US20040022787A1 - Methods for treating an autoimmune disease using a soluble CTLA4 molecule and a DMARD or NSAID - Google Patents

Methods for treating an autoimmune disease using a soluble CTLA4 molecule and a DMARD or NSAID Download PDF

Info

Publication number
US20040022787A1
US20040022787A1 US10/419,008 US41900803A US2004022787A1 US 20040022787 A1 US20040022787 A1 US 20040022787A1 US 41900803 A US41900803 A US 41900803A US 2004022787 A1 US2004022787 A1 US 2004022787A1
Authority
US
United States
Prior art keywords
subject
weight
day
ctla4
molecule
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Abandoned
Application number
US10/419,008
Inventor
Robert Cohen
Suzette Carr
David Hagerty
Robert Peach
Jean-Claude Becker
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Bristol Myers Squibb Co
Original Assignee
Bristol Myers Squibb Co
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
Priority claimed from US09/898,195 external-priority patent/US7455835B2/en
Priority to US10/419,008 priority Critical patent/US20040022787A1/en
Application filed by Bristol Myers Squibb Co filed Critical Bristol Myers Squibb Co
Assigned to BRISTOL-MYERS SQUIBB COMPANY reassignment BRISTOL-MYERS SQUIBB COMPANY ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: HAGERTY, DAVID, PEACH, ROBERT J., BECKER, JEAN-CLAUDE, CARR, SUZETTE, COHEN, ROBERT
Publication of US20040022787A1 publication Critical patent/US20040022787A1/en
Priority to US12/720,064 priority patent/US8227420B2/en
Priority to US13/191,923 priority patent/US8148332B2/en
Priority to US13/404,384 priority patent/US8497247B2/en
Priority to US13/788,970 priority patent/US8722632B2/en
Priority to US13/795,545 priority patent/US8703718B2/en
Priority to US14/193,687 priority patent/US9296808B2/en
Priority to US15/047,849 priority patent/US10052360B2/en
Abandoned legal-status Critical Current

Links

Images

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/16Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • A61K38/17Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • A61K38/177Receptors; Cell surface antigens; Cell surface determinants
    • A61K38/1774Immunoglobulin superfamily (e.g. CD2, CD4, CD8, ICAM molecules, B7 molecules, Fc-receptors, MHC-molecules)
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/0005Vertebrate antigens
    • A61K39/0008Antigens related to auto-immune diseases; Preparations to induce self-tolerance
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/395Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/50Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates
    • A61K47/51Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent
    • A61K47/68Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an antibody, an immunoglobulin or a fragment thereof, e.g. an Fc-fragment
    • A61K47/6801Drug-antibody or immunoglobulin conjugates defined by the pharmacologically or therapeutically active agent
    • A61K47/6803Drugs conjugated to an antibody or immunoglobulin, e.g. cisplatin-antibody conjugates
    • A61K47/6811Drugs conjugated to an antibody or immunoglobulin, e.g. cisplatin-antibody conjugates the drug being a protein or peptide, e.g. transferrin or bleomycin
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P19/00Drugs for skeletal 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
    • 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
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/705Receptors; Cell surface antigens; Cell surface determinants
    • C07K14/70503Immunoglobulin superfamily
    • C07K14/70521CD28, CD152
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2319/00Fusion polypeptide

Definitions

  • the present invention relates generally to the field of immune system diseases, e.g., rheumatic diseases.
  • the invention relates to methods and compositions for treating immune system diseases, e.g., rheumatic diseases, such as rheumatoid arthritis, by administering to a subject an effective amount of soluble CTLA4 molecules alone, or in conjunction with a Disease Modifying Anti-Rheumatic Drug (DMARD).
  • DMARD Disease Modifying Anti-Rheumatic Drug
  • Rheumatic diseases encompass a group of diseases that affect the musculo-skeletal and connective tissues of the body. These diseases are characterized by chronic inflammation that often leads to permanent tissue damage, deformity, atrophy and disability. Rheumatic diseases affect the joints, bone, soft tissue, or spinal cord (Mathies, H. 1983 Rheuma) and are classified as inflammatory rheumatism, degenerative rheumatism, extra-articular rheumatism, or collagen diseases. Some rheumatic diseases are known to be autoimmune diseases caused by a subject's altered immune response.
  • Rheumatoid arthritis is a progressive rheumatic disease, affecting approximately 2% of the adult population of developed countries (Utsinger, P. D., et al., 1985 Rheumatoid Arthritis, p. 140). This disease is characterized by persistent inflammatory synovitis that causes destruction of cartilage and bone erosion, leading to structural deformities in the peripheral joints.
  • the symptoms associated with rheumatoid arthritis include joint swelling, joint tenderness, inflammation, morning stiffness, and pain, especially upon flexing.
  • Subjects having advanced stages of arthritis suffer from structural damage, including joint destruction with bone erosion (in: “Principals of Internal Medicine, Harrison, 13 th edition, pages 1648-1655).
  • patients can present other clinical symptoms of various organic lesions, including lesions of the skin, kidney, heart, lung, central nervous system, and eyes due to vasculitis related to the autoimmune process.
  • erythrocyte sedimentation rates and elevated levels of serum C-reactive protein (CRP) and/or soluble IL-2 receptor (IL-2r).
  • CRP serum C-reactive protein
  • IL-2r soluble IL-2 receptor
  • the erythrocyte sedimentation rate is increased in nearly all patients with active rheumatoid arthritis.
  • the level of serum C-reactive protein is also elevated and correlates with disease activity and the likelihood of progressive joint damage.
  • the level of soluble IL-2r a product of activated T-cells, is elevated in blood serum and synovial fluid of patients with active rheumatoid arthritis (see: “Principals of Internal Medicine, Harrison, 13 th edition, page 1650).
  • T-cell surface receptors CD28 and CTLA4, and their ligands, such as B7-related molecules CD80 (i.e., B7-1) and CD86 (i.e., B7-2), on antigen presenting cells (Linsley, P. and Ledbetter, J. 1993 Ann. Rev. Immunol. 11:191-212).
  • rheumatoid arthritis is thought to be a T-cell-mediated immune system disease
  • one strategy to develop new agents to treat rheumatoid arthritis is to identify molecules that block co-stimulatory signals between T-lymphocytes and antigen presenting cells, by blocking the interaction between endogenous CD28 or CTLA4 and B7.
  • Potential molecules include soluble CTLA4 molecules that are modified (i.e. CTLA4 mutant molecules) to bind to B7 with higher avidity than wildtype CTLA4 (the sequence of which is shown in FIG. 23) or CD28, thereby blocking the co-stimulatory signals.
  • Soluble forms of CD28 and CTLA4 have been constructed by fusing variable (V)-like extracellular domains of CD28 and CTLA4 to immunoglobulin (Ig) constant domains resulting in CD28Ig and CTLA4Ig.
  • V variable
  • Ig immunoglobulin
  • a nucleotide and amino acid sequence of CTLA4Ig is shown in FIG. 24 with the protein beginning with methionine at position +1 or alanine at position ⁇ 1 and ending with lysine at position +357.
  • CTLA4Ig binds both CD80-positive and CD86-positive cells more strongly than CD28Ig (Linsley, P., et al., 1994 Immunity 1:793-80).
  • CTLA4Ig fusion molecules were modified by mutation of amino acids in the CTLA4 portion of the molecules.
  • Regions of CTLA4 that, when mutated, alter the binding affinity or avidity for B7 ligands include the complementarity determining region 1 (CDR-1 as described in U.S. Pat. Nos. 6,090,914, 5,773,253, 5,844,095; in copending U.S. Patent Application Serial No. 60/214,065; and by Peach et al, 1994. J. Exp.
  • CDR-3 complementarity determining region 3
  • CDR-3 is the conserved region of the CTLA4 extracellular domain as described in U.S. Pat. Nos. 6,090,914, 5,773,253 and 5,844,095; in copending U.S. Patent Application Serial No. 60/214,065; and by Peach, R. J., et al J Exp Med 1994 180:2049-2058; the CDR-3-like region encompasses the CDR-3 region and extends, by several amino acids, upstream and/or downstream of the CDR-3 motif).
  • the CDR-3-like region includes a hexapeptide motif MYPPPY (SEQ ID NO.: 20) that is highly conserved in all CD28 and CTLA4 family members. Alanine scanning mutagenesis through the hexapeptide motif in CTLA4, and at selected residues in CD28Ig, reduced or abolished binding to CD80 (Peach, R. J., et al J Exp Med 1994 180:2049-2058; U.S. Pat. No. 5,434,131; U.S. Pat. No. 6,090,914; U.S. Pat. No. 5,773,253.
  • CTLA4Ig molecules Further modifications were made to soluble CTLA4Ig molecules by interchanging homologous regions of CTLA4 and CD28.
  • These chimeric CTLA4/CD28 homologue mutant molecules identified the MYPPPY hexapeptide motif common to CTLA4 and CD28, as well as certain non-conserved amino acid residues in the CDR-1- and CDR-3-like regions of CTLA4, as regions responsible for increasing the binding avidity of CTLA4 with CD80 (Peach, R. J., et al., 1994 J Exp Med 180:2049-2058).
  • Soluble CTLA4 molecules such as CTLA4Ig, CTLA4 mutant molecules or chimeric CTLA4/CD28 homologue mutants as described supra, introduce a new group of therapeutic drugs to treat rheumatic diseases.
  • rheumatic diseases such as rheumatoid arthritis
  • DMARDs Disease Modifying Anti-Rheumatic Drugs
  • TNF ⁇ tumor necrosis factor-alpha
  • these drugs merely slow down the progress of the rheumatoid arthritis, which resumes at an accelerated pace after the therapy is discontinued. Additionally, prolonged therapy with these nonspecific drugs produces toxic side effects, including a tendency towards development of certain malignancies, kidney failure, bone marrow suppression, pulmonary fibrosis, malignancy, diabetes, and liver function disorders. These drugs may also gradually cease being effective after about 2-5 years (Kelley's Textbook of Rheumatology, 6 th Edition, pages 1001-1022). Newer, biologically based, DMARDs such as cytokine blockers may be more potent and may have longer lasting effects than older DMARDS such as hydrochloroquine, however, the long term safety of these newer drugs is still unknown. Reports of multiple sclerosis and lupus exist with the use of TNF blockers.
  • NSAIDS Non-Steroidal Anti-Inflammatory Drugs
  • steroid compounds e.g., corticosteroids or glucocorticoids
  • prednisone and methylprednisolone e.g., prednisone and methylprednisolone.
  • Steroids also have significant toxic side effects associated with their long-term use. (Kelley's Textbook of Rheumatology, 6 th Edition, pages 829-833).
  • the present invention provides compositions and methods for treating immune system diseases, by administering to a subject soluble CTLA4 molecules, which bind to B7 molecules on B7-positive cells, thereby inhibiting endogenous B7 molecules from binding CTLA4 and/or CD28 on T-cells.
  • Soluble CTLA4 molecules used in the methods of the invention include CTLA4Ig and soluble CTLA4 mutant molecule L104EA29YIg.
  • the present invention provides compositions and methods for treating immune system diseases, by administering to a subject a combination of a DMARD and a molecule that blocks B7 interaction with CTLA4 and/or CD28.
  • the present invention also provides methods for treating (e.g. reducing symptoms of) rheumatic diseases, such as rheumatoid arthritis, by administering to a subject suffering from symptoms of arthritis, soluble CTLA4 molecules such as CTLA4Ig and/or soluble CTLA4 mutant molecule L104EA29YIg and/or a mix of any soluble CTLA molecule.
  • soluble CTLA4 molecules such as CTLA4Ig and/or soluble CTLA4 mutant molecule L104EA29YIg and/or a mix of any soluble CTLA molecule.
  • the CTLA4 mutant molecule L104EA29YIg e.g. beginning with methionine at position +1 or alanine at position ⁇ 1 and ending with lysine at position +357, as shown in FIG. 19, is preferred for use in the methods of the invention.
  • the present invention also provides methods for treating (e.g. reducing symptoms of) rheumatic diseases, such as rheumatoid arthritis, by administering to the subject a combination of 1) a DMARD, such as methotrexate or a molecule that blocks TNF interactions, and 2) soluble CTLA4 molecules, such as CTLA4Ig.
  • a DMARD such as methotrexate or a molecule that blocks TNF interactions
  • CTLA4Ig soluble CTLA4 molecules
  • the present invention also provides methods for reducing pathophysiological changes associated with an immune system disease (e.g., rheumatic disease), such as structural damage, by administering to the subject diagnosed with the immune system disease (e.g., rheumatoid arthritis), soluble CTLA4 molecules alone or in conjunction with other therapeutic drugs, such as a DMARD.
  • an immune system disease e.g., rheumatic disease
  • soluble CTLA4 molecules alone or in conjunction with other therapeutic drugs, such as a DMARD.
  • the present invention also provides a pharmaceutical composition for treating immune system diseases, such as rheumatic diseases, comprising a pharmaceutically acceptable carrier and a biologically effective agent, such as soluble CTLA4 molecules, alone or in conjunction with other therapeutic drugs, such as a DMARD, a NSAID, a corticosteroid and/or a glucocorticoid.
  • immune system diseases such as rheumatic diseases
  • a pharmaceutically acceptable carrier such as a biologically effective agent, such as soluble CTLA4 molecules, alone or in conjunction with other therapeutic drugs, such as a DMARD, a NSAID, a corticosteroid and/or a glucocorticoid.
  • Kits comprising pharmaceutical compositions therapeutic for immune system disease are also encompassed by the invention.
  • a kit comprising one or more of the pharmaceutical compositions of the invention is used to treat an immune system disease, e.g. rheumatoid arthritis.
  • the pharmaceutical composition comprises an effective amount of soluble CTLA4 molecules that bind to B7 molecules on B7-positive cells, thereby blocking the B7 molecules from binding CTLA4 and/or CD28 on T-cells.
