual transcription of the associated gene, or “inducible”, and thus regulated by the presence or absence of different substances in the environment.
  • promoters can also either be general, for expression in a broad range of different cell types, or cell-type specific, and thus only active or inducible in a particular cell type, such as a muscle cell.
  • Promoters controlling transcription from vectors may be obtained from various sources, for example, the genomes of viruses such as: polyoma, simian virus 40 (SV40), adenovirus, retroviruses, hepatitis B virus and preferably cytomegalovirus, or from heterologous mammalian promoters, e.g., b-actin promoter.
  • the early and late promoters of the SV40 virus are conveniently obtained as is the immediate early promoter of the human cytomegalovirus.
  • Enhancer refers to cis-acting polynucleotide regions of about from 10-300 basepairs that act on a promoter to enhance transcription from that promoter. Enhancers are relatively orientation and position independent and can be placed 5' or 3' to the transcription unit, within introns, or within the coding sequence itself.
  • intron refers to intervening polynucleotide sequences within a gene or portion of a gene present in a self-vector that is situated upstream of or between "exons", polynucleotide sequences that are retained during RNA processing and most often code for a polypeptide. Introns do not function in coding for protein synthesis and are spliced out of a RNA before it is translated into a polypeptide.
  • RNA molecule refers to the mechanism by which a single functional RNA molecule is generated by the removal of introns and juxtaposition of exons during processing of the primary transcript, or preRNA.
  • Consensus sequences are present at intron-exon junctions that define the 5' end, or donor site, of an intron and the 3' end, or acceptor site, and at a branchpoint site located approximately 20-50 basepairs upstream of the acceptor site within the intron sequence.
  • Most introns start from the sequence GU and end with the sequence AG (in the 5' to 3' direction) with a branchpoint site approximating CU(A/G)A(C/U), where A is conserved in all genes. These sequences signal for the looping out of the intron and its subsequent removal.
  • a "terminator sequence” as used herein means a polynucleotide sequence that signals the end of DNA transcription to the RNA polymerase. Often the 3' end of a RNA generated by the terminator sequence is then processed considerably upstream by polyadenylation.
  • Polyadenylation is used to refer to the non-templated addition of about 50 to about 200 nucleotide chain of polyadenylic acid (polyA) to the 3' end of a transcribed messenger RNA.
  • polyA polyadenylic acid
  • the "polyadenylation signal” (AAUAAA) is found within the 3' untranslated region (UTR) of a mRNA and specifies the site for cleavage of the transcript and addition of the polyA tail.
  • Transcription termination and polyadenylation are functionally linked and sequences required for efficient cleavage/polyadenylation also constitute important elements of termination sequences (Connelly and Manley, 1988).
  • Self-vector means one or more vector(s) which taken together comprise a polynucleotide either DNA or RNA encoding one or more self-protein(s), -polypeptide(s), -peptide(s) (referred to herein as “DNA tolerizing vector”, “DNA tolerizing plasm id” “DNA self-vector” or “RNA self-vector” respectively).
  • a DNA tolerizing vector or DNA tolerizing plasmid when used in the context of a particular autoimmune disease may include the particular autoantigen in the description, for example in the case of Tl D the DNA tolerizing plasmid may be referred to as “Proinsulin tolerizing plasmid (PTP)" when proinsulin is the autoantigen encoded by the self-vector.
  • PTP Proinsulin tolerizing plasmid
  • MTP Myelin basic protein tolerizing plasmid
  • Polynucleotide as used herein is a series of either deoxyribonucleic acids including DNA or ribonucleic acids including RNA, and their derivatives, encoding a self-protein, -polypeptide, or -peptide of this invention.
  • the self- protein, -polypeptide or -peptide coding sequence is inserted into an appropriate plasmid expression self-cassette.
  • the vector is then referred to as a "self- vector."
  • self- vector In the case where polynucleotide encoding more than one self-protein(s),
  • a single self-vector may encode multiple separate self -protein(s), -polypeptide(s) or -peptide(s).
  • DNA encoding several self-protein(s), -polypeptide(s), or -peptide(s) are encoded sequentially in a single self- plasmid utilizing internal ribosomal re-entry sequences (IRES) or other methods to express multiple proteins from a single DNA molecule.
  • IRS internal ribosomal re-entry sequences
  • the DNA expression self-vectors encoding the self-protein(s), -polypeptide(s), or -peptide(s) are prepared and isolated using commonly available techniques for isolation of plasmid DNA such as those commercially available from Qiagen Corporation.
  • the DNA is purified free of bacterial endotoxin for delivery to humans as a therapeutic agent.
  • each self-protein, -polypeptide or -peptide is encoded on a separate DNA expression vector.
  • a "high expression self-vector” or “HESV” refers herein to a modified self-vector that is altered to increase expression of an encoded self-protein(s), -polypeptide(s), or -peptide(s) relative to an unmodified self-vector encoding the same self-protein(s), -polypeptide(s), or - peptide(s).
  • a HESV comprises a polynucleotide encoding and capable of expressing a self- polypeptide associated with an autoimmune disease and a modification to generate increased expression of the self-polypeptide relative to the same self-vector unmodified.
  • a HESV further comprises in operative combination: a promoter; a polynucleotide encoding a self-polypeptide that includes at least one autoantigenic epitope associated with the autoimmune disease; a transcription terminator; and at least one modification for generating increased expression of the self-polypeptide in a host cell, in which the increased expression is relative to an unmodified self-vector comprising the promoter, polynucleotide, and transcription terminator.
  • Modifications of a self-vector to generate a HESV with increased expression of a self-polypeptide are selected from alterations that increase: transcription initiation, transcription termination, mRNA stability, translation efficiency, and/or protein stability.
  • modifications of a self-vector to increase expression of a self-polypeptide are selected from the group consisting of: using a stronger promoter region, addition of enhancer regions, using a more efficient transcription terminator sequence, addition of polyadenylation signals, using a more ideal consensus kozak sequence, optimizing codon usage, inclusion of introns or combinations of the foregoing modifications.
  • Single or multiple modifications may be incorporated into a self-vector to generate a HESV.
  • the modification is the inclusion of an intron downstream of the promoter region and upstream of the start codon of a polynucleotide encoding one or more self-polypeptides.
  • the preferred intron is intron A of the human cytomegalovirus (CMV) or a ⁇ -globin/Ig chimeric intron and most preferably the preferred intron is the ⁇ -globin/Ig chimeric intron.
  • An HESV is generated that expresses increased amounts of a self-protein(s), -polypeptide(s), or peptide(s) associated with an autoimmune disease, such as insulin-dependent diabetes mellitus (T1 D), multiple sclerosis (MS), systemic lupus erythematosus (SLE), or rheumatoid arthritis (RA) compared to an unmodified self-vector encoding the same self-protein(s), -polypeptide(s), or peptide(s).
  • T1 D insulin-dependent diabetes mellitus
  • MS multiple sclerosis
  • SLE systemic lupus erythematosus
  • RA rheumatoid arthritis
  • a HESV is generated that expresses increased amounts of the self-polypeptide preproinsulin compared to the unmodified self-vector.
  • An HESV is generated that expresses increased amounts of a self- polypeptide associated with the autoimmune disease multiple sclerosis (MS) compared to an unmodified self-vector encoding the same self-polypeptide. More particularly the HESV is generated that expresses increased amounts of the self-polypeptide myelin basic protein (MBP) compared to the unmodified self-vector.
  • MBP myelin basic protein
  • a HESV contains a ⁇ - globin/Ig chimeric intron 5' to the start codon of the self-polypeptide MBP.
  • a "non-secreted self-vector” or “N-SSV” or “non-secreted self-vector specific agent” refers herein to a modified self-vector that contains a polynucleotide encoding for an intracellular or non-secreted self-polypeptide version of a extracellular or secreted autoantigen (e.g., a transmembrane protein or secreted soluble factor) associated with an autoimmune disease.
  • a extracellular or secreted autoantigen e.g., a transmembrane protein or secreted soluble factor
  • a N- SSV comprises a polynucleotide encoding and capable of expressing a secreted self-polypeptide associated with an autoimmune disease and a modification to express a non-secreted or non- membrane bound self-polypeptide from a host cell.
  • a N-SSV further comprises in operative combination: a promoter; a polynucleotide encoding an extracellular or secreted self-polypeptide that includes at least one autoantigenic epitope associated with the autoimmune disease; a transcription terminator; and at least one modification to prevent secretion of the self-polypeptide from a host cell relative to an unmodified self-vector comprising the promoter, polynucleotide, and transcription terminator.