  • the kit may contain one or more immunosuppressive agents used in conjunction with the pharmaceutical compositions of the invention.
  • Potential immunosuppressive agents include, but are not limited to, corticosteroids, nonsteroidal antiinflammatory drugs (e.g.
  • Cox-2 inhibitors prednisone, cyclosporine, cyclosporin A, azathioprine, methotrexate, TNF ⁇ blockers or antagonists, hydroxychloroquine, sulphasalazopyrine (sulfasalazine), gold salts, infliximab, etanercept, anakinra and any biological agent targeting an inflammatory cytokine.
  • the present invention also provides methods for reducing the erythrocyte sedimentation rate that is associated with rheumatoid arthritis.
  • the present invention provides methods for reducing the levels of certain components of blood serum which are associated with rheumatoid arthritis, including C-reactive protein, IL-6, TNF- ⁇ , soluble ICAM-1, soluble E-selectin and/or soluble IL-2r.
  • FIG. 1B Demographic data of patient cohorts. Demographic data including gender, age, weight, and disease activity, evaluated by the patient and by the physician, as described in Example 3, infra.
  • FIG. 1C Demographic data of patient cohorts as described in Example 3, infra. Demographic data including disease activity, erythrocyte sedimentation rate (ESR), physical function (disability evaluated by health questionnaire), and C-reactive protein (CRP).
  • ESR erythrocyte sedimentation rate
  • CRP C-reactive protein
  • FIG. 1E Demographic data of patient cohorts as described in Example 3, infra. Demographic data including prior treatments.
  • FIG. 2 Summary of discontinuations at day 85 by reason as described in Example 3, infra.
  • FIG. 3A ACR responses at Day 85 as described in Example 3, infra: ACR-20, ⁇ 50, and ⁇ 70 responses.
  • FIG. 3B ACR-20 responses at Day 85, including placebo response, as described in Example 3, infra: ACR-20 response with 95% confidence limits.
  • FIG. 3C ACR-20 responses at Day 85 as described in Example 3, infra: Difference in ACR-20 response with respect to 95% confidence intervals.
  • FIG. 4A Basic (20% improvement) clinical responses in swollen and tender joint count in percentage of patients at Day 85 as described in Example 3, infra: basic clinical response, ACR-20.
  • FIG. 4B Clinical responses (in percentage improvement) in swollen and tender joint count in percentage of patients at Day 85 as described in Example 3, infra: change in clinical response in percentage improvement.
  • FIG. 5A Pain response (by Likert scale by mean unit change from baseline) in percentage of patients at Day 85 as described in Example 3, infra: pain score changes from baseline.
  • FIG. 5B Patient global disease changes (by Likert scale by mean unit change from baseline) in percentage of patients at Day 85 as described in Example 3, infra: patient global disease activity changes.
  • FIG. 5C Physician global disease changes (by Likert scale by mean unit change from baseline) in percentage of patients at Day 85 as described in Example 3, infra: physician global disease activity changes.
  • FIG. 5D Pain (by Likert scale by mean unit change from baseline) in percentage of patients at Day 85 as described in Example 3, infra: pain changes from baseline.
  • FIG. 6A Patient global assessment of disease activity change from baseline by range of 2 units at Day 85 as described in Example 3, infra; disease activity improvement.
  • FIG. 7A Percent reduction in C-reactive protein (CRP) levels at Day 85 as described in Example 3, infra: percentage reduction in CRP levels from baseline.
  • FIG. 7B Difference in reduction in C-reactive protein (CRP) levels at Day 85 as described in Example 3, infra: percent reduction difference in CRP levels with 95% confidence intervals.
  • FIG. 7C Mean reduction in C-reactive protein (CRP) levels at Day 85 as described in Example 3, infra: mean change from baseline.
  • FIG. 8 Reduction in soluble IL-2 receptor levels mean change from baseline at Day 85 as described in Example 3, infra.
  • FIG. 9A The effect of CTLA4Ig on tender joints over time as described in Example 3, infra: median difference from baseline.
  • FIG. 9B The effect of CTLA4Ig on tender joints over time as described in Example 3, infra: mean difference from baseline.
  • FIG. 10A The effect of CTLA4Ig on swollen joints over time as described in Example 3, infra: median difference from baseline.
  • FIG. 10B The effect of CTLA4Ig on swollen joints over time as described in Example 3, infra: mean difference from baseline.
  • FIG. 11 The effect of CTLA4Ig on pain assessment mean difference from baseline over time as described in Example 3, infra.
  • FIG. 12A The effect of CTLA4Ig on patient assessment of disease activity mean difference from baseline over time as described in Example 3, infra.
  • FIG. 12B The effect of CTLA4Ig on physician assessment of disease activity mean difference from baseline over time as described in Example 3, infra.
  • FIG. 13A The effect of L104EA29YIg on tender joints over time as described in Example 3, infra: median difference from baseline.
  • FIG. 13B The effect of L104EA29YIg on tender joints over time as described in Example 3, infra: mean change from baseline.
  • FIG. 14A The effect of L104EA29YIg on swollen joints over time as described in Example 3, infra: median difference from baseline.
  • FIG. 14B The effect of L104EA29YIg on swollen joints over time as described in Example 3, infra: mean change from baseline.
  • FIG. 15 The effect of L104EA29YIg on pain assessment over time as described in Example 3, infra: mean change from baseline over time.
  • FIG. 16A The effect of L104EA29YIg on patient assessment of disease activity mean difference from baseline over time as described in Example 3, infra.
  • FIG. 17 Percent improvement in patient disability assessed by Health Assessment Questionnaire (HAQ) compared to the baseline at Day 85 with CTLA4Ig and L104EA29YIg treatment as described in Example 3, infra.
  • HAQ Health Assessment Questionnaire
  • FIG. 18 Nucleotide and amino acid sequence of L104EIg (SEQ ID NOs: 6-7) as described in Example 1, infra.
  • FIG. 19 Nucleotide and amino acid sequence of L104EA29YIg (SEQ ID NOs: 8-9) as described in Example 1, infra.
  • FIG. 20 Nucleotide and amino acid sequence of L104EA29LIg (SEQ ID NOs: 10-11) as described in Example 1, infra.
  • FIG. 21 Nucleotide and amino acid sequence of L104EA29TIg (SEQ ID NOs: 12-13) as described in Example 1, infra.
  • FIG. 22 Nucleotide and amino acid sequence of L104EA29WIg (SEQ ID NOs: 14-15) as described in Example 1, infra.
  • FIG. 23 Nucleotide and amino acid sequence of CTLA4 receptor (SEQ ID NOs: 16-17).
  • FIG. 24 Nucleotide and amino acid sequence of CTLA4Ig (SEQ ID NOs: 18-19).
  • FIG. 25 SDS gel (FIG. 25A) for CTLA4Ig (lane 1), L104EIg (lane 2), and L104EA29YIg (lane 3A); and size exclusion chromatographs of CTLA4Ig (FIG. 25B) and L104EA29YIg (FIG. 25C).
  • FIG. 26 (left and right depictions): A ribbon diagram of the CTLA4 extracellular Ig V-like fold generated from the solution structure determined by NMR spectroscopy.
  • FIG. 26 (right depiction) shows an expanded view of the CDR-1 (S25-R33) region and the MYPPPY region indicating the location and side-chain orientation of the avidity enhancing mutations, L104 and A29.
  • FIGS. 27 A & 27 B FACS assays showing binding of L104EA29YIg, L104EIg, and CTLA4Ig to human CD80- or CD86-transfected CHO cells as described in Example 2, infra.
  • FIGS. 28 A & 28 B Graphs showing inhibition of proliferation of CD80-positive and CD86-positive CHO cells as described in Example 2, infra.
  • FIG. 32 A graph showing the equilibrium binding analysis of L104EA29YIg, L104EIg, and wild-type CTLA4Ig to CD86Ig.
  • FIG. 35 A graph showing the summary of ACR50 response by visit day in response to methotrexate alone or methotrexate and CTLA4Ig (2 and 10 mg/kg) therapy, as described in Example 5, infra.
  • FIG. 36 A graph showing the summary of ACR70 response by visit day in response to methotrexate alone or methotrexate and CTLA4Ig (2 and 10 mg/kg) therapy, as described in Example 5, infra.
  • FIG. 39 A bar graph showing the proportion of New Active Joints in response to methotrexate alone or methotrexate and CTLA4Ig (2 and 10 mg/kg) therapy on day 180, as described in Example 5, infra.
  • FIG. 40 A bar graph showing ACR response after therapy with methotrexate alone or methotrexate and CTLA4Ig (2 and 10 mg/kg) on day 180, as described in Example 5, infra.
  • FIG. 41 A graph showing percent improvement in tender joints after therapy with methotrexate alone or methotrexate and CTLA4Ig (2 and 10 mg/kg)—mean percent improvement from baseline, as described in Example 5, infra.
  • FIG. 42 A graph showing percent improvement in swollen joints after therapy with methotrexate alone or methotrexate and CTLA4Ig (2 and 10 mg/kg)—mean percent improvement from baseline, as described in Example 5, infra.
  • FIG. 43 A graph showing percent improvement in pain after therapy with methotrexate alone or methotrexate and CTLA4Ig (2 and 10 mg/kg)—mean percent improvement from baseline, as described in Example 5, infra.
  • FIG. 44 A graph showing percent improvement in regard to disease activity as reported by the subject after therapy with methotrexate alone or methotrexate and CTLA4Ig (2 and 10 mg/kg)—mean percent improvement from baseline, as described in Example 5, infra.
  • FIG. 45 A graph showing percent improvement in regard to disease activity as reported by the physician after therapy with methotrexate alone or methotrexate and CTLA4Ig (2 and 10 mg/kg)—mean percent improvement from baseline, as described in Example 5, infra.
  • FIG. 46 A graph showing percent improvement regarding physical function after therapy with methotrexate alone or methotrexate and CTLA4Ig (2 and 10 mg/kg)—mean percent improvement from baseline as measured by HAQ, as described in Example 5, infra.
  • FIG. 47 A graph showing percent improvement in CRP levels function after therapy with methotrexate alone or methotrexate and CTLA4Ig (2 and 10 mg/kg)—mean percent improvement from baseline, as described in Example 5, infra.
  • FIG. 48 A graph showing percent improvement in CRP levels function after therapy with methotrexate alone or methotrexate and CTLA4Ig (2 and 10 mg/kg)—median percent improvement from baseline, as described in Example 5, infra.
  • FIG. 49 A graph showing the difference in ACR response rate on day 180 in two groups after therapy with CTLA4Ig (2 and 10 mg/kg) in comparison to a group treated with methotrexate (MTX) only (95% Confidence Limits), as described in Example 5, infra.
  • MTX methotrexate
  • FIG. 50 A graph showing the change from baseline for SF-36 Physical Health Component on day 180, in two groups after therapy with CTLA4Ig (2 and 10 mg/kg) compared to a group treated with methotrexate only (95% Confidence Limits), as described in Example 5, infra.
  • FIG. 51 A graph showing the change from baseline for SF-36 Mental Health Component on Day 180, in two groups after therapy with CTLA4Ig (2 and 10 mg/kg) compared to a group treated with methotrexate only (95% Confidence Limits), as described in Example 5, infra.
  • FIG. 52 A bar graph showing CRP levels at day 180 after therapy with methotrexate alone or methotrexate and CTLA4Ig (2 and 10 mg/kg), as described in Example 5, infra.
  • FIG. 54 A bar graph showing IL-2r levels on day 180 after therapy with methotrexate alone or methotrexate and CTLA4Ig (2 and 10 mg/kg), as described in Example 5, infra.
  • FIG. 55 A bar graph showing IL-6 levels on day 180 after therapy with methotrexate alone or methotrexate and CTLA4Ig (2 and 10 mg/kg), as described in Example 5, infra.
  • FIG. 56 A bar graph showing TNF ⁇ levels on day 180 after therapy with methotrexate alone or methotrexate and CTLA4Ig (2 and 10 mg/kg), as described in Example 5, infra.
  • FIG. 58 A table of the univariate methotrexate dose at screening/enrollment for treatment group BMS 2-treated with CTLA4Ig at 2 mg/kg body weight as described in Example 5, infra.
  • FIG. 59 A table of the univariate methotrexate dose at screening/enrollment for the placebo group, as described in Example 5, infra.
  • FIG. 60 A table of the univariate methotrexate dose up to and including day 180 of the study for treatment group BMS 10-treated with CTLA4Ig at 10 mg/kg body weight as described in Example 5, infra.
  • FIG. 61 A table of the univariate methotrexate dose up to and including day 180 of the study for treatment group BMS 2-treated with CTLA4Ig at 2 mg/kg body weight as described in Example 5, infra.
  • FIG. 62 A table of the univariate methotrexate dose up to and including day 180 of the study for the placebo group, as described in Example 5, infra.
  • FIG. 63 A bar graph showing the difference in modified ACR response rates on day 180 in two groups after therapy with etanercept alone (25 mg twice weekly) or in combination with CTLA4Ig (2 mg/kg), as described in Example 6, infra.
  • FIGS. 64 A-C Graphs showing percentage improvement of individual components of the modified ACR criteria as assessed on each visit day after therapy with etanercept alone (25 mg twice weekly) or in combination with CTLA4Ig (2 mg/kg) as described in Example 6, infra.
  • FIG. 65 A graph showing the change from baseline for SF-36 Physical Health Component on day 180, in two groups after therapy with etanercept (25 mg biweekly) alone or in combination with CTLA4Ig (2 mg/kg) (95% Confidence Limits), as described in Example 6, infra.
  • B A graph showing the change from baseline for SF-36 Mental Health Component on day 180, in two groups after therapy with etanercept (25 mg biweekly) alone or in combination with CTLA4Ig (2 mg/kg) (95% Confidence Limits), as described in Example 6, infra.
  • FIG. 66 Nucleotide sequence of a CTLA4Ig encoding a signal peptide; a wild type amino acid sequence of the extracellular domain of CTLA4 starting at methionine at position +1 to aspartic acid at position +124, or starting at alanine at position ⁇ 1 to aspartic acid at position +124; and an Ig region (SEQ ID NO.: 21).
  • FIG. 67 Amino acid sequence of a CTLA4Ig having a signal peptide; a wild type amino acid sequence of the extracellular domain of CTLA4 starting at methionine at position +1 to aspartic acid at position +124, or starting at alanine at position ⁇ 1 to aspartic acid at position +124; and an Ig region (SEQ ID NO.: 22).
  • FIG. 68 A schematic diagram showing the disposition of subjects into three cohorts as described in Example 7, infra.
  • FIG. 70 A Kaplan-Meier plot of the cumulative proportion of subjects who discontinued due to lack of efficacy during the first 12 months of study, as described in Example 7, infra.
  • FIG. 71A A graph showing the ACR Responses on Day 180 for patients administered methotrexate alone or methotrexate and CTLA4Ig (2 or 10 mg/kg body weight) as described in Example 7, infra.
  • FIG. 71B A graph showing the 95 Percent Confidence Intervals for Differences in ACR Responses on Day 180 for patients administered methotrexate alone or methotrexate and CTLA4Ig (2 or 10 mg/kg body weight) as described in Example 7, infra.
  • FIG. 72A A graph showing the ACR Responses on Day 360 for patients administered methotrexate alone or methotrexate and CTLA4Ig (2 or 10 mg/kg body weight) as described in Example 7, infra.
  • FIG. 72B A graph showing the 95 Percent Confidence Intervals for Differences in ACR Responses on Day 360 for patients administered methotrexate alone or methotrexate and CTLA4Ig (2 or 10 mg/kg body weight) as described in Example 7, infra.
  • FIG. 73A A graph summarizing the ACR 20 Response by Visit during a one year interval for patients administered methotrexate alone or methotrexate and CTLA4Ig (2 or 10 mg/kg body weight) as described in Example 7, infra.
  • FIG. 74 A graph showing the Mean ACR-N over a one year time interval for patients administered methotrexate alone or methotrexate and CTLA4Ig (2 or 10 mg/kg body weight) as described in Example 7, infra.
  • FIG. 75 A graph showing the Proportion of New Active Joints at Day 180 for patients administered methotrexate alone or methotrexate and CTLA4Ig (2 or 10 mg/kg body weight) as described in Example 7, infra.
  • FIG. 76A A graph showing the Frequency of New Tender Joints per Subject at Day 180 for patients administered methotrexate alone or methotrexate and CTLA4Ig (2 or 10 mg/kg body weight) as described in Example 7, infra.
  • FIG. 76B A graph showing the Frequency of New Tender Joints per Subject at Day 360 for patients administered methotrexate alone or methotrexate and CTLA4Ig (2 or 10 mg/kg body weight) as described in Example 7, infra.
  • FIG. 77A A graph showing the Frequency of New Swollen Joints per Subject at Day 180 for patients administered methotrexate alone or methotrexate and CTLA4Ig (2 or 10 mg/kg body weight) as described in Example 7, infra.
  • FIG. 77B A graph showing the Frequency of New Swollen Joints per Subject at Day 360 for patients administered methotrexate alone or methotrexate and CTLA4Ig (2 or 10 mg/kg body weight) as described in Example 7, infra.
  • FIG. 78 A graph showing the Proportion of New Active Joints at Day 360 for patients administered methotrexate alone or methotrexate and CTLA4Ig (2 or 10 mg/kg body weight) as described in Example 7, infra.
  • FIG. 79 Graphs showing the: A) Change from Baseline in the Physical Health Domains on Day 180, and B) Change from Baseline in the Mental Health Domains on 180, for patients administered methotrexate alone or methotrexate and CTLA4Ig (2 or 10 mg/kg body weight) as described in Example 7, infra.
  • FIG. 80 Graphs showing the: A) Change from Baseline in the Physical Health Domains on Day 360, and B) Change from Baseline in the Mental Health Domains on Day 360, for patients administered methotrexate alone or methotrexate and CTLA4Ig (2 or 10 mg/kg body weight) as described in Example 7, infra.
  • FIG. 81 A graph showing the Soluble IL-2r Levels at Baseline, Days 180 and 360 for patients administered methotrexate alone or methotrexate and CTLA4Ig (2 or 10 mg/kg body weight) as described in Example 7, infra.
  • FIG. 82 A graph showing the Rheumatoid Factor Levels at Baseline, Days 180 and 360 for patients administered methotrexate alone or methotrexate and CTLA4Ig (2 or 10 mg/kg body weight) as described in Example 7, infra.
  • FIG. 83 A graph showing the ICAM-1 Levels at Baseline, Days 180 and 360 for patients administered methotrexate alone or methotrexate and CTLA4Ig (2 or 10 mg/kg body weight) as described in Example 7, infra.
  • FIG. 84 A graph showing the e-Selectin Levels at Baseline, Days 180 and 360 for patients administered methotrexate alone or methotrexate and CTLA4Ig (2 or 10 mg/kg body weight) as described in Example 7, infra.
  • FIG. 85 A graph showing the Serum IL-6 at Baseline, Days 180 and 360 for patients administered methotrexate alone or methotrexate and CTLA4Ig (2 or 10 mg/kg body weight) as described in Example 7, infra.
  • FIG. 86A A graph showing the CRP Levels at Baseline, Days 180 and 360 for patients administered methotrexate alone or methotrexate and CTLA4Ig (2 or 10 mg/kg body weight) as described in Example 7, infra.
  • FIG. 86B A graph showing the TNF ⁇ Levels at Baseline, Days 180 and 360 for patients administered methotrexate alone or methotrexate and CTLA4Ig (2 or 10 mg/kg body weight) as described in Example 7, infra.
  • DMARD refers to a Disease Modifying Anti-Rheumatic Drug.
  • a DMARD is any agent that modifies the symptoms and/or progression associated with an immune system disease, including autoimmune diseases (e.g. rheumatic diseases), graft-related disorders and immunoproliferative diseases.
  • DMARDs modify one or more of the symptoms and/or disease progression associated with rheumatic disease.
  • Symptoms of rheumatic diseases include the following: joint swelling, pain, tenderness, morning stiffness, structural damage, an elevated level of serum C-reactive protein (CRP), an elevated level of soluble IL-2r, an elevated level of soluble ICAM-1, an elevated level of soluble E-selectin, an elevated level of rheumatoid factor, an elevated level of IL-6 or an elevated erythrocyte sedimentation rate (ESR).
  • CRP serum C-reactive protein
  • ICAM-1 an elevated level of soluble E-selectin
  • ESR erythrocyte sedimentation rate
  • DMARDs include, but are not limited to, dihydrofolic acid reductase inhibitors e.g., methotrexate; cyclophosphamide; cyclosporine; cyclosporin A; chloroquine; hydroxychloroquine; sulfasalazine (sulphasalazopyrine) gold salts D-penicillamine; leflunomide; azathioprine; anakinra; TNF blockers e.g., infliximab (REMICADER) or etanercept; and a biological agent that targets an inflammatory cytokine.
  • dihydrofolic acid reductase inhibitors e.g., methotrexate; cyclophosphamide; cyclosporine; cyclosporin A; chloroquine; hydroxychloroquine; sulfasalazine (sulphasalazopyrine) gold salts D-penicill
  • NSAID refers to a Non-Steroidal Anti-Inflammatory Drug.
  • NSAIDs reduce inflammatory reactions in a subject.
  • NSAIDs include, but are not limited to acetyl salicylic acid, choline magnesium salicylate, diflunisal, magnesium salicylate, salsalate, sodium salicylate, diclofenac, etodolac, fenoprofen, flurbiprofen, indomethacin, ketoprofen, ketorolac, meclofenamate, naproxen, nabumetone, phenylbutazone, piroxicam, sulindac, tolmetin, acetaminophen, ibuprofen, Cox-2 inhibitors, meloxicam and tramadol.
  • ligand refers to a molecule that specifically recognizes and binds another molecule
  • a ligand for CTLA4 is a B7 molecule.
  • a ligand for the B7 molecule is a CTLA4 and/or CD28 molecule.
  • CTLA4 interaction with its ligand B7 can be blocked by administration of CTLA4Ig molecules.
  • Tumor Necrosis Factor (TNF) interaction with its ligand, TNF receptor (TNFR) can be blocked by administration of etanercept or other TNF/TNFR blocking molecules.
  • TNF Tumor Necrosis Factor
  • wild type CTLA4 or “non-mutated CTLA4” has the amino acid sequence of naturally occurring, full length CTLA4 as shown in FIG. 23 (also as described in U.S. Pat. Nos. 5,434,131, 5,844,095, and 5,851,795 herein incorporated by reference in their entirety), or any portion or derivative thereof, that recognizes and binds a B7 or interferes with a B7 so that it blocks binding to CD28 and/or CTLA4 (e.g., endogenous CD28 and/or CTLA4).
  • CD28 and/or CTLA4 e.g., endogenous CD28 and/or CTLA4
  • the extracellular domain of wild type CTLA4 begins with methionine at position +1 and ends at aspartic acid at position +124, or the extracellular domain of wild type CTLA4 begins with alanine at position ⁇ 1 and ends at aspartic acid at position +124 as shown in FIG. 23.
  • Wild type CTLA4 is a cell surface protein, having an N-terminal extracellular domain, a transmembrane domain, and a C-terminal cytoplasmic domain. The extracellular domain binds to target molecules, such as a B7 molecule.
  • target molecules such as a B7 molecule.
  • the naturally occurring, wild type CTLA4 protein is translated as an immature polypeptide, which includes a signal peptide at the N-terminal end.
  • the immature polypeptide undergoes post-translational processing, which includes cleavage and removal of the signal peptide to generate a CTLA4 cleavage product having a newly generated N-terminal end that differs from the N-terminal end in the immature form.
  • post-translational processing includes cleavage and removal of the signal peptide to generate a CTLA4 cleavage product having a newly generated N-terminal end that differs from the N-terminal end in the immature form.
  • additional post-translational processing may occur, which removes one or more of the amino acids from the newly generated N-terminal end of the CTLA4 cleavage product.
  • the signal peptide may not be removed completely, generating molecules that begin before the common starting amino acid methionine.
  • the mature CTLA4 protein may start at methionine at position +1 or alanine at position ⁇ 1.
  • the mature form of the CTLA4 molecule includes the extracellular domain or any portion thereof, which
  • a “CTLA4 mutant molecule” means wildtype CTLA4 as shown in FIG. 23 or any portion or derivative thereof, that has a mutation or multiple mutations (preferably in the extracellular domain of wildtype CTLA4).
  • a CTLA4 mutant molecule has a sequence that it is similar but not identical to the sequence of wild type CTLA4 molecule, but still binds a B7.
  • the mutations may include one or more amino acid residues substituted with an amino acid having conservative (e.g., substitute a leucine with an isoleucine) or non-conservative (e.g., substitute a glycine with a tryptophan) structure or chemical properties, amino acid deletions, additions, frameshifts, or truncations.
  • CTLA4 mutant molecules may include a non-CTLA4 molecule therein or attached thereto.
  • the mutant molecules may be soluble (i.e., circulating) or bound to a cell surface.
  • Additional CTLA4 mutant molecules include those described in U.S. patent application Ser. Nos. 09/865,321, 60/214,065 and 60/287,576; in U.S. Pat. Nos. 6,090,914 5,844,095 and 5,773,253; and as described by Peach, R. J., et al., in J Exp Med 180:2049-2058 (1994)).
  • CTLA4 mutant molecules can be made synthetically or recombinantly.
  • CTLA4Ig is a soluble fusion protein comprising an extracellular domain of wildtype CTLA4 that binds B7, or a portion thereof, joined to an immunoglobulin constant region (Ig), or a portion thereof.
  • a particular embodiment comprises the extracellular domain of wild type CTLA4 (as shown in FIG.
  • CTLA4Ig-24 a Chinese Hamster Ovary (CHO) cell line expressing CTLA4Ig was deposited on May 31, 1991 with ATCC identification number CRL-10762).
  • the soluble CTLA4Ig molecules used in the methods and/or kits of the invention may or may not include a signal (leader) peptide sequence. Typically, in the methods and/or kits of the invention, the molecules do not include a signal peptide sequence.
  • L104EA29YIg is a fusion protein that is a soluble CTLA4 mutant molecule comprising an extracellular domain of wildtype CTLA4 with amino acid changes A29Y (a tyrosine amino acid residue substituting for an alanine at position 29) and L104E (a glutamic acid amino acid residue substituting for a leucine at position +104), or a portion thereof that binds a B7 molecule, joined to an Ig tail (included in FIG. 19; DNA encoding L104EA29YIg was deposited on Jun. 20, 2000 with ATCC number PTA-2104; copending in U.S. patent application Ser. Nos.
  • the soluble L104EA29YIg molecules used in the methods and/or kits of the invention may or may not include a signal (leader) peptide sequence. Typically, in the methods and/or kits of the invention, the molecules do not include a signal peptide sequence.
  • soluble refers to any molecule, or fragments and derivatives thereof, not bound or attached to a cell, i.e., circulating.
  • CTLA4, B7 or CD28 can be made soluble by attaching an immunoglobulin (Ig) moiety to the extracellular domain of CTLA4, B7 or CD28, respectively.
  • Ig immunoglobulin
  • a molecule such as CTLA4 can be rendered soluble by removing its transmembrane domain.
  • the soluble molecules used in the methods, compositions and/or kits of the invention do not include a signal (or leader) sequence.
  • soluble CTLA4 molecules means non-cell-surface-bound (i.e. circulating) CTLA4 molecules or any functional portion of a CTLA4 molecule that binds B7 including, but not limited to: CTLA4Ig fusion proteins (e.g.