  • Modifications to a self-vector to generate a N-SSV encoding and expressing a non-secreted or non-membrane bound version of a secreted or membrane bound self-polypeptide include but are not limited to eliminating the signal sequence, mutating the signal sequence, and adding alternative protein localization (ER retention, plasma membrane attachment, etc.) protein degradation signals or modifying or deleting, transmembrane domains or hydrophobic regions of the self-polypeptide.
  • a "non-secreted high expression self-vector” or “N-SHESV” or “non-secreted high expression self-vector specific agent” refers to a modified self-vector that is altered to increase expression of an encoded intracellular or non-secreted version of an extracellular or secreted self-polypeptide or non-membrane bound version of a membrane bound self-polypeptide in which expression and secretion is relative to an unmodified self-vector.
  • a N-SHESV specific agent comprises a polynucleotide encoding and capable of expressing a secreted or membrane bound self-polypeptide associated with an autoimmune disease and a modification to generate increased expression of the self-polypeptide in a non-secreted or non-membrane bound form relative to the unmodified self-vector.
  • a N-SHESV further comprises in operative combination: a promoter; a polynucleotide encoding a extracellular or secreted self-polypeptide that includes at least one autoantigenic epitope associated with the autoimmune disease; a transcription terminator; and at least one modification for generating increased expression of the self- polypeptide and at least one modification to express the non-secreted or non-membrane bound self-polypeptide from a host cell where both modifications are relative to an unmodified self- vector comprising the promoter, polynucleotide, and transcription terminator.
  • a "secreted self-vector” or “SSV” or “secreted self-vector specific agent” refers herein to a modified self-vector that contains a polynucleotide encoding a secreted self-polypeptide version of a membrane associated or intracellular autoantigen associated with an autoimmune disease.
  • a SSV comprises a polynucleotide encoding and capable of expressing a membrane associated or intracellular self-polypeptide associated with an autoimmune disease and a modification to allow secretion of the self-polypeptide from a host cell.
  • a SSV comprises a polynucleotide encoding and capable of expressing a membrane associated or intracellular self- polypeptide associated with an autoimmune disease and a modification to allow secretion of the self-polypeptide from a host cell.
  • a SSV further comprises in operative combination: a promoter; a polynucleotide encoding a membrane associated or intracellular self-polypeptide that includes at least one autoantigenic epitope associated with the autoimmune disease; a
  • Modifications to a self-vector to generate a SSV encoding and expressing a secreted version of an intracellular self-polypeptide include, but are not limited to, addition of a signal sequence. Additionally, the modification may further include signals for membrane association including, for example, a transmembrane domain or a GPI anchor so that intracellular epitope(s) are presented extracellularly.
  • Modifications to a self-vector to generate a SSV encoding and expressing a secreted version of a membrane associated self-polypeptide include but are not limited to: removal of a transmembrane domain; removal of a GPI linkage, removal of an extracellular and transmembrane domain with addition of a signal sequence to an intracellular domain; and removal of a transmembrane domain and intracellular domain.
  • a "secreted high expression self-vector” or “SHESV” or “secreted high expression self- vector specific agent” as used herein refers to a modified self-vector that is altered to increase expression of an encoded secreted version of a membrane associated or intracellular self- polypeptide in which expression and secretion is relative to an unmodified self-vector.
  • a SHESV comprises a polynucleotide encoding and capable of expressing a membrane associated or intracellular self-polypeptide associated with an autoimmune disease and a modification to generate increased expression of the self-polypeptide in a secreted or extracellular membrane associated form relative to the same self-vector unmodified.
  • a SHESV further comprises in operative combination: a promoter; a polynucleotide encoding a membrane associate or intracellular self-polypeptide that includes at least one autoantigenic epitope associated with the autoimmune disease; a transcription terminator; and at least one modification for generating increased expression of the self-polypeptide and at least one modification to allow secretion of the self-polypeptide from a host cell where both modifications are relative to an unmodified self- vector comprising the promoter, polynucleotide, and transcription terminator.
  • vector backbone refers to the portion of a plasmid vector other than the sequence encoding a self-antigen, -protein, -polypeptide, or -peptide.
  • an "immunosuppressive vector backbone” refers to a vector backbone that either (i) elicits a reduced immune response in comparison to a parent vector backbone, or (ii) prevents or inhibits an immune response.
  • the immune response can be measured using in vitro or in vivo assays known in the art. For example, the immune response can be determined by measuring proliferation of lymphocytes exposed to the vector backbone, or by measuring production of cytokines (in cell culture media, in serum, etc.) indicative of immune stimulation ⁇ e.g., IL-2, lFN- ⁇ , IL-6).
  • an immunosuppressive vector backbone contains fewer immunostimulatory sequences (e.g., CpG sequences) in comparison to a parent vector backbone.
  • an immunosuppressive vector backbone contains one or more
  • an immunosuppressive vector backbone promotes a Th2 immune response and inhibits a Thl immune response.
  • the method for treating autoimmune disease further includes the administration of a polynucleotide comprising an inhibitory immune modulatory sequence or an immune inhibitory sequence (IIS).
  • IISs useful in accordance with the present invention comprise the following core hexamer:
  • X and Y are any naturally occurring or synthetic nucleotide, except that X and Y cannot be cytosine-guanine.
  • the core hexamer of IMSs can be flanked 5' and/or 3' by any composition or number of nucleotides or nucleosides.
  • IMSs range between 6 and 100 base pairs in length, and most preferably 16-50 base pairs in length.
  • IMSs can also be delivered as part of larger pieces of DNA, ranging from 100 to 100,000 base pairs.
  • IMSs can be incorporated in, or already occur in, DNA plasmids, viral vectors and genomic DNA. Most preferably IMSs can also range from 6 (no flanking sequences) to 10,000 base pairs, or larger, in size. Sequences present which flank the hexamer core can be constructed to substantially match flanking sequences present in any known immunoinhibitory sequences (IIS). For example, the flanking sequences
  • flanking sequences TTGACTGTG -Pu-Pyr-X-Y-Pyr-Pyr-AGAGATGA, where TTGACTGTG and AGAGATGA are flanking sequences.
  • Another preferred flanking sequence incorporates a series of pyrimidines (C, T, and U), either as an individual pyrimidine repeated two or more times, or a mixture of different pyrimidines two or more in length. Different flanking sequences have been used in testing inhibitory modulatory sequences. Further examples of flanking sequences for inhibitory oligonucleotides are contained in the following references: U.S. Patent Nos.
  • An IIS contains the core hexamer region having the sequence AAGGTT. That sequence is referred to herein as an immune modulatory sequence or IMS.
  • IMS immune modulatory sequence
  • dinucleotide cores GGGGTT, AGGGTT, GAGGTT, AAGGTT, GGGGCT, AGGGCT, GAGGCT, AAGGCT, GGGGTC, AGGGTC, GAGGTC,
  • dinucleotide cores GGGCTT, AGGCTT, GAGCTT, AAGCTT, GGGCCT, AGGCCT, GAGCCT, AAGCCT, GGGCTC, AGGCTC, GAGCTC,
  • the core hexamer region of the IMS is flanked at either the 5' or 3' end, or at both the 5' and 3' ends, by a polyG region.
  • a "polyG region” or “polyG motif as used herein means a nucleic acid region consisting of at least two (2) contiguous guanine bases, typically from 2 to 30 or from 2 to 20 contiguous guanines. In some embodiments, the polyG region has from 2 to 10, from 4 to 10, or from 4 to 8 contiguous guanine bases. In certain preferred embodiments, the flanking polyG region is adjacent to the core hexamer.
  • the polyG region is linked to the core hexamer by a non-polyG region (non-polyG linker); typically, the non-polyG linker region has no more than 6, more typically no more than 4 nucleotides, and most typically no more than 2 nucleotides.
  • the immune modulatory nucleic acids can contain IMSs alone or incorporated in cis or in trans with other nucleic acid regions such as, for example, into a recombinant self-vector (plasmid, cosmid, virus or retrovirus) which may in turn code for any self- protein(s), - polypeptide(s), or -peptide(s) deliverable by a recombinant expression vector.
  • the IMSs are incorporated into a vector such as, for example, an expression vector, which may be accomplished, for example, using conventional techniques as known to one of ordinary skill in the art (see, e.g., Ausubel, Current Protocols in Molecular Biology, supra).
  • construction of recombinant expression vectors employs standard ligation techniques.