  • CTLA4 immunoglobulin
  • Ig immunoglobulin
  • IgC ⁇ 1 IgCgamma1
  • IgC ⁇ 2 IgCgamma2
  • IgC ⁇ 3 IgCgamma3
  • IgC ⁇ 4 IgCgamma4
  • IgC ⁇ (IgCmu) IgC ⁇ 1 (IgCalpha1), IgC ⁇ 2 (IgCalpha2), IgC ⁇ (IgCdelta) or IgC ⁇ (IgCepsilon
  • proteins with the extracellular domain of CTLA4 fused or joined with a portion of a biologically active or chemically active protein such as the papillomavirus E7 gene product (CTLA4-E7), melanoma-associated antigen p97 (CTLA4-p97) or HIV env protein
  • the extracellular domain of CTLA4 is the portion of CTLA4 that recognizes and binds CTLA4 ligands, such as B7 molecules.
  • an extracellular domain of CTLA4 comprises methionine at position +1 to aspartic acid at position +124 (FIG. 23).
  • an extracellular domain of CTLA4 comprises alanine at position ⁇ 1 to aspartic acid at position +124 (FIG. 23).
  • the extracellular domain includes fragments or derivatives of CTLA4 that bind a B7 molecule.
  • the extracellular domain of CTLA4 as shown in FIG. 23 may also include mutations that change the binding avidity of the CTLA4 molecule for a B7 molecule.
  • mutation means a change in the nucleotide or amino acid sequence of a wildtype molecule, for example, a change in the DNA and/or amino acid sequences of the wild-type CTLA4 extracellular domain.
  • a mutation in DNA may change a codon leading to a change in the amino acid sequence.
  • a DNA change may include substitutions, deletions, insertions, alternative splicing, or truncations.
  • An amino acid change may include substitutions, deletions, insertions, additions, truncations, or processing or cleavage errors of the protein.
  • mutations in a nucleotide sequence may result in a silent mutation in the amino acid sequence as is well understood in the art.
  • nucleotide codons encode the same amino acid.
  • examples include nucleotide codons CGU, CGG, CGC, and CGA encoding the amino acid, arginine (R); or codons GAU, and GAC encoding the amino acid, aspartic acid (D).
  • a protein can be encoded by one or more nucleic acid molecules that differ in their specific nucleotide sequence, but still encode protein molecules having identical sequences.
  • the amino acid coding sequence is as follows: One Letter Amino Acid Symbol Symbol Codons Alanine Ala A GCU, GCC, GCA, GCG Cysteine Cys C UGU, UGC Aspartic Acid Asp D GAU, GAC Glutamic Acid Glu E GAA, GAG Phenylalanine Phe F UUU, UUC Glycine Gly G GGU, GGC, GGA, GGG Histidine His H CAU, CAC Isoleucine Ile I AUU, AUC, AUA Lysine Lys K AAA, AAG Leucine Leu L UUA, UUG, CUU, CUC, CUA, CUG Methionine Met M AUG Asparagine Asn N AAU, AAC Proline Pro P CCU, CCC, CCA, CCG Glutamine Gln Q CAA, CAG Arginine Arg R CGU, CGC, CGA, CGG, AGA, AGG Serine Ser S UCU, UCC, UCA, UCG,
  • the mutant molecule may have one or more mutations.
  • a “non-CTLA4 protein sequence” or “non-CTLA4 molecule” means any protein molecule that does not bind B7 and does not interfere with the binding of CTLA4 to its target.
  • the non-CTLA4 molecule, attached to the extracellular domain of a CTLA4 molecule can alter the solubility or affinity of the CTLA4 molecule.
  • An example includes, but is not limited to, an immunoglobulin (Ig) constant region or portion thereof.
  • the Ig constant region is a human or monkey Ig constant region, e.g., human C(gamma)1, including the hinge, CH2 and CH3 regions.
  • the Ig constant region can be mutated to reduce its effector functions (U.S. Pat. Nos. 5,637,481, 5,844,095 and 5,434,131).
  • a “fragment” or “portion” is any part or segment of a molecule e.g. CTLA4 or CD28, preferably the extracellular domain of CTLA4 or CD28 or a part or segment thereof, that recognizes and binds its target, e.g., a B7 molecule.
  • B7 refers to the B7 family of molecules including, but not limited to, B7-1 (CD80) (Freeman et al, 1989, J. Immunol. 143:2714-2722, herein incorporated by reference in its entirety), B7-2 (CD86) (Freeman et al, 1993, Science 262:909-911 herein incorporated by reference in its entirety; Azuma et al, 1993, Nature 366:76-79 herein incorporated by reference in its entirety) that may recognize and bind CTLA4 and/or CD28.
  • a B7 molecule can be expressed on an activated B cell.
  • CD28 refers to the molecule that recognizes and binds B7 as described in U.S. Pat. Nos. 5,580,756 and 5,521,288 (herein incorporated by reference in their entirety).
  • B7-positive cells are any cells with one or more types of B7 molecules expressed on the cell surface.
  • a “derivative” is a molecule that shares sequence similarity and activity of its parent molecule.
  • a derivative of CTLA4 includes a soluble CTLA4 molecule having an amino acid sequence at least 70% similar to the extracellular domain of wildtype CTLA4, and which recognizes and binds B7 e.g. CTLA4Ig or soluble CTLA4 mutant molecule L104EA29YIg.
  • a derivative means any change to the amino acid sequence and/or chemical quality of the amino acid e.g., amino acid analogs.
  • an immune response is to activate, stimulate, up-regulate, inhibit, block, down-regulate or modify the immune response.
  • the auto-immune diseases described herein may be treated by regulating an immune response e.g., by regulating functional CTLA4- and/or CD28-positive cell interactions with B7-positive cells.
  • a method for regulating an immune response comprises contacting the B7-positive cells with a soluble CTLA4 molecule of the invention so as to form soluble CTLA4/B7 complexes, the soluble CTLA4 molecule interfering with reaction of an endogenous CTLA4 and/or CD28 molecule with said B7 molecule.
  • a receptor, signal or molecule means to interfere with the activation of the receptor, signal or molecule, as detected by an art-recognized test. For example, blockage of a cell-mediated immune response can be detected by determining reduction of Rheumatic Disease associated symptoms. Blockage or inhibition may be partial or total.
  • blocking B7 interaction means to interfere with the binding of B7 to its ligands, such as CD28 and/or CTLA4, thereby obstructing T-cell and B7-positive cell interactions.
  • agents that block B7 interactions include, but are not limited to, molecules such as an antibody (or portion or derivative thereof) that recognizes and binds to the any of CTLA4, CD28 or B7 molecules (e.g. B7-1, B7-2); a soluble form (or portion or derivative thereof) of the molecules such as soluble CTLA4; a peptide fragment or other small molecule designed to interfere with the cell signal through the CTLA4/CD28/B7-mediated interaction.
  • the blocking agent is a soluble CTLA4 molecule, such as CTLA4Ig (ATCC 68629) or L104EA29YIg (ATCC PTA-2104), a soluble CD28 molecule such as CD28Ig (ATCC 68628), a soluble B7 molecule such as B7Ig (ATCC 68627), an anti-B7 monoclonal antibody (e.g. ATCC HB-253, ATCC CRL-2223, ATCC CRL-2226, ATCC HB-301, ATCC HB-11341 and monoclonal antibodies as described in by Anderson et al in U.S. Pat. No. 6,113,898 or Yokochi et al., 1982. J.
  • CTLA4Ig ATCC 68629
  • L104EA29YIg ATCC PTA-2104
  • a soluble CD28 molecule such as CD28Ig (ATCC 68628)
  • a soluble B7 molecule such as B7Ig (ATCC 68627)
  • Immun., 128(2)823-827 an anti-CTLA4 monoclonal antibody (e.g. ATCC HB-304, and monoclonal antibodies as described in references 82-83) and/or an anti-CD28 monoclonal antibody (e.g. ATCC HB 11944 and mAb 9.3 as described by Hansen (Hansen et al., 1980. Immunogenetics 10: 247-260) or Martin (Martin et al., 1984. J. Clin. Immun., 4(1):18-22)).
  • an anti-CTLA4 monoclonal antibody e.g. ATCC HB-304, and monoclonal antibodies as described in references 82-83
  • an anti-CD28 monoclonal antibody e.g. ATCC HB 11944 and mAb 9.3 as described by Hansen (Hansen et al., 1980. Immunogenetics 10: 247-260) or Martin (Martin et al., 1984. J. Clin. Immun., 4(1):18-22)
  • an “effective amount” of a molecule is defined as an amount that blocks the interaction of the molecule with its ligand.
  • an effective amount of a molecule that blocks B7 interaction with CTLA4 and/or CD28 may be defined as the amount of the molecule that, when bound to B7 molecules on B7-positive cells, inhibit B7 molecules from binding endogenous ligands such as CTLA4 and CD28.
  • an effective amount of a molecule that blocks B7 interaction with CTLA4 and/or CD28 may be defined as the amount of the molecule that, when bound to CTLA4 and/or CD28 molecules on T cells, inhibit B7 molecules from binding endogenous ligands such as CTLA4 and CD28.
  • the inhibition or blockage may be partial or complete.
  • treating means to manage a disease by medicinal or other therapies.
  • Treatment of a disease may ameliorate the symptoms of a disease, reduce the severity of a disease, alter the course of disease progression and/or ameliorate or cure the basic disease problem.
  • to treat an auto-immune disease may be accomplished by regulating an immune response e.g., by regulating functional CTLA4- and/or CD28-positive cell interactions with B7-positive cells.
  • treating an auto-immune disease may be accomplished by preventing the disease from occurring or progressing through the use of the compositions described herein.
  • immune system disease means any disease mediated by T-cell interactions with B7-positive cells including, but not limited to, autoimmune diseases, graft related disorders and immunoproliferative diseases.
  • immune system diseases include graft versus host disease (GVHD) (e.g., such as may result from bone marrow transplantation, or in the induction of tolerance), immune disorders associated with graft transplantation rejection, chronic rejection, and tissue or cell allo- or xenografts, including solid organs (e.g., kidney transplants), skin, islets, muscles, hepatocytes, neurons.
  • GVHD graft versus host disease
  • immunoproliferative diseases include, but are not limited to, psoriasis, T-cell lymphoma, T-cell acute lymphoblastic leukemia, testicular angiocentric T-cell lymphoma, benign lymphocytic angiitis, lupus (e.g. lupus erythematosus, lupus nephritis), Hashimoto's thyroiditis, primary myxedema, Graves' disease, pernicious anemia, autoimmune atrophic gastritis, Addison's disease, diabetes (e.g.
  • insulin dependent diabetes mellitis type I diabetes mellitis, type II diabetes mellitis
  • good pasture's syndrome myasthenia gravis, pemphigus, Crohn's disease, sympathetic ophthalmia, autoimmune uveitis, multiple sclerosis, autoimmune hemolytic anemia, idiopathic thrombocytopenia, primary biliary cirrhosis, chronic action hepatitis, ulceratis colitis, Sjogren's syndrome, rheumatic diseases (e.g. rheumatoid arthritis), polymyositis, scleroderma, and mixed connective tissue disease.
  • Gene therapy is a process to treat a disease by genetic manipulation. Gene therapy involves introducing a nucleic acid molecule into a cell and the cell expressing a gene product encoded by the nucleic acid molecule.
  • introducing the nucleic acid molecule into a cell may be performed by introducing an expression vector containing the nucleic acid molecule of interest into cells ex vivo or in vitro by a variety of methods including, for example, calcium phosphate precipitation, diethyaminoethyl dextran, polyethylene glycol (PEG), electroporation, direct injection, lipofection or viral infection (Sambrook et al., Molecular Cloning: A Laboratory Manual (Cold Spring Harbor Laboratory Press 1989); Kriegler M. Gene Transfer ad Expression: A Laboratory Manual (W. H.
  • nucleotide sequences of interest may be introduced into a cell in vivo using a variety of vectors and by a variety of methods including, for example: direct administration of the nucleic acid into a subject (Williams et al, 1991 PNAS 88:2726-2730); or insertion of the nucleic acid molecule into a viral vector, production of the recombinant virus or viral particle, and infection of the subject with the recombinant virus (Battleman et al, 1993 J Neurosci 13:94-951; Carroll et al, 1993 J Cell Biochem 17E:241; Lebkowski et al, U.S.
  • HAQs Health Questionnaire Assessments
  • ACR refers to clinical response studies based on criteria established by the American College of Rheumatology. A subject satisfied the “ACR20” criterion if there was about a 20 percent improvement in tender and swollen joint counts and 20 percent improvement in three of five remaining symptoms measured, such as patient and physician global disease changes, pain, physical disability, and an acute phase reactant such as CRP or ESR (Felson, D. T., et al., 1993 Arthritis and Rheumatism 36:729-740; Felson, D. T., et al., 1995 Arthritis and Rheumatism 38:1-9).
  • a subject satisfied the “ACR50” or “ACR70” criterion if there was about a 50 or 70 percent improvement, respectively, in tender and swollen joint counts and about 50 or 70 percent improvement, respectively, in three of five remaining symptoms measured, such as patient and physician global disease changes, pain, physical disability, and an acute phase reactant such as CRP or ESR.
  • the “Medical Outcomes Study Short Form-36 (SF-36)” refers to forms used to evaluate the impact of a DMARD (e.g., methotrexate or etanercept) and CTLA4Ig therapy on health-related quality of life (HRQOL).
  • the SF-36 consists of 36 items which covers four physical and four mental domains (physical function, role-physical, bodily pain, general health, vitality, social function, role emotional, and mental health). These individual domains are used to derive the physical and mental component summary scores which range from about 0 to 100, with higher scores indicating better quality of life. Absolute differences of 5 or more in the SF-36 scores were considered clinically meaningful.
  • an immune disease e.g., rheumatic disease
  • an immune disease including, but not limited to, joint swelling, pain, tenderness, morning stiffness, structural damage, an elevated level of serum C-reactive protein (CRP), an elevated level of soluble IL-2r, an elevated level of soluble ICAM-1, an elevated level of soluble E-selectin, an elevated level of rheumatoid factor, an elevated level of IL-6 or an elevated erythrocyte sedimentation rate.
  • CRP serum C-reactive protein
  • ICAM-1 an elevated level of soluble IL-2r
  • an elevated level of soluble E-selectin an elevated level of rheumatoid factor
  • an elevated level of IL-6 or an elevated erythrocyte sedimentation rate e.g., rheumatic disease
  • the present invention provides compositions and methods for treating immune system diseases, such as rheumatic diseases, by administering to a subject a combination of an effective amount of 1) a DMARD (such as methotrexate or a molecule that blocks TNF interactions, e.g., blocks TNF interactions with its ligand) or other therapeutic agent, plus 2) an effective amount of a molecule that blocks B7 interaction with CTLA4 and/or CD28 such as soluble CTLA4 molecules (e.g., CTLA4Ig, CTLA4Ig/CD28Ig, CTLA4-E7, CTLA4-p97, CTLA4-env gp120, L104EA29YIg, L104EA29LIg, L104EA29TIg and/or L104EA29WIg), soluble CD28 molecules, soluble B7-1 molecules, soluble B7-2 molecules, and monoclonal antibodies that recognize and bind B7, CD28 and/or CTLA4 (e.g., an anti-associated a
  • An effective amount of a molecule that blocks B7 interaction with CTLA4 and/or CD28 may be defined as the amount of anti-B7 monoclonal antibodies, soluble CTLA4 and/or soluble CD28 molecules that, when bound to B7 molecules on B7-positive cells, inhibit B7 molecules from binding endogenous ligands such as CTLA4 and CD28. The inhibition may be partial or complete.
  • an effective amount of a molecule that blocks B7 interaction with CTLA4 and/or CD28 may be defined as the amount of anti-CTLA4 monoclonal antibody, anti-CD28 monoclonal antibody or soluble B7 (B7-1 or B7-2) molecules that, when bound to CTLA4 and/or CD28 molecules on T cells, inhibit B7 molecules from binding endogenous ligands such as CTLA4 and CD28.
  • the inhibition may be partial or complete.
  • An effective amount of a molecule that blocks B7 interaction with CTLA4 and/or CD28 is an amount about 0.1 to 100 mg/kg weight of a subject.
  • the effective amount is an amount about 0.5 to 5 mg/kg weight of a subject, 0.1 to 5 mg/kg weight of a subject, about 5 to 10 mg/kg weight of a subject, about 10 to 15 mg/kg weight of a subject, about 15 to 20 mg/kg weight of a subject, about 20 to 25 mg/kg weight of a subject, about 25 to 30 mg/kg weight of a subject, about 30 to 35 mg/kg weight of a subject, about 35 to 40 mg/kg weight of a subject, about 40 to 45 mg/kg of a subject, about 45 to 50 mg/kg weight of a subject, about 50 to 55 mg/kg weight of a subject, about 55 to 60 mg/kg weight of a subject, about 60 to 65 mg/kg weight of a subject, about 65 to 70 mg/kg weight of a subject, about 70 to 75 mg/kg
  • the effective amount of a molecule that blocks B7 interaction with CTLA4 and/or CD28 is an amount about 2 mg/kg to about 10 mg/kg weight of a subject.
  • the preferred amount is 10 mg/kg weight of a subject.
  • the effective amount is an amount about 0.1 to 4 mg/kg weight of a subject.
  • the effective amount is an amount about 0.1 to 0.5 mg/kg weight of a subject, about 0.5 to 1.0 mg/kg weight of a subject, about 1.0 to 1.5 mg/kg weight of a subject, about 1.5 to 2.0 mg/kg weight of a subject, about 2.0 to 2.5 mg/kg weight of a subject, about 2.5 to 3.0 mg/kg weight of a subject, about 3.0 to 3.5 mg/kg weight of a subject, about 3.5 to 4.0 mg/kg weight of a subject, about 4.0 to 4.5 mg/kg weight of a subject, about 4.5 to 5.0 mg/kg weight of a subject, about 5.0 to 5.5 mg/kg weight of a subject, about 5.5 to 6.0 mg/kg weight of a subject, about 6.0 to 6.5 mg/kg weight of a subject, about 6.5 to 7.0 mg/kg weight of a subject, about 7.0 to 7.5 mg/kg weight of a subject, about 7.5 to 8.0 mg/kg weight of a subject, about 8.0 to 8.5 mg/kg weight of
  • the effective amount is an amount about 0.1 to 20 mg/kg weight of a subject. In another embodiment, the effective amount is an amount about 0.1 to 2 mg/kg weight of a subject, about 2 to 4 mg/kg weight of a subject, about 4 to 6 mg/kg weight of a subject, about 6 to 8 mg/kg weight of a subject, about 8 to 10 mg/kg weight of a subject, about 10 to 12 mg/kg weight of a subject, about 12 to 14 mg/kg weight of a subject, about 14 to 16 mg/kg weight of a subject, about 16 to 18 mg/kg weight of a subject or about 18 to 20 mg/kg weight of a subject.
  • the effective amount is about 2 mg/kg weight of a subject. In yet another embodiment, the effective amount is about 10 mg/kg weight of a subject.
  • the molecule that blocks B7 interaction with CTLA4 and/or CD28 is soluble CTLA4 and the effective amount of a soluble CTLA4 molecule is about 2 mg/kg weight of a subject. In another specific embodiment, the effective amount of a soluble CTLA4 molecule is about 10 mg/kg weight of a subject. In another specific embodiment, an effective amount of a soluble CTLA4 is 500 mg for a subject weighing less than 60 kg, 750 mg for a subject weighing between 60-100 kg and 1000 mg for a subject weighing more than 100 kg.
  • An effective amount of the molecule that blocks B7 interaction with CTLA4 and/or CD28 is soluble CTLA4 may be administered to a subject daily, weekly, monthly and/or yearly, in single or multiple times per hour/day/week/month/year, depending on need.
  • the molecule may initially be administered once every two weeks for a month, and then once every month thereafter.
  • the immune disease is a rheumatic disease.
  • Rheumatic diseases are any diseases which are characterized by (i) inflammation or degeneration of musculo-skeletal or connective tissue structures of the body, particularly the joints, and including muscles, tendons, cartilage, synovial and fibrous tissues, (ii) accompanied by joint swelling, joint tenderness, inflammation, morning stiffness, and/or pain, or impairment of locomotion or function of those structures and, in some cases, (iii) often accompanied by serological evidence of rheumatoid factor and other inflammatory surrogate markers.
  • Rheumatic diseases include, but are not limited to, rheumatoid arthritis.
  • the symptoms of rheumatoid arthritis include joint swelling, joint tenderness, inflammation, morning stiffness, and pain leading to physical disability.
  • Subjects afflicted with the advanced stages of arthritis suffer from symptoms of structural damage and debilitating pain.
  • Other organs also can be impaired by the autoimmune mechanism.
  • the DMARD is methotrexate or a molecule that blocks TNF interactions such as etanercept
  • the molecule that blocks B7 interaction with CTLA4 and/or CD28 is a soluble CTLA4.
  • the methods of the invention comprise administering to a subject an effective amount of methotrexate or a molecule that blocks TNF interactions in combination with an effective amount of soluble CTLA4 in order to treat rheumatic diseases such as rheumatoid arthritis.
  • Effective amounts of methotrexate range about 0.1 to 40 mg/week.
  • the effective amount includes ranges of about 0.1 to 5 mg/week, about 5 to 10 mg/week, about 10 to 15 mg/week, about 15 to 20 mg/week, about 20 to 25 mg/week, about 25 to 30 mg/week, about 30 to 35 mg/week, or about 35 to 40 mg/week.
  • methotrexate is administered in an amount ranging about 5 to 30 mg/week.
  • the effective amount of a soluble CTLA4 molecule is about 2 mg/kg weight subject and the effective amount of methotrexate is about 10 to 30 mg/week. In another embodiment, the effective amount of a soluble CTLA4 molecule is about 10 mg/kg weight subject and the effective amount of methotrexate is about 10 to 30 mg/week.
  • the DMARD is etanercept and the molecule that blocks B7 interaction with CTLA4 and/or CD28 is a soluble CTLA4.
  • the methods of the invention comprise administering to a subject an effective amount of etanercept in combination with an effective amount of soluble CTLA4 in order to treat rheumatic diseases such as rheumatoid arthritis.
  • Effective amounts of etanercept range about 0.1 to 100 mg/week.
  • the effective amount includes ranges of about 10 to 100 mg/week, about 0.1 to 50 mg/week, about 0.1 to 5 mg/week, about 5 to 10 mg/week, about 10 to 15 mg/week, about 15 to 20 mg/week, about 20 to 25 mg/week, about 25 to 30 mg/week, about 30 to 35 mg/week, about 35 to 40 mg/week, about 40 to 45 mg/week, about 45 to 50 mg/week, about 50 to 55 mg/week, about 55 to 60 mg/week, about 60 to 65 mg/week, about 65 to 70 mg/week, about 70 to 75 mg/week, about 75 to 80 mg/week, about 80 to 85 mg/week, about 85 to 90 mg/week, about 90 to 95 mg/week or about 95 to 100 mg/week.
  • etanercept is administered in an amount of about 50 mg/week, alternatively etanercept may be administered in an amount of about 25 mg twice weekly.
  • the effective amount of a soluble CTLA4 molecule is about 2 mg/kg weight subject and the effective amount of etanercept is about 25 mg twice a week. In another embodiment, the effective amount of a soluble CTLA4 molecule is about 10 mg/kg weight subject and the effective amount of etanercept is about 25 mg twice a week.
  • the invention also provides compositions and methods for treating immune system diseases, such as rheumatic diseases, by administering to a subject a combination of an effective amount of an NSAID and/or other therapeutic agent plus an effective amount of a molecule that blocks B7 interaction with CTLA4 and/or CD28.
  • immune system diseases such as rheumatic diseases
  • the invention also provides compositions and methods for treating immune system diseases, such as rheumatic diseases, by administering to a subject a an effective amount of a glucocorticoid, corticosteroid and/or other therapeutic agent plus an effective amount of a molecule that blocks B7 interaction with CTLA4 and/or CD28.
  • immune system diseases such as rheumatic diseases
  • the present invention provides compositions for treating immune diseases, such as rheumatic diseases, comprising soluble CTLA4 molecules. Further, the present invention provides compositions comprising a biological agent that inhibits T-cell function but not T-cell depletion in a human by contacting B7-positive cells in the human with a soluble CTLA4.
  • soluble CTLA4 include CTLA4Ig and soluble CTLA4 mutant molecules such as L104EA29YIg (FIG. 19), L104EA29LIg (FIG. 20), L104EA29TIg (FIG. 21), and L104EA29WIg (FIG. 22).
  • CTLA4 molecules with mutant or wildtype sequences, may be rendered soluble by deleting the CTLA4 transmembrane segment (Oaks, M. K., et al., 2000 Cellular Immunology 201:144-153).
  • soluble CTLA4 molecules may be fusion proteins, wherein the CTLA4 molecules are fused to non-CTLA4 moieties such as immunoglobulin (Ig) molecules that render the CTLA4 molecules soluble.
  • a CTLA4 fusion protein may include the extracellular domain of CTLA4 fused to an immunoglobulin constant domain, resulting in the CTLA4Ig molecule (FIG. 24) (Linsley, P. S., et al., 1994 Immunity 1:793-80).
  • immunoglobulin domains that may be fused to CTLA4 include, but are not limited to IgC ⁇ 1 (IgCgamma1), IgC ⁇ 2 (IgCgamma2), IgC ⁇ 3 (IgCgamma3), IgC ⁇ 4 (IgCgamma4), IgC ⁇ (IgCmu), IgC ⁇ 1 (IgCalpha1), IgC ⁇ 2 (IgCalpha2), IgC ⁇ (IgCdelta) or IgC ⁇ (IgCepsilon).
  • the immunoglobulin moiety does not elicit a detrimental immune response in a subject.
  • the preferred moiety is the immunoglobulin constant region, including the human or monkey immunoglobulin constant regions.
  • One example of a suitable immunoglobulin region is human C ⁇ 1, including the hinge, CH2 and CH3 regions which can mediate effector functions such as binding to Fc receptors, mediating complement-dependent cytotoxicity (CDC), or mediate antibody-dependent cell-mediated cytotoxicity (ADCC).
  • the immunoglobulin moiety may have one or more mutations therein, (e.g., in the CH2 domain, to reduce effector functions such as CDC or ADCC) where the mutation modulates the binding capability of the immunoglobulin to its ligand, by increasing or decreasing the binding capability of the immunoglobulin to Fc receptors.
  • mutations in the immunoglobulin moiety may include changes in any or all its cysteine residues within the hinge domain, for example, the cysteines at positions +130, +136, and +139 are substituted with serine (FIG. 24).
  • the immunoglobulin moiety may also include the proline at position +148 substituted with a serine, as shown in FIG. 24.
  • the mutations in the immunoglobulin moiety may include having the leucine at position +144 substituted with phenylalanine, leucine at position +145 substituted with glutamic acid, or glycine at position +147 substituted with alanine.
  • Additional non-CTLA4 moieties for use in the soluble CTLA4 molecules or soluble CTLA4 mutant molecules include, but are not limited to, p97 molecule, env gp120 molecule, E7 molecule, and ova molecule (Dash, B. et al. 1994 J. Gen. Virol. 75 (Pt 6):1389-97; Ikeda, T., et al. 1994 Gene 138(1-2):193-6; Falk, K., et al. 1993 Cell. Immunol. 150(2):447-52; Fujisaka, K. et al. 1994 Virology 204(2):789-93).