  • the ligation mixtures may be used to transform a host cell and successful transformants selected by antibiotic resistance where appropriate.
  • Vectors from the transformants are prepared, analyzed by restriction and/or sequenced by, for example, the method of Messing, et al., Nucleic Acids Res., 9:309, 1981 , the method of Maxam, et al , Methods in Enzymology, 65:499, 1980, or other suitable methods which will be known to those skilled in the art. Size separation of cleaved fragments is performed using conventional gel electrophoresis as described, for example, by Maniatis, et al , Molecular Cloning, pp. 133-134, 1982.
  • Host cells may be transformed with the expression vectors of this invention and cultured in conventional nutrient media modified as is appropriate for inducing promoters, selecting transformants or amplifying genes.
  • the 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 ordinarily skilled artisan.
  • plasmids and cosmids are particularly preferred for their lack of pathogenicity.
  • plasmids and cosmids are subject to degradation in vivo more quickly than viruses and therefore may not deliver an adequate dosage of IMS-ON to prevent or treat an inflammatory or autoimmune disease.
  • a nucleic acid vector in which a non-CpG dinucleotide is substituted for one or more CpG dinucleotides of the formula 5'-punne-pyrimidine-C-G- pyrimidine-pyrimidine-3' or 5'-purine-purine-C-G-pyrimidine-pyrimidine-3', thereby producing a vector in which US-associated immunostimulatory activity is reduced.
  • Such vectors are useful, for example, in methods for administering immune modulatory nucleic acids and/or for administering a self vector encoding one or more self-protein(s), -polypeptides(s), or -peptide(s).
  • the cytosine of the CpG dinucleotide can be substituted with guanine, thereby yielding an IMS region having a GpG motif of the formula 5'-purine-pyrimidine-G-G- pyrimidine-pyrimidine-3' or 5'-purine-purine-G-G-pyrimidine-pyrimidine-3'.
  • the cytosine can also be substituted with any other non-cytosine nucleotide. The substitution can be
  • the substituted CpG motifs are those CpGs that are not located in important control regions of the vector ⁇ e.g., promoter regions).
  • the non-cytosine substitution is typically selected to yield a silent mutation or a codon corresponding to a conservative substitution of the encoded amino acid.
  • a modified pVAXl vector in which one or more CpG dinucleotides of the formula 5'-purine-pyrimidine-C-G-pyrimidine-pyrimidine- 3' is mutated by substituting the cytosine of the CpG dinucleotide with a non-cytosine nucleotide.
  • the pVAXl vector is known in the art and is commercially available from Invitrogen (Carlsbad, CA).
  • the modified pVAXl vector has the following cytosine to non-cytosine substitutions within a CpG motif: cytosine to guanine at nucleotides 784, 1 161 , 121 8, and 1966; cytosine to adenine at nucleotides 1264, 1337, 1829, 1874, 1940, and 1997; and cytosine to thymine at nucleotides 1 158, and 1987; with additional cytosine to guanine mutations at nucleotides 1831 , 1876, 1942, and 1999.
  • the nucleotide number designations as set forth above are according to the numbering system for pVAXl provided by Invitrogen.
  • the vector thus constructed was named pBHTl .
  • Nucleotide sequences selected for use in the self-vector can be derived from known sources, for example, by isolating the nucleic acid from cells containing a desired gene or nucleotide sequence using standard techniques. Similarly, the nucleotide sequences can be generated synthetically using standard modes of polynucleotide synthesis that are well known in the art. See, e.g. , Edge et al , Nature 292:756, 1981 ; Nambair et al , Science 223: 1299, 1984; Jay et al, J. Biol Chem. 259:631 1 , 1984. Generally, synthetic oligonucleotides can be prepared by either the phosphotriester method as described by Edge et al. ⁇ supra) and Duckworth et al.
  • Synthetic oligonucleotides can also be prepared using commercially available automated oligonucleotide synthesizers.
  • the nucleotide sequences can thus be designed with appropriate codons for a particular amino acid sequence. In general, one will select preferred codons for expression in the intended host.
  • the complete sequence is assembled from overlapping oligonucleotides prepared by standard methods and assembled into a complete coding sequence.
  • nucleic acid sequences for use herein is by recombinant means.
  • a desired nucleotide sequence can be excised from a plasmid carrying the nucleic acid using standard restriction enzymes and procedures.
  • Site specific DNA cleavage is performed by treating with the suitable restriction enzymes and procedures.
  • Site specific DNA cleavage is performed under conditions which are generally understood in the art, and the particulars of which are specified by manufacturers of commercially available restriction enzymes.
  • size separation of the cleaved fragments may be performed by
  • RNA is isolated from, for example, cells, tissues, or whole organisms by techniques known to one skilled in the art.
  • Complementary DNA cDNA is then generated using poly-dT or random hexamer primers, deoxynucleotides, and a suitable reverse transcriptase enzyme.
  • the desired polynucleotide can then be amplified from the generated cDNA by PCR.
  • the polynucleotide of interest can be directly amplified from an appropriate cDNA library.
  • Primers that hybridize with both the 5' and 3' ends of the polynucleotide sequence of interest are synthesized and used for the PCR.
  • the primers may also contain specific restriction enzyme sites at the 5' end for easy digestion and ligation of amplified sequence into a similarly restriction digested plasmid vector.
  • the expression cassette of the modified self-vector will employ a promoter that is functional in host cells.
  • vectors containing promoters and control sequences that are derived from species compatible with the host cell are used with the particular host cell.
  • Promoters suitable for use with prokaryotic hosts illustratively include the beta-lactamase and lactose promoter systems, alkaline phosphatase, the tryptophan (trp) promoter system and hybrid promoters such as tac promoter.
  • trp tryptophan
  • other functional bacterial promoters are suitable.
  • eukaryotic microbes such as yeast cultures may also be used.
  • Promoters controlling transcription from vectors in mammalian host cells may be obtained from various sources, for example, the genomes of viruses such as: polyoma, simian virus 40 (SV40), adenovirus, retroviruses, hepatitis B virus and preferably cytomegalovirus (CMV), or from heterologous mammalian promoters, e.g. ⁇ -actin promoter.
  • viruses such as: polyoma, simian virus 40 (SV40), adenovirus, retroviruses, hepatitis B virus and preferably cytomegalovirus (CMV), or from heterologous mammalian promoters, e.g. ⁇ -actin promoter.
  • the early and late promoters of the SV 40 virus are conveniently obtained as an SV40 restriction fragment which also contains the SV40 viral origin of replication.
  • the immediate early promoter of the human cytomegalovirus is conveniently obtained as a Hindlll restriction fragment.
  • DNA encoding two or more self-protein(s), -polypeptide(s), or - peptide(s) are encoded sequentially in a single self-vector utilizing internal ribosomal re-entry sequences (IRES) or other elements for expression of multiple proteins from a single DNA molecule.
  • IRS internal ribosomal re-entry sequences
  • the vectors used herein may contain a selection gene, also termed a selectable marker.
  • a selection gene encodes a protein, necessary for the survival or growth of a host cell transformed with the vector.
  • suitable selectable markers for mammalian cells include the dihydrofolate reductase gene (DHFR), the ornithine decarboxylase gene, the multidrug resistance gene (mdr), the adenosine deaminase gene, and the glutamine synthase gene.
  • the first category is based on a cell's metabolism and the use of a mutant cell line which lacks the ability to grow independent of a supplemented media.
  • the second category is referred to as dominant selection which refers to a selection scheme used in any cell type and does not require the use of a mutant cell line. These schemes typically use a drug to arrest growth of a host cell. Those cells which have a novel gene would express a protein conveying drug resistance and would survive the selection. Examples of such dominant selection use the drugs neomycin (Southern and Berg (1982) J. Molec. Appl. Genet. 1, 327), mycophenolic acid (Mulligan and Berg ( 1980) Science 209, 1422), or hygromycin (Sugden et al.
  • the vectors used herein are propagated in a host cell using antibiotic-free selection based on repressor titration (Cranenburgh et al., 2001 ).
  • the vectors are modified to contain the lac operon either as part of the lac promoter or with the lacOi and lacOs operators with the optimal spacing found in the pUC series of plasmid vectors.
  • the lacOj operator or palindromic versions of the lacO can be used in isolation as single or multiple copies (Cranenburgh et al., 2004).
  • the lac operon sequence may be incorporated at single or multiple sites anywhere within the vector so as not to interfere with other functional components of the vector.
  • a synthetic Escherichia coli lac operon dimer operator (Genbank Acc. Num. K02913) is used.
  • the lac operon may be added to a vector that lacks a suitable selective marker to provide selection, be added in addition to another selectable marker, or used to replace a selectable marker, especially an antibiotic resistance marker, to make the vector more suitable for therapeutic applications.
  • Vectors containing the lac operon can be selected in genetically modified E. coli with an essential gene, including dapD, under the control of the lac promoter (lacOP) thus allowing the modified host cell to survive by titrating the lac repression from the lacOP and allowing expression of dapD.
  • Suitable E. coli stains include OmiacdapD and OH ⁇ lacP2dapD (Cranenburgh et al., 2001 )
  • the self-vector specific agents of this invention can be formulated as polynucleotide salts for use as pharmaceuticals.
  • Polynucleotide salts can be prepared with non-toxic inorganic or organic bases.
  • Inorganic base salts include sodium, potassium, zinc, calcium, aluminum, magnesium, etc.
  • Organic non-toxic bases include salts of primary, secondary and tertiary amines, etc.
  • Such self-DNA polynucleotide salts can be formulated in lyophilized form for reconstitution prior to delivery, such as sterile water or a salt solution.
  • self-DNA polynucleotide salts can be formulated in solutions, suspensions, or emulsions involving water- or oil-based vehicles for delivery.
  • the DNA is lyophilized in phosphate buffered saline with physiologic levels of calcium (0.9 mM) and then reconstituted with sterile water prior to administration.
  • the DNA is formulated in solutions containing higher quantities of Ca ++ , between 1 mM and 2M.
  • the DNA can also be formulated in the absence of specific ion species.
  • the polynucleotide encoding a self-polypeptide can be formulated with cationic polymers including cationic liposomes.
  • Other liposomes also represent effective means to formulate and deliver self-polynucleotide.
  • the self DNA can be incorporated into a viral vector, viral particle, or bacterium for pharmacologic delivery.
  • Viral vectors can be infection competent, attenuated (with mutations that reduce capacity to induce disease), or replication-deficient.
  • Methods utilizing self-DNA to prevent the deposition, accumulation, or activity of pathogenic self proteins may be enhanced by use of viral vectors or other delivery systems that increase humoral responses against the encoded self-protein.
  • the DNA can be conjugated to solid supports including gold particles, polysaccharide-based supports, or other particles or beads that can be injected, inhaled, or delivered by particle bombardment (ballistic delivery).
  • particle bombardment ballistic delivery.
  • Methods for delivering nucleic acid preparations are known in the art. See, e.g. ; U.S. Patent Nos. 5,399,346, 5,580,859, and 5,589,466.
  • a number of viral based systems have been developed for transfer into mammalian cells. For example, retroviral systems have been described (U.S.
  • a number of adenovirus vectors have also been described, see e.g., (Haj-Ahmad et al. , J. Virol. 57:267-274, 1986; Bett er al. , J. Virol. 67:591 1 - 5921 , 1993; Mittereder et al, Human Gene Therapy 5:717-729, 1994; Seth et al, J. Virol.
  • Adeno-associated virus (AAV) vector systems have also been developed for nucleic acid delivery.
  • AAV vectors can be readily constructed using techniques well known in the art. See, e.g., U.S. Patent Nos. 5, 173,414 and 5, 139,941 ; International Publication Nos. WO 92/01070 and WO 93/03769; Lebkowski et al, Molec. Cell. Biol. 8:3988-3996, 1988; Vincent et al, Vaccines 90 (Cold Spring Harbor
  • the polynucleotide of this invention can also be delivered without a viral vector.
  • the molecule can be packaged in liposomes prior to delivery to the subject.
  • Lipid encapsulation is generally accomplished using liposomes which are able to stably bind or entrap and retain nucleic acid.
  • liposomes as carriers for delivery of nucleic acids, see, e.g., Hug et al, Biochim. Biophys. Acta. 1097: 1 -17, 1991 ; Straubinger et al, in Methods of Enzymology, Vol. 101, pp. 512-527, 1983.
  • Therapeutically effective amounts of self-vector are in the range of about 0.001 mg to about 1 g.
  • a preferred therapeutic amount of self-vector is in the range of about 10 ng to about 10 mg.
  • a most preferred therapeutic amount of self-vector is in the range of about 0.025 mg to 6 mg.
  • the self- vector is administered monthly for 6-12 months, and then every 3-12 months as a maintenance dose.
  • Alternative treatment regimens may be developed and may range from daily, to weekly, to every other month, to yearly, to a one-time
  • the polynucleotide is delivered by intramuscular injection.
  • the polynucleotide is delivered intranasally, orally, subcutaneously, intradermal ly, intravenously, mucosally, impressed through the skin, or attached to gold particles delivered to or through the dermis (see, e.g. , WO 97/46253).
  • nucleic acid can be delivered into skin cells by topical application with or without liposomes or charged lipids (see e.g. U.S. Patent No. 6,087,341 ).
  • Yet another alternative is to deliver the nucleic acid as an inhaled agent.
  • the polynucleotide is formulated in phosphate buffered saline with physiologic levels of calcium (0.9 raM). Alternatively, the polynucleotide is formulated in solutions containing higher quantities of Ca ++ , between 1 mM and 2M. The polynucleotide may be formulated with other cations such as zinc, aluminum, and others. Alternatively, or in addition, the polynucleotide may be formulated either with a cationic polymer, cationic liposome-forming compounds, or in non-cationic liposomes. Examples of cationic liposomes for DNA delivery include liposomes generated using l ,2-bis(oleoyloxy)-3-(trimethylammionio) propane (DOTAP) and other such molecules.
  • DOTAP l ,2-bis(oleoyloxy)-3-(trimethylammionio) propane
  • the delivery site Prior to delivery of the polynucleotide, the delivery site can be preconditioned by treatment with bupivicane, cardiotoxin or another agent that may enhance the subsequent delivery of the polynucleotide.
  • Such preconditioning regimens are generally delivered 12 to 96 hours prior to delivery of therapeutic polynucleotide; more frequently 24 to 48 hours prior to delivery of the therapeutic polynucleotide. Alternatively, no preconditioning treatment is given prior to polynucleotide therapy.
  • Antigen refers to any molecule that can be recognized by the immune system that is by B cells or T cells, or both.
  • Autoantigen refers to an endogenous molecule, typically a protein or fragment thereof, that elicits a pathogenic immune response.
  • the autoantigen or epitope thereof is associated with an autoimmune disease,” it is understood to mean that the autoantigen or epitope is involved in the pathophysiology of the disease either by inducing the pathophysiology (i.e., associated with the etiology of the disease), mediating or facilitating a pathophysiologic process; and/or by being the target of a pathophysiologic process.
  • the immune system aberrantly targets autoantigens, causing damage and dysfunction of cells and tissues in which the autoantigen is expressed and/or present.
  • autoantigens are ignored by the host immune system through the elimination, inactivation, or lack of activation of immune cells that have the capacity to recognize the autoantigen through a process designated "immune tolerance.”
  • epitope is understood to mean a portion of a polypeptide having a particular shape or structure that is recognized by either B-cells or T-cells of the animal's immune system.
  • Autoantigenic epitope or “pathogenic epitope” refers to an epitope of an autoantigen that elicits a pathogenic immune response.
  • polypeptide As used interchangeably herein to refer to a polymer of amino acid residues. The terms apply to amino acid polymers in which one or more amino acid residue is an artificial chemical mimetic of a corresponding naturally occurring amino acid, as well as to naturally occurring amino acid polymers and non-naturally occurring amino acid polymers.
  • Self-protein self-polypeptide
  • self-peptide any protein, polypeptide, or peptide, or fragment or derivative thereof that: is encoded within the genome of the animal; is produced or generated in the animal; may be modified post- translationally at some time during the life of the animal; and, is present in the animal non- physiologically.
  • non-physiological or “non-physiologically” when used to describe the self-protein(s), -polypeptide(s), or -peptide(s) of this invention means a departure or deviation from the normal role or process in the animal for that self-protein, -polypeptide, or - peptide.
  • the self-protein, -polypeptide or -peptide when referring to the self-protein, -polypeptide or -peptide as "associated with a disease” or “involved in a disease” it is understood to mean that the self-protein, -polypeptide, or -peptide may be modified in form or structure and thus be unable to perform its physiological role or process or may be involved in the pathophysiology of the condition or disease either by inducing the pathophysiology; mediating or facilitating a pathophysiologic process; and/or by being the target of a pathophysiologic process.
  • autoimmune disease the immune system aberrantly attacks self-proteins causing damage and dysfunction of cells and tissues in which the self-protein is expressed and/or present.
  • the self-protein, - polypeptide or -peptide can itself be expressed at non-physiological levels and/or function non- physiologically.
  • self-proteins are aberrantly expressed, and aggregate in lesions in the brain thereby causing neural dysfunction.
  • the self-protein aggravates an undesired condition or process.
  • in neurodegenerative diseases self-proteins are aberrantly expressed, and aggregate in lesions in the brain thereby causing neural dysfunction.