  • Other molecules are also possible (Gerard, C. et al. 1994 Neuroscience 62(3):721; Byrn, R. et al. 1989 63(10):4370; Smith, D. et al. 1987 Science 238:1704; Lasky, L. 1996 Science 233:209).
  • the soluble CTLA4 molecule of the invention can include a signal peptide sequence linked to the N-terminal end of the extracellular domain of the CTLA4 portion of the molecule.
  • the signal peptide can be any sequence that will permit secretion of the molecule, including the signal peptide from oncostatin M (Malik, et al., (1989) Molec. Cell. Biol. 9: 2847-2853), or CD5 (Jones, N. H. et al., (1986) Nature 323:346-349), or the signal peptide from any extracellular protein.
  • the soluble CTLA4 molecule of the invention can include the oncostatin M signal peptide linked at the N-terminal end of the extracellular domain of CTLA4, and the human immunoglobulin molecule (e.g., hinge, CH2 and CH3) linked to the C-terminal end of the extracellular domain (wildtype or mutated) of CTLA4.
  • This molecule includes the oncostatin M signal peptide encompassing an amino acid sequence having methionine at position ⁇ 26 through alanine at position ⁇ 1, the CTLA4 portion encompassing an amino acid sequence having methionine at position +1 through aspartic acid at position +124, a junction amino acid residue glutamine at position +125, and the immunoglobulin portion encompassing an amino acid sequence having glutamic acid at position +126 through lysine at position +357.
  • the soluble CTLA4 mutant molecules of the invention comprising the mutated CTLA4 sequences described infra, can be fusion molecules comprising human Ig, e.g., IgC(gamma)1 (i.e. IgC ⁇ 1) moieties fused to the mutated CTLA4 fragments.
  • IgC(gamma)1 i.e. IgC ⁇ 1
  • the soluble CTLA4 mutant molecules comprise IgC ⁇ 1 (IgCgamma1) fused to an extracellular domain of CTLA4 comprising a single-site mutation in the extracellular domain.
  • the extracellular domain of CTLA4 comprises methionine at position +1 through aspartic acid at position +124 (e.g., FIG. 23).
  • the extracellular domain of the CTLA4 can comprise alanine at position ⁇ 1 through aspartic acid at position +124 (e.g., FIG. 23).
  • single-site mutations include the following wherein the leucine at position +104 is changed to any other amino acid.
  • Codon change L104EIg Glutamic acid GAG L104SIg Serine AGT L104TIg Threonine ACG L104AIg Alanine GCG L104WIg Tryptophan TGG L104QIg Glutamine CAG L104KIg Lysine AAG L104RIg Arginine CGG L104GIg Glycine GGG
  • mutant molecules having the extracellular domain of CTLA4 with two mutations, fused to an Ig C ⁇ 1 (IgCgamma1) moiety include the following wherein the leucine at position +104 is changed to another amino acid (e.g.
  • Double-site mutants Codon change: L104EG105FIg Phenylalanine TTC L104EG105WIg Tryptophan TGG L104EG105LIg Leucine CTT L104ES25RIg Arginine CGG L104ET30GIg Glycine GGG L104ET30NIg Asparagine AAT L104EA29YIg Tyrosine TAT L104EA29LIg Leucine TTG L104EA29TIg Threonine ACT L104EA29WIg Tryptophan TGG
  • mutant molecules having the extracellular domain of CTLA4 comprising three mutations, fused to an IgC ⁇ 1 (IgCgamma1) moiety.
  • IgCgamma1 IgCgamma1
  • Examples include the following wherein the leucine at position +104 is changed to another amino acid (e.g. glutamic acid), the alanine at position +29 is changed to another amino acid (e.g. tyrosine) and the serine at position +25 is changed to another amino acid:
  • Triple-site Mutants Codon changes: L104EA29YS25KIg Lysine AAA L104EA29YS25KIg Lysine AAG L104EA29YS25NIg Asparagine AAC L104EA29YS25RIg Arginine CGG
  • Soluble CTLA4 mutant molecules may have a junction amino acid residue which is located between the CTLA4 portion and the Ig portion of the molecule.
  • the junction amino acid can be any amino acid, including glutamine.
  • the junction amino acid can be introduced by molecular or chemical synthesis methods known in the art.
  • the soluble CTLA4 proteins of the invention can be generated by chemical synthesis methods.
  • the principles of solid phase chemical synthesis of polypeptides are well known in the art and may be found in general texts relating to this area (Dugas, H. and Penney, C. 1981 Bioorganic Chemistry, pp 54-92, Springer-Verlag, New York).
  • the soluble CTLA4 proteins may be synthesized by solid-phase methodology utilizing an Applied Biosystems 430A peptide synthesizer (Applied Biosystems, Foster City, Calif.) and synthesis cycles supplied by Applied Biosystems.
  • Protected amino acids such as t-butoxycarbonyl-protected amino acids, and other reagents are commercially available from many chemical supply houses.
  • the present invention provides CTLA4 mutant molecules including a signal peptide sequence linked to the N-terminal end of the extracellular domain of the CTLA4 portion of the mutant molecule.
  • the signal peptide can be any sequence that will permit secretion of the mutant molecule, including the signal peptide from oncostatin M (Malik, et al., 1989 Molec. Cell. Biol. 9: 2847-2853), or CD5 (Jones, N. H. et al., 1986 Nature 323:346-349), or the signal peptide from any extracellular protein.
  • the invention provides soluble CTLA4 mutant molecules comprising a single-site mutation in the extracellular domain of CTLA4 such as L104EIg (as included in FIG. 18) or L104SIg, wherein L104EIg and L104SIg are mutated in their CTLA4 sequences so that leucine at position +104 is substituted with glutamic acid or serine, respectively.
  • the single-site mutant molecules further include CTLA4 portions encompassing methionine at position +1 through aspartic acid at position +124, a junction amino acid residue glutamine at position +125, and an immunoglobulin portion encompassing glutamic acid at position +126 through lysine at position +357.
  • the immunoglobulin portion of the mutant molecule may also be mutated so that the cysteines at positions +130, +136, and +139 are substituted with serine, and the proline at position +148 is substituted with serine.
  • the single-site soluble CTLA4 mutant molecule may have a CTLA4 portion encompassing alanine at position ⁇ 1 through aspartic acid at position +124.
  • the invention provides soluble CTLA4 mutant molecules comprising a double-site mutation in the extracellular domain of CTLA4, such as L104EA29YIg, L104EA29LIg, L104EA29TIg or L104EA29WIg, wherein leucine at position +104 is substituted with a glutamic acid and alanine at position +29 is changed to tyrosine, leucine, threonine and tryptophan, respectively.
  • soluble CTLA4 mutant molecules comprising a double-site mutation in the extracellular domain of CTLA4, such as L104EA29YIg, L104EA29LIg, L104EA29TIg or L104EA29WIg, wherein leucine at position +104 is substituted with a glutamic acid and alanine at position +29 is changed to tyrosine, leucine, threonine and tryptophan, respectively.
  • the sequences for L104EA29YIg, L104EA29LIg, L104EA29TIg and L104EA29WIg, starting at methionine at position +1 and ending with lysine at position +357, plus a signal (leader) peptide sequence are shown in FIGS. 19 - 22 respectively.
  • the double-site mutant molecules further comprise CTLA4 portions encompassing methionine at position +1 through aspartic acid at position +124, a junction amino acid residue glutamine at position +125, and an immunoglobulin portion encompassing glutamic acid at position +126 through lysine at position +357.
  • the immunoglobulin portion of the mutant molecule may also be mutated, so that the cysteines at positions +130, +136, and +139 are substituted with serine, and the proline at position +148 is substituted with serine.
  • these mutant molecules can have a CTLA4 portion encompassing alanine at position ⁇ 1 through aspartic acid at position +124.
  • the immunoglobulin portion of the may also be mutated, so that the cysteines at positions +130, +136, and +139 are substituted with serine, and the proline at position +148 is substituted with serine.
  • these mutant molecules can have a CTLA4 portion encompassing alanine at position ⁇ 1 through aspartic acid at position +124.
  • L104ES25RIg which is a double-site mutant molecule comprising a CTLA4 portion encompassing methionine at position +1 through aspartic acid at position +124, a junction amino acid residue glutamine at position +125, and the immunoglobulin portion encompassing glutamic acid at position +126 through lysine at position +357.
  • the portion having the extracellular domain of CTLA4 is mutated so that serine at position +25 is substituted with arginine, and leucine at position +104 is substituted with glutamic acid.
  • L104ES25RIg can have a CTLA4 portion encompassing alanine at position ⁇ 1 through aspartic acid at position +124.
  • the immunoglobulin portion of the mutant molecule may also be mutated, so that the cysteines at positions +130, +136, and +139 are substituted with serine, and the proline at position +148 is substituted with serine.
  • these mutant molecules can have a CTLA4 portion encompassing alanine at position ⁇ 1 through aspartic acid at position +124.
  • the invention provides soluble CTLA4 mutant molecules comprising a triple-site mutation in the extracellular domain of CTLA4, such as L104EA29YS25KIg, L104EA29YS25NIg, L104EA29YS25RIg, wherein leucine at position +104 is substituted with a glutamic acid, alanine at position +29 is substituted with tyrosine, and serine at position +25 is substituted with lysine, asparagine and arginine, respectively.
  • a triple-site mutation in the extracellular domain of CTLA4 such as L104EA29YS25KIg, L104EA29YS25NIg, L104EA29YS25RIg, wherein leucine at position +104 is substituted with a glutamic acid, alanine at position +29 is substituted with tyrosine, and serine at position +25 is substituted with lysine, asparagine and arginine, respectively.
  • the triple-site mutant molecules further comprise CTLA4 portions encompassing methionine at position +1 through aspartic acid at position +124, a junction amino acid residue glutamine at position +125, and an immunoglobulin portion encompassing glutamic acid at position +126 through lysine at position +357.
  • the immunoglobulin portion of the mutant molecule may also be mutated, so that the cysteines at positions +130, +136, and +139 are substituted with serine, and the proline at position +148 is substituted with serine.
  • these mutant molecules can have a CTLA4 portion encompassing alanine at position ⁇ 1 through aspartic acid at position +124.
  • soluble CTLA4 mutant molecules include chimeric CTLA4/CD28 homologue mutant molecules that bind a B7 (Peach, R. J., et al., 1994 J Exp Med 180:2049-2058).
  • chimeric CTLA4/CD28 mutant molecules include HS1, HS2, HS3, HS4, HS5, HS6, HS4A, HS4B, HS7, HS8, HS9, HS10, HS11, HS12, HS13 and HS14 (U.S. Pat. No. 5,773,253)
  • Preferred embodiments of the invention are soluble CTLA4 molecules such as CTLA4Ig (as shown in FIG.
  • the invention further provides nucleic acid molecules comprising nucleotide sequences encoding the amino acid sequences corresponding to the soluble CTLA4 molecules of the invention.
  • the nucleic acid molecule is a DNA (e.g., cDNA) or a hybrid thereof.
  • a CTLA4Ig molecule can comprise a GCT or GCC codon, encoding alanine, at nucleotide position +49 to +51 as shown in FIG. 24.
  • a CTLA4Ig molecule can comprise a GGT or GGG codon, encoding glycine, at nucleotide position +436 to +438 as shown in FIG. 24.
  • a CTLA4Ig molecule can comprise a CGG or CGT codon, encoding arginine, at nucleotide position +631 to +633 as shown in FIG. 24.
  • DNA encoding CTLA4Ig (FIG. 24) was deposited on May 31, 1991 with the American Type Culture Collection (ATCC), 10801 University Boulevard., Manassas, Va. 20110-2209 and has been accorded ATCC accession number ATCC 68629.
  • DNA encoding L104EA29YIg (sequence included in FIG. 19) was deposited on Jun. 19, 2000 with ATCC and has been accorded ATCC accession number PTA-2104.
  • the nucleic acid molecules are RNA or a hybrid thereof.
  • the nucleic acid molecules of the invention also include derivative nucleic acid molecules which differ from DNA or RNA molecules, and anti-sense molecules.
  • Derivative molecules include peptide nucleic acids (PNAs), and non-nucleic acid molecules including phosphorothioate, phosphotriester, phosphoramidate, and methylphosphonate molecules, that bind to single-stranded DNA or RNA in a base pair-dependent manner (Zamecnik, P. C., et al., 1978 Proc. Natl. Acad. Sci. 75:280284; Goodchild, P. C., et al., 1986 Proc. Natl. Acad. Sci. 83:4143-4146).
  • Peptide nucleic acid molecules comprise a nucleic acid oligomer to which an amino acid residue, such as lysine, and an amino group have been added. These small molecules, also designated anti-gene agents, stop transcript elongation by binding to their complementary (template) strand of nucleic acid (Nielsen, P. E., et al., 1993 Anticancer Drug Des 8:53-63). Reviews of methods for synthesis of DNA, RNA, and their analogues can be found in: Oligonucleotides and Analogues, eds. F. Eckstein, 1991, IRL Press, New York; Oligonucleotide Synthesis, ed. M. J. Gait, 1984, IRL Press, Oxford, England.
  • the vectors can permit expression of the soluble CTLA4 transcript or polypeptide sequences in prokaryotic or eukaryotic host cells.
  • the vectors include expression vectors, comprising an expression control element, such as a promoter sequence, which enables transcription of the inserted soluble CTLA4 nucleic acid sequences and can be used for regulating the expression (e.g., transcription and/or translation) of an operably linked soluble CTLA4 sequence in an appropriate host cell.
  • Expression control elements are known in the art and include, but are not limited to, inducible promoters, constitutive promoters, secretion signals, enhancers, transcription terminators, and other transcriptional regulatory elements. Other expression control elements that are involved in translation are known in the art, and include the Shine-Dalgarno sequence (e.g., prokaryotic host cells), and initiation and termination codons.