  • the self-protein aggravates an undesired condition or process.
  • self-proteins including collagenases and matrix metalloproteinases aberrantly degrade cartilage covering the articular surface of joints.
  • posttranslational modifications of self-protein(s), -polypeptide(s) or -peptide(s) are glycosylation, addition of lipid groups, reversible phosphorylation, addition of dimethylarginine residues, citrullination, and proteolysis, and more specifically citrullination of fillagrin and fibrin by peptidyl arginine deiminase (PAD), alpha ⁇ -crystallin phosphorylation, citrullination of MBP, and SLE
  • self-protein, -polypeptide or -peptide would all be considered host self-antigens and under normal physiological conditions are ignored by the host immune system through the elimination, inactivation, or lack of activation of immune cells that have the capacity to recognize self-antigens through a process designated "immune tolerance.”
  • a self-protein, -polypeptide, or -peptide does not include immune proteins, polypeptides, or peptides which are molecules expressed physiologically exclusively by cells of the immune system for the purpose of regulating immune function.
  • the immune system is the defense mechanism that provides the means to make rapid, highly specific, and protective responses against the myriad of potentially pathogenic microorganisms inhabiting the animal's world.
  • immune protein(s), polypeptide(s) or peptide(s) are proteins comprising the T-cell receptor, immunoglobulins, cytokines including the type I interleukins, and the type II cytokines, including the interferons and IL- 10, TNF, lymphotoxin, and the
  • chemokines such as macrophage inflammatory protein -1 alpha and beta, monocyte-chemotactic protein and RANTES, and other molecules directly involved in immune function such as Fas- ligand.
  • immune protein(s), polypeptide(s) or peptide(s) that are included in the self-protein, -polypeptide or -peptide of the invention and they are: class I MHC membrane glycoproteins, class II MHC glycoproteins and osteopontin.
  • Self-protein, -polypeptide or - peptide does not include proteins, polypeptides, and peptides that are absent from the subject, either entirely or substantially, due to a genetic or acquired deficiency causing a metabolic or functional disorder, and are replaced either by administration of said protein, polypeptide, or peptide or by administration of a polynucleotide encoding said protein, polypeptide or peptide (gene therapy).
  • disorders include Duchenne' muscular dystrophy, Becker's muscular dystrophy, cystic fibrosis, phenylketonuria, galactosemia, maple syrup urine disease, and homocystinuria.
  • Self-protein, -polypeptide or -peptide does not include proteins, polypeptides, and peptides expressed specifically and exclusively by cells which have characteristics that distinguish them from their normal counterparts, including: (1 ) clonality, representing proliferation of a single cell with a genetic alteration to form a clone of malignant cells, (2) autonomy, indicating that growth is not properly regulated, and (3) anaplasia, or the lack of normal coordinated cell differentiation. Cells have one or more of the foregoing three criteria are referred to either as neoplastic, cancer or malignant cells.
  • Modulation of refers to any alteration of an existing or potential immune responses against self-molecules, including, e.g. , nucleic acids, lipids, phospholipids, carbohydrates, self-polypeptides, protein complexes, or ribonucleoprotein complexes, that occurs as a result of administration of a polynucleotide encoding a self-polypeptide.
  • modulation includes any alteration in presence, capacity, or function of any immune cell involved in or capable of being involved in an immune response.
  • Immune cells include B cells, T cells, NK cells, NK T cells, professional antigen-presenting cells, non-professional antigen-presenting cells, inflammatory cells, or any other cell capable of being involved in or influencing an immune response.
  • “Modulation” includes any change imparted on an existing immune response, a developing immune response, a potential immune response, or the capacity to induce, regulate, influence, or respond to an immune response. Modulation includes any alteration in the expression and/or function of genes, proteins and/or other molecules in immune cells as part of an immune response.
  • Modulation of an immune response includes, for example, the following:
  • immune cells elimination, deletion, or sequestration of immune cells; induction or generation of immune cells that can modulate the functional capacity of other cells such as autoreactive lymphocytes, antigen presenting cells (APCs), or inflamatory cells; induction of an unresponsive state in immune cells (i.e. , anergy); increasing, decreasing, or changing the activity or function of immune cells or the capacity to do so, including but not limited to altering the pattern of proteins expressed by these cells. Examples include altered production and/or secretion of certain classes of molecules such as cytokines, chemokines, growth factors, transcription factors, kinases, costimulatory molecules, or other cell surface receptors; or any combination of these modulatory events.
  • Preventing refers to the administration of a polynucleotide encoding a self- protein ⁇ ), -polypeptide(s), or -peptide(s) either alone or in combination with another compound as described herein, to prevent the occurrence or onset of a disease or disorder or some or all of the symptoms of a disease or disorder or to lessen the likelihood of the onset of a disease or disorder.
  • treatment includes the administration of one or both agent(s) of the instant invention prior to or following the onset of a symptom of an autoimmune disease or disorder thereby preventing or removing all signs of the autoimmune disease or disorder.
  • agent(s) after clinical manifestation of the autoimmune disease to combat the symptoms of the disease.
  • administration of one or both agent(s) of the instant invention after onset and after clinical symptoms have developed where administration affects clinical parameters of the autoimmune disease or disorder, such as the degree of tissue injury, whether or not the treatment leads to amelioration of the autoimmune disease, comprises "treatment” or "therapy” within the context of the invention.
  • “Therapeutically effective amounts" of the self-vector comprising polynucleotide encoding one or more self-protein(s), -polypeptide(s) or -peptide(s) is administered in accord with the teaching of this invention and will be sufficient to treat or prevent the disease as for example by ameliorating or eliminating symptoms and/or the cause of the disease.
  • therapeutically effective amounts fall within broad range(s) and are determined through clinical trials and for a particular patient is determined based upon factors known to the ordinarily skilled clinician including the severity of the disease, weight of the patient, age and other factors.
  • Therapeutically effective amounts of self-vector are in the range of about 0.001 micrograms to about 1 gram.
  • a preferred therapeutic amount of self-vector is in the range of about
  • a most preferred therapeutic amount of self-vector is in the range of about 0.025 mg to 5 mg.
  • Polynucleotide therapy is delivered monthly for 6- 12 months, and then every 3-12 months as a maintenance dose.
  • Alternative treatment regimens may be developed and may range from daily, to weekly, to every other month, to yearly, to a one-time administration depending upon the severity of the disease, the age of the patient, the self- protein(s), -polypeptide(s) or -peptide(s) being administered and such other factors as would be considered by the ordinary treating physician.
  • the method of treating an autoimmune disease by administering a combination of specific agents comprising a B cell specific agent in combination with a DNA antigen specific therapeutic agent comprising a polynucleotide encoding a self-polypeptide can modulate an immune response by eliminating, sequestering, or inactivating immune cells mediating or capable of mediating an undesired immune response; inducing, generating, or turning on immune cells that mediate or are capable of mediating a protective immune response; changing the physical or functional properties of immune cells; or a combination of these effects.
  • Examples of measurements of the modulation of an immune response include, but are not limited to, examination of the presence or absence of immune cell populations (using flow cytometry, immunohistochemistry, histology, electron microscopy, polymerase chain reaction (PCR)); measurement of the functional capacity of immune cells including ability or resistance to proliferate or divide in response to a signal (such as using T cell proliferation assays and pepscan analysis based on 3 H-thymidine incorporation following stimulation with anti-CD3 antibody, anti-T cell receptor antibody, anti-CD28 antibody, calcium ionophores, PMA, antigen presenting cells loaded with a peptide or protein antigen; B cell proliferation assays); measurement of the ability to kill or lyse other cells (such as cytotoxic T cell assays); measurements of the cytokines, chemokines, cell surface molecules, antibodies and other products of the cells (e.g., by flow cytometry, enzyme-linked immunosorbent assays, Western blot analysis, protein microarray analysis, immunoprecipit
  • mice 50 uL of blood were taken daily and the amount of anti-CD20 in serum was quantified by ELISA using a synthetic CD20-peptide containing its extracellular sequence from amino acids 140 to 184, with biotin fusion at amino acid 140, as described previously (P. J. Adamson, H. Zola, I. C. Nicholson, G. Pilkington, A. Hohmann, Leuk Res 25, 1047 (Dec, 2001 )). Mice were given varying doses (5, 10, 25, 50, or 100 ⁇ g) of the B cell specific agent, i.e., anti-CD20 mAb, only on day 1 (lx) or on days 1 , 4, 7 and 10 (4x) intravenously.
  • the B cell specific agent i.e., anti-CD20 mAb
  • mice not treated with an antibody or those mice treated with an isotype matched control antibody were used as controls.
  • blood glucose was monitored in treated and untreated mice twice weekly for 10 weeks. Mice were considered protected when the blood glucose values were ⁇ 200ug/dL.
  • Expression of insulin in the pancreas, 5 weeks post treatment, was determined by immunohistochemistry (IHC) staining of 6 ⁇ frozen pancreatic sections with guinea-pig anti- swine insulin (1 :300) antibody, as described (D. Bresson et al , J Clin Invest 116, 1371 (May, 2006), U.
  • IHC immunohistochemistry
  • splenocytes or PLN cells were surface-stained with anti-CD4, -CD8, -CD25, -CD 127, -CTLA4 ( 1 :20 dilution, BD Biosciences), followed by intracellular staining with anti-Foxp3 (ebioscience) or anti-GITR (R&D systems) (D. Bresson et al, J Clin Invest 116, 1371 (May, 2006)).
  • Frequencies of nTreg cells were determined by analyzing the expression of Foxp3, CTLA4 or GITR in CD4 + CD127 low CD25 + or CD4 + CD25- cells, and as a control in CD8 + cells. Further, production of cytokines was assessed by ELISA, ELISPOT or ICS.
  • Splenocytes or PLN cells were stimulated for 3-days in vitro using anti-CD3/CD28 or isotype matched control antibodies (l ug/mL each).
  • Culture supernatants were harvested and cytokines were quantified using IFN- ⁇ , TNF-a, IL-10, IL-4 (BD pharmingen), TGF- ⁇ (Quantikine kit, R&D systems) ELISA kits or using multiplexed cytokine assay (Beadlyte Mouse Multi-Cytokine Detection System; Upstate USA Inc), as per manufacturer's instructions. Intracellular cytokine staining and analysis were performed as described (D.
  • cytokine assays were performed following stimulation with insulin 9-23 and proinsulin peptides (IA g7 restricted). Additionally, to determine the effect on autoaggressive CD8 + T cells, frequencies of antigen specific T cells were determined in the peripheral blood by FACS analysis, or their presence were detected in frozen pancreatic sections by IHC, using NRP-V7 tetramers (obtained from Pere Santamaria, University of Calgary, Canada) (D. Bresson et al., J Clin Invest 116, 1371 (May, 2006)).
  • mice were administered a single 10, 50 and 100 ⁇ g dose of B cell specific agent, i.e., murine anti-CD20 antibody 5D2.
  • DNA plasmid vector encoding proinsulin (SEQ ID NO: 20) was prepared, (see WO 2007/044394) and used at >95% purity.
  • mice were administered DNA plasmid vector encoding proinsulin on days 1 , 8, 15 and 22.
  • Recent onset T1 D NOD-mice treated with anti- CD20 only, insulin only or untreated mice were included as controls.
  • mice with BG>400mg/dl hardly ever revert from recent-onset diabetes, because the remaining beta cell mass is too low.
  • mice with increased efficacy of protection pancreatic insulin expression, insulitis, and the effect on antigen specific CD8 + T cells was determined.
  • splenocytes or peripheral lymph node (PLN) cells or CD4 + or CD8 + or B220 + cells were used as described above.
  • nTregs and adaptive Tregs were enumerated and tested for cytokine secretion as described above.
  • a one time administration of anti-CD20 in combination with either a one time (Fig. 4A) or four times (Fig. 4C) administration of Proinsulin plasmid exhibited synergy in protecting the NOD mice from T1 D onset. Further, a four-time administration of Proinsulin plasmid exhibited more synergy than a one time administration of Proinsulin plasmid.
  • proinsulin plasmid administration of proinsulin plasmid was able to exhibit synergy is a significant finding.
  • the combination therapy of anti-CD20 and proinsulin plasmid is more efficacious than using either of these reagents alone.
  • Pemphigus vulgaris is a skin blistering disease caused by a targeted autoantibody immune response against the self protein, desmoglein III (DMGIII) (Perez and Patton. Drugs Aging.. 26:833-46, 2009).
  • Current first line therapies include high dose systemic corticosteroids (e.g., prednisone l mg/kg/day) .
  • Steroid-sparing immunosuppressives such as azathioprine, cyclosphosphamide, mycophenolate mofetil are also utilized extensively.
  • Rituximab anti-CD20 has been used to treat pemphigus vulgaris that is refractory to conventional therapy (Fernando and Broadfoot. G.
  • IVIG Intravenous immunoglobulin
  • DMGIII Human desmogleinlll
  • SEQ ID NO: 3 An animal model of PV used to screen potential therapeutics for treating PV is developed using anti-DMGIII antibodies obtained from immunized DMGIII knockout mouse (Koch et al. J Cell Biol. 137: 1091 -1 102, 1997; Amagai et al. J Clin. Invest.. 105:625-631 , 2000).
  • Anti- DMGIII antibodies are adoptively transferred to wild type mice that subsequently develop skin lesions similar in pathophysiology to those in patients with PV.
  • a pDNA self-vector is constructed encoding the self-antigen polypeptide, desmoglein III (SEQ ID NO: 1 ), comprising the appropriate control and regulatory elements (See for exampleWO 2007/044394).
  • a second pDNA self-vector is constructed encoding DMGIII non- secreted (SEQ ID NO: 2) and is also tested as follows. Treatment commences in DMGIII knockout animals after antibodies to DMGIII are induced and detectable by ELISA.
  • a dose range is tested from 5 to 250ug/mouse of anti-CD20 alone defining the kinetics of response and extent of disease modification of anti-CD20 in this disease model.
  • a sub-optimal and optimal dose level of anti-CD20 is tested in combination with pDNA self vector encoding DMGIII.
  • a sub-optimal dose of anti-CD20 mAb is evaluated to identify a lower dose of anti-CD20 mAb for administration in combination with the DNA antigen specific therapeutic agent encoding DMGIII to improve the anti-CD20 mAb's safety profile while maintaining efficacy.
  • Anti- CD20 mAb (both sub-optimal and optimal dose levels) and pDNA self vector encoding DMGIII is administered to DMGIII knock out mice randomized to 7 treatment groups based on antibody levels. The treatment regimens are shown in Table 3.
  • Group 1 is left untreated. Three groups are treated with sub-optimal anti-CD20 mAb, including two with the combination therapy of this invention, and three groups are treated with the optimal anti-CD20 mAb dose, including two with the combination therapy of this invention.
  • the effect of the anti-CD20 mAb alone is seen in Groups 2 and 5.
  • the effect of combination therapy, by combining the pDNA self vector encoding autoantigen with the anti-CD20 mAb is seen in groups 3, 4, 6 and 7.
  • the effect of the timing of the administration (simultaneous therapy versus sequential therapy) on the efficacy is demonstrated by comparing groups 3 with 4 and 6 with 7.
  • pDNA self vector is administered weekly by IM injection (5C ⁇ g/mouse, 25 ⁇ g/quad). For groups 4 and 7, the pDNA therapy is started at 2, 4 or 6 weeks after the anti-CD20 mAb is administered. Once started, the pDNA therapy is continued weekly until sera is collected for the adoptive transfer.
  • MS DNA Antigen Specific Therapeutic Agent to Treat Multiple Sclerosis
  • EAE experimentally induced autoimmune encephalomyelitis
  • Antigen specific therapy has been previously demonstrated to decrease symptoms of EAE.
  • One of the myelin proteins targeted during the autoimmune response in EAE is the protein, proteolipid protein or PLP.
  • PLP proteolipid protein
  • pDNA self-vector encoding PLP has demonstrated ability to lower relapse rates in preclinical EAE studies (Garren et al. Immunity., 15(1 ): 15-22, 2001 ) in mice, however, animals continue to have symptoms of disease and could benefit from combination therapy of a DNA antigen specific therapeutic agent combined with another immunomodulatory compound such as B cell specific agent.
  • Murine PLP is a polypeptide of 30.08 molecular weight.
  • EAE is induced in the SJL mouse model using PLP peptide emulsified in CFA.
  • a DNA self-vector is constructed encoding the self antigen polypeptide, proteolipid protein, PLP (SEQ ID NO: 5), comprising the appropriate control and regulatory elements (See WO 2007/044394).
  • the anti-CD20 monoclonal antibody is tested across a dose range from 5 to 250ug/mouse of anti-CD20 alone defining the kinetics of response and extent of disease modification of anti-CD20 in this model.
  • a sub-optimal and optimal dose level of anti-CD20 mAb is tested alone and in combination with pDNA self-vector encoding PLP.
  • a sub-optimal dose of anti-CD20 mAb is evaluated to identify a lower dose of anti-CD20 mAb for administration in combination with the DNA antigen specific therapeutic agent encoding PLP to improve the anti-CD20 mAb's safety profile while maintaining efficacy.
  • Anti-CD20 (both sub-optimal and optimal dose levels) and pDNA self- vector encoding PLP is administered to EAE mice randomized to seven treatment groups based on disease severity. The treatment regimen is shown in Table 3 above.
  • Group 1 is left untreated. Three groups are treated with sub-optimal anti-CD20 mAb, and two groups are treated with the optimal anti-CD20 mAb dose. The effect of the anti-CD20 mAb alone is seen in Groups 2 and 5. The effect of combination therapy by combining the pDNA self-vector encoding autoantigen with anti-CD20 mAb is seen in groups 3, 4, 6 and 7. The effect of the timing of the administration (simultaneous therapy versus sequential therapy) on the efficacy is demonstrated by comparing groups 3 with 4 and 6 with 7.