  • Specific initiation signals may also be required for efficient translation of a soluble CTLA4 sequence. These signals include the ATG-initiation codon and adjacent sequences. In cases where the soluble CTLA4 initiation codon and upstream sequences are inserted into the appropriate expression vector, no additional translational control signals may be needed. However, in cases where only the coding sequence, or a portion thereof, is inserted, exogenous transcriptional control signals including the ATG-initiation codon may be provided. Furthermore, the initiation codon should be in the correct reading-frame to ensure translation of the entire insert. Exogenous transcriptional elements and initiation codons can be of various origins, both natural and synthetic.
  • the preferred vectors for expression of the soluble CTLA4 sequences in eukaryote host cells include expression control elements, such as the baculovirus polyhedrin promoter for expression in insect cells.
  • Other expression control elements include promoters or enhancers derived from the genomes of plant cells (e.g., heat shock, RUBISCO, storage protein genes), viral promoters or leader sequences or from plant viruses, and promoters or enhancers from the mammalian genes or from mammalian viruses.
  • the preferred vectors for generating soluble CTLA4 transcripts and/or the encoded soluble CTLA4 polypeptides are expression vectors which are compatible with prokaryotic host cells.
  • Prokaryotic cell expression vectors are well known in the art and are available from several commercial sources.
  • pET vectors e.g., pET-21, Novagen Corp.
  • BLUESCRIPT phagemid (Stratagene, LaJolla, Calif.)
  • pSPORT Gibco BRL, Rockville, Md.
  • ptrp-lac hybrids may be used to express soluble CTLA4 polypeptides in bacterial host cells.
  • the preferred expression vectors for generating soluble CTLA4 transcripts and/or the encoded soluble CTLA4 polypeptides are expression vectors which are compatible with eukaryotic host cells.
  • the more preferred vectors are those compatible with vertebrate cells.
  • Eukaryotic cell expression vectors are well known in the art and are available from several commercial sources. Typically, such vectors are provided containing convenient restriction sites for insertion of the desired DNA segment. Typical of such vectors are PSVL and pKSV-10 (Pharmacia), pBPV-1/pML2d (International Biotechnologies, Inc.), pTDT1 (ATCC, #31255), and similar eukaryotic expression vectors.
  • a host vector system comprises the vector of the invention in a suitable host cell.
  • suitable host cells include, but are not limited to, prokaryotic and eukaryotic cells.
  • eukaryotic cells are also suitable host cells.
  • eukaryotic cells include any animal cell, whether primary or immortalized, yeast (e.g., Saccharomyces cerevisiae, Schizosaccharomyces pombe , and Pichia pastoris ), and plant cells.
  • Exemplary animal cells include cells from bovine, ovine, porcine, murine, equine, monkey and ape.
  • CHO-K1 Tet-On cell line CCL-61
  • CHO designated ECACC 85050302 (CAMR, Salisbury, Wiltshire, UK)
  • CHO clone 13 (GEIMG, Genova, IT)
  • CHO clone B (GEIMG, Genova, IT)
  • CHO-K1/SF designated ECACC 93061607 (CAMR, Salisbury, Wiltshire, UK)
  • RR-CHOK1 designated ECACC 92052129 (CAMR, Salisbury, Wiltshire, UK).
  • Exemplary plant cells include whole plants, cell culture, or callus, from tobacco, corn, soybean, and rice cells. Corn, soybean, and rice seeds are also acceptable.
  • the CTLA4 mutant molecules of the invention may be isolated as naturally-occurring polypeptides, or from any source whether natural, synthetic, semi-synthetic or recombinant. Accordingly, the CTLA4 mutant polypeptide molecules may be isolated as naturally-occurring proteins from any species, particularly mammalian, including bovine, ovine, porcine, murine, equine, and preferably human. Alternatively, the CTLA4 mutant polypeptide molecules may be isolated as recombinant polypeptides that are expressed in prokaryote or eukaryote host cells, or isolated as a chemically synthesized polypeptide.
  • CTLA4 mutant molecules and fragments or derivatives thereof can be produced by recombinant methods. Accordingly, an isolated nucleotide sequence encoding wild-type CTLA4 molecules may be manipulated to introduce mutations, resulting in nucleotide sequences that encode the CTLA4 mutant polypeptide molecules.
  • the nucleotide sequences encoding the CTLA4 mutant molecules may be generated by site-directed mutagenesis methods, using primers and PCR amplification.
  • the primers can include specific sequences designed to introduce desired mutations. Alternatively, the primers can be designed to include randomized or semi-randomized sequences to introduce random mutations.
  • the invention includes pharmaceutical compositions comprising pharmaceutically effective amounts of a molecule that blocks B7 interaction with CTLA4 and/or CD28 such as soluble CTLA4 molecules, CD28 molecules, B7 (B7-1 or B7-2) molecules, anti-CTLA4 monoclonal antibodies, anti-CD28 monoclonal antibodies or anti-B7 (B7-1 or B7-2) monoclonal antibodies.
  • a molecule that blocks B7 interaction with CTLA4 and/or CD28 such as soluble CTLA4 molecules, CD28 molecules, B7 (B7-1 or B7-2) molecules, anti-CTLA4 monoclonal antibodies, anti-CD28 monoclonal antibodies or anti-B7 (B7-1 or B7-2) monoclonal antibodies.
  • the pharmaceutical compositions of the invention are useful for treatment of immune system diseases.
  • immune system diseases are mediated by CD28/CTLA4/B7 interactions.
  • the soluble CTLA4 molecules are preferably soluble CTLA4 molecules with wildtype sequence and/or soluble CTLA4 molecules having one
  • the pharmaceutical composition can include soluble CTLA4 protein molecules and/or nucleic acid molecules, and/or vectors encoding the molecules.
  • the soluble CTLA4 molecules have the amino acid sequence of the extracellular domain of CTLA4 as shown in either FIG. 24 or 19 (CTLA4Ig or L104EA29Y, respectively). Even more preferably, the soluble CTLA4 mutant molecule is L104EA29YIg as disclosed herein.
  • the compositions may additionally include other therapeutic agents, including, but not limited to, DMARDs, NSAIDs, corticosteroids, glucocorticoids, drug toxins, alkylating agents, anti-neoplastic drugs, enzymes, antibodies, or conjugates.
  • An embodiment of the pharmaceutical composition of the invention comprises an effective amount of a molecule that blocks B7 interaction with CTLA4 and/or CD28, such as the molecules and the suitable amounts of the molecules described supra, and an effective amount of a DMARD.
  • an effective amount can be in a range of about 1 to about 5000 mg/day. This range can be modified to an amount of about 1 to 10 mg/day, about 10 to 50 mg/day, about 50 to 100 mg/day, about 100 to 150 mg/day, about 150 to 200 mg/day, about 200 to 250 mg/day, about 250 to 300 mg/day, about 300 to 350 mg/day, about 350 to 400 mg/day, about 400 to 450 mg/day, about 450 to 500 mg/day, about 500 to 550 mg/day, about 550 to 600 mg/day, about 600 to 650 mg/day, about 650 to 700 mg/day, about 700 to 750 mg/day, about 750 to 800 mg/day, about 800 to 850 mg/day, about 850 to 900 mg/day, about 900 to 950 mg/day, about 950 to 1000 mg/day, about 1000 to 1100 mg/day, about 1100 to 1200 mg/day, about 1200 to 1300 mg/day, about 1300
  • an effective amount of a DMARD can be in a range of about 0.1 mg/week to 40 mg/week; 0.1 mg/week to 5 mg/week; 5 mg/week to 10 mg/week; 10 mg/week to 30 mg/week; 30 mg/week to 35 mg/week; 0.1 mg/week to 100 mg/week; or 30 mg/week to 50 mg/week.
  • a DMARD can be administered in an amount of about 50 mg/week or 25 mg twice weekly. It would be clear to one skilled in the art that dosage range will vary depending on the particular DMARD being used, for example see below.
  • Methotrexate is an antimetabolite molecule that interferes with DNA synthesis, repair and cellular replication. Methotrexate functions as an inhibitor of dihydrofolic acid reductase i.e. it is a folic acid antagonist. Methotrexate is commonly administered in an amount about 0.1 to 40 mg per week with a common dosage ranging about 5 to 30 mg per week. Methotrexate may be administered to a subject in various increments: about 0.1 to 5 mg/week, about 5 to 10 mg/week, about 10 to 15 mg/week, about 15 to 20 mg/week, about 20 to 25 mg/week, about 25 to 30 mg/week, about 30 to 35 mg/week, or about 35 to 40 mg/week. In one embodiment, an effective amount of a DMARD, including methotrexate, is an amount about 10 to 30 mg/week.
  • Cyclophosphamide an alkylating agent
  • Cyclosporine e.g. NEORAL R
  • Cyclosporin A is commonly administered in dosages ranging from about 1 to 10 mg/kg body weight per day. Dosages ranging about 2.5 to 4 mg per body weight per day are commonly used.
  • Chloroquine or hydroxychloroquine is commonly administered in dosages ranging about 100 to 1000 mg daily. Preferred dosages range about 200-600 mg administered daily.
  • Sulfasalazine e.g., AZULFIDINE EN-tabs R
  • AZULFIDINE EN-tabs R is commonly administered in amounts ranging about 50 to 5000 mg per day, with a common dosage of about 2000 to 3000 mg per day for adults. Dosages for children are commonly about 5 to 100 mg/kg of body weight, up to 2 grams per day.
  • Gold salts are formulated for two types of administration: injection or oral. Injectable gold salts are commonly prescribed in dosages about 5 to 100 mg doses every two to four weeks. Orally administered gold salts are commonly prescribed in doses ranging about 1 to 10 mg per day.
  • D-penicillamine or penicillamine (CUPRIMINE R ) is commonly administered in dosages about 50 to 2000 mg per day, with preferred dosages about 125 mg per day up to 1500 mg per day.
  • Azathioprine is commonly administered in dosages of about 10 to 250 mg per day. Preferred dosages range about 25 to 200 mg per day.
  • Anakinra e.g. KINERET R
  • KINERET R is an interleukin-1 receptor antagonist.
  • a common dosage range for anakinra is about 10 to 250 mg per day, with a recommended dosage of about 100 mg per day.
  • Infliximab (REMICADE R ) is a chimeric monoclonal antibody that binds to tumor necrosis factor alpha (TNF ⁇ ) and inhibits the activity of TNF ⁇ . Infliximab is commonly administered in dosages about 1 to 20 mg/kg body weight every four to eight weeks. Dosages of about 3 to 10 mg/kg body weight may be administered every four to eight weeks depending on the subject.
  • Etanercept is a dimeric fusion protein that binds the tumor necrosis factor (TNF) and blocks its interactions with TNF receptors.
  • TNF tumor necrosis factor
  • Commonly administered dosages of etanercept are about 10 to 100 mg per week for adults with a preferred dosage of about 50 mg per week. Dosages for juvenile subjects range about 0.1 to 50 mg/kg body weight per week with a maximum of about 50 mg per week. For adult patients, etanercept is commonly administered e.g., injected, in 25 mg doses twice weekly e.g., 72-96 hours apart in time.
  • Leflunomide (ARAVA R ) is commonly administered at dosages about 1 and 100 mg per day. A common daily dosage is about 10 to 20 mg per day.
  • a further embodiment of the invention is a pharmaceutical composition
  • a pharmaceutical composition comprising an effective amount of a soluble CTLA4, such as CTLA4Ig, and an effective amount of a DMARD, such as methotrexate or etanercept.
  • a pharmaceutical composition comprising soluble CTLA4 can be used for methods for blocking B7 interaction with CTLA4 and/or CD28; or for treating immune system diseases.
  • Effective amounts of soluble CTLA4 in the pharmaceutical composition range about 0.1 to 100 mg/kg weight of the subject.
  • the effective amount is an amount about 0.5 to 5 mg/kg weight of a subject, about 5 to 10 mg/kg weight of a subject, about 10 to 15 mg/kg weight of a subject, about 15 to 20 mg/kg weight of a subject, about 20 to 25 mg/kg weight of a subject, about 25 to 30 mg/kg weight of a subject, about 30 to 35 mg/kg weight of a subject, about 35 to 40 mg/kg weight of a subject, about 40 to 45 mg/kg of a subject, about 45 to 50 mg/kg weight of a subject, about 50 to 55 mg/kg weight of a subject, about 55 to 60 mg/kg weight of a subject, about 60 to 65 mg/kg weight of a subject, about 65 to 70 mg/kg weight of a subject, about 70 to 75 mg/kg weight of a subject, about 75 to 80 mg/kg weight of a subject, about 80 to 85 mg/kg weight of a subject, about 85 to 90 mg/kg weight of a subject, about 90 to 95 mg/kg weight of a subject, about
  • the effective amount of soluble CTLA4 is an amount about 2 mg/kg to about 10 mg/kg weight of a subject. In another embodiment, the effective amount is an amount about 0.1 to 4 mg/kg weight of a subject. In another embodiment the effective amount is an amount about 0.1 to 0.5 mg/kg weight of a subject, about 0.5 to 1.0 mg/kg weight of a subject, about 1.0 to 1.5 mg/kg weight of a subject, about 1.5 to 2.0 mg/kg weight of a subject, about 2.0 to 2.5 mg/kg weight of a subject, about 2.5 to 3.0 mg/kg weight of a subject, about 3.0 to 3.5 mg/kg weight of a subject or about 3.5 to 4.0 mg/kg weight of a subject.