  • the pDNA antigen specific therapy is continued weekly for the length of the study .
  • the pDNA self- vector is administered weekly by IM injection at a 50ug/mouse dose (25ug/quad).
  • the pDNA antigen specific therapy is started at 2, 4 or 6 weeks after the anti-CD20 is administered.
  • the pDNA therapy is continued weekly for the length of the study.
  • Effectiveness of the combination approach is determined by comparing disease scores and brain histology from mice that received only the anti-CD20 antibody with animals that received combination therapy with anti-CD20 mAb and pDNA self-vector encoding PLP.
  • Fibrinogen is a polypeptide of 69.76 molecular weight.
  • a DNA self-vector is constructed encoding the self antigen polypeptide, fibrinogen (SEQ ID NO:6), comprising the appropriate control and regulatory elements (See WO 2007/044394).
  • the anti-CD20 monoclonal antibody is tested across a dose range from 5 to 250ug/mouse of anti-CD20 alone defining the kinetics of response and extent of disease modification of anti-CD20 in this model.
  • a sub-optimal and optimal dose level of anti- CD20 mAb is tested alone and in combination with pDNA self-vector encoding fibrinogen.
  • a sub-optimal dose of anti-CD20 mAb is evaluated to identify a lower dose of anti-CD20 mAb for administration in combination with the DNA antigen specific therapeutic agent encoding fibrinogen to improve the anti-CD20 mAb's safety profile while maintaining efficacy.
  • Anti- CD20 (both sub-optimal and optimal dose levels) and pDNA self vector encoding fibrinogen is administered to EAE mice randomized to seven treatment groups based on disease severity. The treatment regimen is shown in Table 3 above.
  • Group 1 is left untreated. Three groups are treated with sub-optimal anti-CD20 mAb, and two groups are treated with the optimal anti-CD20 mAb dose. The effect of the anti-CD20 mAb alone is seen in Groups 2 and 5. The effect of combination therapy by combining the pDNA self vector encoding autoantigen with anti-CD20 mAb is seen in groups 3, 4, 6 and 7. The effect of the timing of the administration (simultaneous therapy versus sequential therapy) on the efficacy is demonstrated by comparing groups 3 with 4 and 6 with 7. Once started, the pDNA antigen specific therapy is continued weekly for the length of the study.
  • the pDNA self vector is administered weekly by IM injection at a 50ug/mouse dose (25ug/quad).
  • a 50ug/mouse dose 25ug/quad.
  • the pDNA antigen specific therapy is started at 2, 4 or 6 weeks after the anti-CD20 is administered. Once started, the pDNA therapy is continued weekly for the length of the study.
  • Effectiveness of the combination approach is determined by comparing disease scores and joint histology from mice that received only the anti-CD20 antibody with animals that received combination therapy of anti-CD20 mAb and pDNA self vector encoding fibrinogen.
  • Idiopathic pulmonary fibrosis is a chronic progressive autoimmune disease that is ultimately a fatal lung disease of unknown etiology. Its prognosis is poor and the outcome even worse than in many malignant diseases.
  • the bleomycin mouse model of IPF (bIPF) has been used for decades as a model system for evaluating therapeutics for evaluation in clinical studies (Moeller et al., Int. J. Biochem Cell Biol. 2008. 40(3):362-382).
  • bleomycin causes inflammatory and fibrotic reactions within a short period of time.
  • Proinflammatory cytokines are elevated followed by increased expression of pro-fibrotic markers with a peak at about 14 days.
  • the switch between inflammation and fibrosis occurs about day 9. Part of the inflammation that is induced is thought to be the result of an autoimmune response to collagen V that is exposed during the remodeling of lung tissue after exposure to injury (Wilkes, DS., Arch. Immunol. Ther. Exp. 2003. 51 :227-30).
  • Collagen V is a polypeptide of 183.56 molecular weight.
  • a DNA self- vector is constructed encoding the self-antigen polypeptide, collagen V (SEQ ID NO: 10), comprising the appropriate control and regulatory elements (See WO 2007/044394).
  • a second pDNA self-vector is constructed encoding collagen V non-secreted and is also tested as follows. The anti-CD20 monoclonal antibody is tested across a dose range from 5 to 250ug/mouse of anti-CD20 alone defining the kinetics of response and extent of disease modification of anti- CD20 in this model.
  • a sub-optimal and optimal dose level of anti-CD20 mAb is tested alone and in combination with pDNA self-vector encoding collagen V.
  • a sub-optimal dose of anti- CD20 mAb is evaluated to identify a lower dose of anti-CD20 mAb for administration in combination with the DNA antigen specific therapeutic agent encoding collagen V to improve the anti-CD20 mAb's safety profile while maintaining efficacy.
  • Anti-CD20 (both sub-optimal and optimal dose levels) and pDNA self vector encoding collagen is administered to EAE mice randomized to seven treatment groups based on disease severity. The treatment regimen is shown in Table 3 above.
  • Group 1 is left untreated. Three groups are treated with sub-optimal anti-CD20 mAb, and two groups are treated with the optimal anti-CD20 mAb dose. The effect of the anti-CD20 mAb alone is seen in Groups 2 and 5. The effect of combination therapy by combining the pDNA self vector encoding autoantigen with anti-CD20 mAb is seen in groups 3, 4, 6 and 7. The effect of the timing of the administration (simultaneous therapy versus sequential therapy) on the efficacy is demonstrated by comparing groups 3 with 4 and 6 with 7. Once started, the pDNA antigen specific therapy is continued weekly for the length of the study.
  • the pDNA self vector is administered weekly by IM injection at a 50ug/mouse dose (25ug/quad).
  • a 50ug/mouse dose 25ug/quad.
  • the pDNA antigen specific therapy is started at 2, 4 or 6 weeks after the anti-CD20 is administered. Once started, the pDNA therapy is continued weekly for the length of the study.
  • Effectiveness of the combination approach is determined by comparing disease scores and histopathological evaluation of lung tissues from mice that received only the anti-CD20 antibody with animals that received combination therapy of anti-CD20 mAb and pDNA self vector encoding collagen V.
  • MG Myasthenia gravis
  • AChR nicotinic acetylcholine receptor
  • the disease is mediated by circulating autoantibodies that block acetylcholine receptors at the post-synaptic neuromuscular junction inhibiting the stimulative effect of the
  • AChR is a polypeptide of 51 .84 molecular weight.
  • a DNA self-vector is constructed encoding the self-antigen polypeptide acetylcholine receptor (SEQ ID NO: 14) comprising the appropriate control and regulatory elements (See WO 2007/044394).
  • the anti-CD20 monoclonal antibody is tested across a dose range from 5 to 250ug/rat of anti-CD20 alone defining the kinetics of response and extent of disease modification of anti-CD20 mAb in the EAMG model.
  • a sub-optimal and optimal dose level of anti-CD20 mAb is tested alone and in combination with pDNA self-vector encoding AChR.
  • a sub-optimal dose of anti-CD20 mAb is evaluated to identify a lower dose of anti-CD20 mAb for administration in combination with the DNA antigen specific therapeutic agent encoding AChR to improve the anti-CD20 mAb's safety profile while maintaining efficacy.
  • Anti-CD20 (both sub-optimal and optimal dose levels) and pDNA self vector encoding AChR is administered to EAMG rat randomized to seven treatment groups based on disease severity. The treatment regimen is shown in Table 3 above.
  • Group 1 is left untreated. Three groups are treated with sub-optimal anti-CD20 mAb, and two groups are treated with the optimal anti-CD20 mAb dose. The effect of the anti-CD20 mAb alone is seen in Groups 2 and 5. The effect of combination therapy by combining the pDNA self vector encoding autoantigen with anti-CD20 mAb is seen in groups 3, 4, 6 and 7. The effect of the timing of the administration (simultaneous therapy versus sequential therapy) on the efficacy is demonstrated by comparing groups 3 with 4 and 6 with 7. Once started, the pDNA antigen specific therapy is continued weekly for the length of the study.
  • the pDNA self vector is administered weekly by IM injection at a 50ug/rat dose (25ug/quad). For groups 4 and 7, the pDNA antigen specific therapy is started at 2, 4 or 6 weeks after the anti-CD20 is administered. Once started, the pDNA therapy is continued weekly for the length of the study. [0247] Effectiveness of the combination approach is determined by comparing disease scores, muscle fatiguability and histopathological evaluation of muscle tissues from rats that received only the anti-CD20 antibody with animals that received combination therapy of anti-CD20 mAb and pDNA self vector encoding AChR.
  • This example provides data showing that DNA vaccine (rBHT-3034) and corticosteroid (Solu-Medrol) combination therapy is more effective than either monotherapy at suppressing disease scores in a rat model of EAMG.
  • EAMG was induced in Lewis rats as previously described. Prior to the onset of disease, animals were randomized to treatment groups and dosing of Solu-Medrol began 7 days post immunization. Weekly IM injections of rBHT-3034 began at Day 28. Animals were scored weekly for disease and disease scores are plotted above ⁇ SEM. P-values ⁇ 0.05 were obtained from days 76-105 comparing the Solu-Medrol+rBHT-3034 to the PBS control group using a ruskal-Wallis test.

Landscapes

  • Health & Medical Sciences (AREA)
  • Veterinary Medicine (AREA)
  • Medicinal Chemistry (AREA)
  • Public Health (AREA)
  • General Health & Medical Sciences (AREA)
  • Animal Behavior & Ethology (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • General Chemical & Material Sciences (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Engineering & Computer Science (AREA)
  • Immunology (AREA)
  • Diabetes (AREA)
  • Rheumatology (AREA)
  • Neurology (AREA)
  • Physical Education & Sports Medicine (AREA)
  • Orthopedic Medicine & Surgery (AREA)
  • Transplantation (AREA)
  • Emergency Medicine (AREA)
  • Obesity (AREA)
  • Neurosurgery (AREA)
  • Pain & Pain Management (AREA)
  • Endocrinology (AREA)
  • Hematology (AREA)
  • Biomedical Technology (AREA)
  • Dermatology (AREA)
  • Epidemiology (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
  • Peptides Or Proteins (AREA)
  • Medicines Containing Antibodies Or Antigens For Use As Internal Diagnostic Agents (AREA)

Abstract

La présente invention concerne le traitement de maladies auto-immunes avec une combinaison d'un agent spécifique aux lymphocytes B et un agent thérapeutique spécifique à un antigène d'ADN codant pour une protéine endogène associée à la maladie auto-immune. Plus spécifiquement, la présente invention concerne une combinaison d'un agent spécifique aux lymphocytes B, tel qu'un anticorps avec un agent thérapeutique spécifique à un antigène d'ADN, tel qu'un vecteur plasmidique à ADN codant pour un antigène endogène associé à la maladie auto-immune. La présente invention concerne en outre des compositions pharmaceutiques et des articles de fabrication tels que des trousses comprenant les agents et des combinaisons de ceux-ci.
PCT/US2011/021862 2010-01-20 2011-01-20 Thérapie d'association pour traiter des maladies auto-immunes WO2011091138A1 (fr)

Priority Applications (2)

Application Number Priority Date Filing Date Title
JP2012550114A JP2013517329A (ja) 2010-01-20 2011-01-20 自己免疫疾患を処置するための併用治療
EP11735172A EP2525825A1 (fr) 2010-01-20 2011-01-20 Thérapie d'association pour traiter des maladies auto-immunes

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US29683910P 2010-01-20 2010-01-20
US61/296,839 2010-01-20

Publications (1)

Publication Number Publication Date
WO2011091138A1 true WO2011091138A1 (fr) 2011-07-28

Family

ID=44307201

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2011/021862 WO2011091138A1 (fr) 2010-01-20 2011-01-20 Thérapie d'association pour traiter des maladies auto-immunes

Country Status (3)

Country Link
EP (1) EP2525825A1 (fr)
JP (1) JP2013517329A (fr)
WO (1) WO2011091138A1 (fr)

Cited By (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2015162504A1 (fr) * 2014-04-23 2015-10-29 Novartis Ag Nouveau dosage et nouvelles utilisations de l'ofatumumab
WO2019028503A1 (fr) * 2017-08-07 2019-02-14 St Vincent's Institute Of Medical Research Therapie du diabète de type 1
US11279745B2 (en) 2019-04-26 2022-03-22 Novo Nordisk A/S Tolerogenic DNA vaccine
US11466278B2 (en) 2016-11-01 2022-10-11 Novo Nordisk A/S Temperature based plasmid regulation system

Citations (7)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2007044394A2 (fr) * 2005-10-05 2007-04-19 Bayhill Therapeutics, Inc. Compositions et methodes de traitement de maladies auto-immunes
US20070190045A1 (en) * 2004-02-04 2007-08-16 Kevan Herold Anti-CD3 and antigen-specific immunotherapy to treat autoimmunity
US20080095771A1 (en) * 2005-01-13 2008-04-24 Genentech, Inc. Treatment Method
US20080108585A1 (en) * 1999-03-12 2008-05-08 The Board Of Trustees Of The Leland Stanford Junior University Dna vaccination for treatment of autoimmune disease
US20090016968A1 (en) * 2005-08-06 2009-01-15 Qinghua Wang Composition and method for prevention and treatment of type i diabetes
US20090060921A1 (en) * 2006-01-17 2009-03-05 Biolex Therapeutics, Inc. Glycan-optimized anti-cd20 antibodies
US20090311255A1 (en) * 2004-05-05 2009-12-17 Genentech, Inc. Preventing autoimmune disease

Patent Citations (7)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20080108585A1 (en) * 1999-03-12 2008-05-08 The Board Of Trustees Of The Leland Stanford Junior University Dna vaccination for treatment of autoimmune disease
US20070190045A1 (en) * 2004-02-04 2007-08-16 Kevan Herold Anti-CD3 and antigen-specific immunotherapy to treat autoimmunity
US20090311255A1 (en) * 2004-05-05 2009-12-17 Genentech, Inc. Preventing autoimmune disease
US20080095771A1 (en) * 2005-01-13 2008-04-24 Genentech, Inc. Treatment Method
US20090016968A1 (en) * 2005-08-06 2009-01-15 Qinghua Wang Composition and method for prevention and treatment of type i diabetes
WO2007044394A2 (fr) * 2005-10-05 2007-04-19 Bayhill Therapeutics, Inc. Compositions et methodes de traitement de maladies auto-immunes
US20090060921A1 (en) * 2006-01-17 2009-03-05 Biolex Therapeutics, Inc. Glycan-optimized anti-cd20 antibodies

Non-Patent Citations (4)

* Cited by examiner, † Cited by third party
Title
BRESSON ET AL.: "Anti-CD3 and nasal proinsulin combination therapy enhances remission from recent-onset autoimmune diabetes by inducing Tregs.", J. CLIN. INVEST., vol. 116, no. 5, May 2006 (2006-05-01), pages 1371 - 1381, XP008167683, DOI: doi:10.1172/JCI27191 *
BRESSON ET AL.: "Immunotherapy for the prevention and treatment of type 1 diabetes: optimizing the path from bench to bedside.", DIABETES CARE., vol. 32, no. 10, October 2009 (2009-10-01), pages 1753 - 1768, XP008167682, DOI: doi:10.2337/dc09-0373 *
EVERY ET AL.: "Intranasal vaccination with proinsulin DNA induces regulatory CD4+ T cells that prevent experimental autoimmune diabetes.", J. IMMUNOL, vol. 176, no. 8, 15 April 2006 (2006-04-15), pages 4608 - 4615, XP055020156, DOI: doi:10.4049/jimmunol.176.8.4608 *
SOLVASON ET AL.: "Improved efficacy of a tolerizing DNA vaccine for reversal of hyperglycemia through enhancement of gene expression and localization to intracellular sites.", J. IMMUNOL., vol. 181, no. 12, 15 December 2008 (2008-12-15), pages 8298 - 8307, XP008167684 *

Cited By (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2015162504A1 (fr) * 2014-04-23 2015-10-29 Novartis Ag Nouveau dosage et nouvelles utilisations de l'ofatumumab
US11466278B2 (en) 2016-11-01 2022-10-11 Novo Nordisk A/S Temperature based plasmid regulation system
WO2019028503A1 (fr) * 2017-08-07 2019-02-14 St Vincent's Institute Of Medical Research Therapie du diabète de type 1
US11806386B2 (en) 2017-08-07 2023-11-07 St. Vincent's Institute Of Medical Research Type I diabetes therapy
US11279745B2 (en) 2019-04-26 2022-03-22 Novo Nordisk A/S Tolerogenic DNA vaccine

Also Published As

Publication number Publication date
EP2525825A1 (fr) 2012-11-28
JP2013517329A (ja) 2013-05-16

Similar Documents

Publication Publication Date Title
EP1951304B1 (fr) Procede destine a traiter une lesion articulaire
TWI433682B (zh) Cd20抗體於治療多發性硬化症之用途及用於該用途之物品
US20090169550A1 (en) Therapy of rituximab-refractory rheumatoid arthritis patients
US20090004189A1 (en) Biological markers predictive of rheumatoid arthritis response to b-cell antagonists
US20060062787A1 (en) Method for treating Sjogren's syndrome
US20060233797A1 (en) Treatment of inflammatory bowel disease (IBD)
US20090204489A1 (en) Biological markers predictive of rheumatoid arthritis response to b-cell antagonists
JP2008501706A (ja) 疾患の治療方法
JP2008515890A (ja) 血管炎の治療方法
JP2008501706A5 (fr)
JP2007536246A (ja) 自己免疫疾患の予防法
CA2706899C (fr) Agents de depletion de cellules-b, comme les anticorps anti-cd20 ou leurs fragments, pour le traitement du syndrome de fatigue chronique
US20160051671A1 (en) COMBINATION OF BLyS INHIBITION AND ANTI-CD 20 AGENTS FOR TREATMENT OF AUTOIMMUNE DISEASE
WO2010075249A2 (fr) Méthode de traitement de la polyarthrite rhumatoïde avec des antagonistes de cellules b
US7914785B2 (en) B-cell depleting agents, like anti-CD20 antibodies or fragments thereof for the treatment of chronic fatigue syndrome
EP2525825A1 (fr) Thérapie d'association pour traiter des maladies auto-immunes
WO2009134738A1 (fr) Réponses à des immunisations chez des patients atteints de polyarthrite rhumatoïde traités avec un anticorps anti-cd20
US20110142836A1 (en) B-cell depleting agents for the treatment of chronic fatigue syndrome

Legal Events

Date Code Title Description
121 Ep: the epo has been informed by wipo that ep was designated in this application

Ref document number: 11735172

Country of ref document: EP

Kind code of ref document: A1

WWE Wipo information: entry into national phase

Ref document number: 2012550114

Country of ref document: JP

NENP Non-entry into the national phase

Ref country code: DE

WWE Wipo information: entry into national phase

Ref document number: 2011735172

Country of ref document: EP