  • the effective amount is an amount about 0.1 to 20 mg/kg weight of a subject. In another embodiment, the effective amount is an amount about 0.1 to 2 mg/kg weight of a subject, about 2 to 4 mg/kg weight of a subject, about 4 to 6 mg/kg weight of a subject, about 6 to 8 mg/kg weight of a subject, about 8 to 10 mg/kg weight of a subject, about 10 to 12 mg/kg weight of a subject, about 12 to 14 mg/kg weight of a subject, about 14 to 16 mg/kg weight of a subject, about 16 to 18 mg/kg weight of a subject or about 18 to 20 mg/kg weight of a subject. In an embodiment, the effective amount is 2 mg/kg weight of a subject. In another embodiment, the effective amount is about 10 mg/kg weight of a subject.
  • an effective amount of soluble CTLA4 is 500 mg for a subject weighing less than 60 kg, 750 mg for a subject weighing between 60-100 kg and 1000 mg for a subject weighing more than 100 kg.
  • Effective amounts of methotrexate in the pharmaceutical composition range about 0.1 to 40 mg/week.
  • the effective amount is an amount about 0.1 to 5 mg/week, about 5 to 10 mg/week, about 10 to 15 mg/week, about 15 to 20 mg/week, about 20 to 25 mg/week, about 25 to 30 mg/week, about 30 to 35 mg/week, or about 35 to 40 mg/week.
  • an effective amount of a DMARD, including methotrexate is an amount about 10 to 30 mg/week.
  • the effective amount of a soluble CTLA4 molecule is about 2 mg/kg weight subject and the effective amount of methotrexate is about 10 to 30 mg/week. In another embodiment, the effective amount of a soluble CTLA4 molecule is about 10 mg/kg weight subject and the effective amount of methotrexate is about 10 to 30 mg/week.
  • Effective amounts of etanercept in the pharmaceutical composition range about 0.1 to 100 mg/week. In one embodiment, the effective amount is ranges about 0.1 to 5 mg/week, about 5 to 10 mg/week, about 10 to 15 mg/week, about 15 to 20 mg/week, about 20 to 25 mg/week, about 25 to 30 mg/week, about 30 to 35 mg/week, about 35 to 40 mg/week, about 40 to 45 mg/week, about 45 to 50 mg/week, about 50 to 55 mg/week, about 55 to 60 mg/week, about 60 to 65 mg/week, about 65 to 70 mg/week, about 70 to 75 mg/week, about 75 to 80 mg/week, about 80 to 85 mg/week, about 85 to 90 mg/week, about 90 to 95 mg/week or about 95 to 100 mg/week. In one embodiment, etanercept is administered in an amount ranging about 50 mg/week e.g., 25 mg administered twice weekly.
  • the effective amount of a soluble CTLA4 molecule is about 2 mg/kg weight subject and the effective amount of etanercept is about 25 mg twice a week. In another embodiment, the effective amount of a soluble CTLA4 molecule is about 10 mg/kg weight subject and the effective amount of etanercept is about 25 mg twice a week.
  • compositions of the invention further encompass a pharmaceutical composition comprising soluble CTLA4 in combination with other treatments for rheumatic disease including, but not limited to: collagen, dnaJ, molecules that block TNF function (e.g., pegsunercept), molecules that block cytokine function (e.g., AMG719), molecules that block LFA-1 function (e.g., efalizumab) and stem cell transplants.
  • TNF function e.g., pegsunercept
  • cytokine function e.g., AMG719
  • LFA-1 function e.g., efalizumab
  • Collagen for example in the form of bovine II collagen, may be orally administered to a patient suffering from rheumatoid arthritis in order to alleviate one or more symptoms of rheumatoid arthritis.
  • DnaJ is a small peptide which mimics a protein contained in a gene in many patients with rheumatoid arthritis.
  • the peptide is derived from E. coli bacteria heat shock protein.
  • DnaJ may be orally administered to a patient suffering from rheumatoid arthritis in order to alleviate one or more symptoms of rheumatoid arthritis.
  • TNF is a molecule involved in the inflammatory response of patients with rheumatoid arthritis.
  • any molecule that blocks TNF function e.g., by blocking TNF binding to the TNF receptor (TNFR)
  • TNF receptor TNF receptor
  • TNF blockers such as infliximab and etanercept, have been shown to be efficacious in treating rheumatoid arthritis.
  • Other TNF blockers such as pegsunercept are being developed and tested (Phase II clinical trial) for their efficacy in treating rheumatoid arthritis.
  • Cytokines e.g., Interleukin-1 (IL-1), are cell secreted molecules involved in mediating immune responses. Conceivably, any molecule that blocks cytokine function e.g., by blocking IL-1 interaction with its receptor, may help modify the progression of rheumatoid arthritis and alleviate one or more of its symptoms.
  • IL-1 Interleukin-1
  • AMG719 An IL-1 inhibitor, AMG719, is being developed and tested (Phase II clinical trial) for its efficacy in treating rheumatoid arthritis.
  • Lymphocyte function associated molecule 1 is a molecule composed of two subunits, CD11a and CD18, which functions by mediating lymphocyte adhesion to various cell types such as endothelium. Conceivably, interference of LFA-1 function may help modify the progression of rheumatoid arthritis and alleviate one or more of its symptoms.
  • An anti-LFA-1 antibody, efalizumab is being developed and tested (Phase II clinical trial) for its efficacy in treating rheumatoid arthritis.
  • Blockage of TNF, cytokine or LFA-1 interaction to their ligands by a potentially therapeutic molecule can be determined by any number of assays known to those skilled in the art.
  • competition assays may be used to test blockage by the molecule of interest e.g., a molecule can be exposed to a TNF/TNFR binding pair in order to compete with TNF to bind to TNFR.
  • functional assays can be performed to test blockage e.g., a molecule can be tested for its ability to inhibit an inflammatory cascade, or any part of an inflammatory reaction such as swelling, redness or pain, caused by a cytokine.
  • the present invention also provides pharmaceutical compositions comprising the molecules of the invention e.g., CTLA4Ig and an acceptable carrier or adjuvant which is known to those of skill of the art.
  • the pharmaceutical compositions preferably include suitable carriers and adjuvants which include any material which when combined with the molecules of the invention (e.g., a soluble CTLA4 molecule, such as, CTLA4Ig or L104EA29Y) retain the molecule's activity, and is non-reactive with the subject's immune system.
  • carriers and adjuvants include, but are not limited to, ion exchangers, alumina, aluminum stearate, lecithin, serum proteins, such as human serum albumin, buffer substances such as phosphates, glycine, sorbic acid, potassium sorbate, partial glyceride mixtures of saturated vegetable fatty acids, phosphate buffered saline solution, water, emulsions (e.g. oil-water emulsion), salts or electrolytes such as protamine sulfate, disodium hydrogen phosphate, potassium hydrogen phosphate, sodium chloride, zinc salts, colloidal silica, magnesium trisilicate, polyvinyl pyrrolidone, cellulose-based substances and polyethylene glycol.
  • buffer substances such as phosphates, glycine, sorbic acid, potassium sorbate, partial glyceride mixtures of saturated vegetable fatty acids, phosphate buffered saline solution, water, emulsions (e.g
  • compositions comprising such carriers are formulated by well known conventional methods. Such compositions may also be formulated within various lipid compositions, such as, for example, liposomes as well as in various polymeric compositions, such as polymer microspheres.
  • kits i.e., a packaged combination of reagents with instructions
  • the molecules of the invention useful for blocking B7 interactions with its ligands and/or for treating an immune system disease.
  • the kit can contain a pharmaceutical composition that includes one or more agents, for example, a soluble CTLA4 molecule alone, or with a second agent, and an acceptable carrier or adjuvant, e.g., pharmaceutically acceptable buffer, such as phosphate-buffered saline, Ringer's solution and dextrose solution. It may further include other materials desirable from a commercial and user standpoint, including other buffers, diluents, filters, needles, syringes, and package inserts with instructions for use.
  • the agents may be provided as dry powders, usually lyophilized, including excipients that upon dissolving will provide a reagent solution having the appropriate concentration.
  • Second agents can include the following: steroids, glucocorticoids, drug toxins, alkylating agents, anti-neoplastic drugs, enzymes, antibodies, conjugates, immunosuppressive agents, corticosteroids, DMARDs, nonsteroidal antiinflammatory drugs (NSAIDs), prednisone, azathioprine, methotrexate, TNF ⁇ blockers or antagonists, infliximab, any biological agent targeting an inflammatory cytokine, chloroquine, hydroxychloroquine, sulfasalazine (sulphasalazopryine), gold salts, etanercept, anakinra, cyclophosphamide, leflunomide, collagen, dnaJ, a molecule that blocks TNF receptors (e.g., pegsunercept), a molecule that blocks cytokine function (e.g., AMG719), a molecule that blocks LFA-1 function (e.g.,
  • the kit comprises a container with a label and/or instructions.
  • Suitable containers include, for example, bottles, vials, and test tubes.
  • the containers can be formed from a variety of materials such as glass or plastic.
  • the container can have a sterile access port (for example the container can be an intravenous solution bag or a vial having a stopper pierceable by a needle such as a hypodermic injection needle).
  • the container can hold a pharmaceutical composition such as a pharmaceutical composition having an agent that is effective for blocking B7 interactions with its ligand and/or treating an immune system disease.
  • the kit can also comprise a second container comprising one or more second agents as described herein (e.g., any of the DMARDS or NSAIDS) and/or a pharmaceutically acceptable buffer, such as phosphate-buffered saline, Ringer's solution and dextrose solution. It may further include other materials desirable from a commercial and user standpoint, including other buffers, diluents, filters, needles, syringes, and package inserts with instructions for use.
  • a second agent e.g., any of the DMARDS or NSAIDS
  • a pharmaceutically acceptable buffer such as phosphate-buffered saline, Ringer's solution and dextrose solution. It may further include other materials desirable from a commercial and user standpoint, including other buffers, diluents, filters, needles, syringes, and package inserts with instructions for use.
  • the kit may also suitably include a label and/or instructions on, or associated with the container.
  • the label can provide directions for carrying out the preparation of the agents for example, dissolving of the dry powders, and/or treatment for a specific immune system disease.
  • the label and/or the instructions can indicate directions for either in vivo or in vitro use of the pharmaceutical composition.
  • the label and/or the instructions can indicate that the pharmaceutical composition is used alone, or in combination with a second agent.
  • the label can indicate appropriate dosages for the molecules of the invention.
  • the label can indicate that dosages for a molecule that is effective for blocking B7 interactions with its ligand and/or treating an immune system disease is about 0.1 to 100 mg/kg weight of the subject, about 0.5 to 5 mg/kg weight of a subject, about 5 to 10 mg/kg weight of a subject, about 10 to 15 mg/kg weight of a subject, about 15 to 20 mg/kg weight of a subject, about 20 to 25 mg/kg weight of a subject, about 25 to 30 mg/kg weight of a subject, about 30 to 35 mg/kg weight of a subject, about 35 to 40 mg/kg weight of a subject, about 40 to 45 mg/kg of a subject, about 45 to 50 mg/kg weight of a subject, about 50 to 55 mg/kg weight of a subject, about 55 to 60 mg/kg weight of a subject, about 60 to 65 mg/kg weight of a subject, about 65 to 70 mg/kg weight of a subject, about 70 to 75 mg
  • the label and/or instructions can also indicate dosages for a second agent, such as a DMARD, is about 1 to about 5000 mg/day, about 1 to 10 mg/day, about 10 to 50 mg/day, about 50 to 100 mg/day, about 100 to 150 mg/day, about 150 to 200 mg/day, about 200 to 250 mg/day, about 250 to 300 mg/day, about 300 to 350 mg/day, about 350 to 400 mg/day, about 400 to 450 mg/day, about 450 to 500 mg/day, about 500 to 550 mg/day, about 550 to 600 mg/day, about 600 to 650 mg/day, about 650 to 700 mg/day, about 700 to 750 mg/day, about 750 to 800 mg/day, about 800 to 850 mg/day, about 850 to 900 mg/day, about 900 to 950 mg/day, about 950 to 1000 mg/day, about 1000 to 1100 mg/day, about 1100 to 1200 mg/day, about 1200 to 1300 mg/day, about 1300 to
  • the label and/or the instructions can also indicate that the pharmaceutical composition can be used alone, or in combination, with a second agent to treat a condition of choice e.g., immune system diseases, autoimmune diseases, immunoproliferative diseases, graft-related disorders, graft versus host disease (GVHD) (e.g., such as may result from bone marrow transplantation, or in the induction of tolerance), immune disorders associated with graft transplantation rejection, immune disorders associated with chronic rejection, immune disorders associated with tissue or cell allo- or xenografts (e.g., kidneys, skin, islets, muscles, hepatocytes, neurons, solid organs and the like), psoriasis, T cell lymphoma, T cell acute lymphoblastic leukemia, testicular angiocentric T cell lymphoma, benign lymphocytic angiitis, as lupus (e.g., lupus erythematosus, lupus nephriti
  • insulin dependent diabetes mellitus type I diabetes mellitus, type II diabetes mellitus
  • good pasture's syndrome myasthenia gravis, pemphigus, Crohn's disease, sympathetic ophthalmia, autoimmune uveitis, multiple sclerosis, autoimmune hemolytic anemia, idiopathic thrombocytopenia, primary biliary cirrhosis, chronic action hepatitis, ulcerative colitis, Sjogren's syndrome, rheumatic diseases (e.g., rheumatoid arthritis), polymyositis, scleroderma, mixed connective tissue disease, and the like.
  • rheumatic diseases e.g., rheumatoid arthritis
  • polymyositis scleroderma
  • mixed connective tissue disease and the like.
  • the kit comprises a pharmaceutical composition comprising a pharmaceutically acceptable carrier and an effective amount of a first agent, wherein the first agent is a molecule that blocks B7 interaction with CTLA4 and/or CD28 such as soluble CTLA4 molecules, CD28 molecules, B7 (B7-1 or B7-2) molecules, anti-CTLA4 monoclonal antibodies, anti-CD28 monoclonal antibodies or anti-B7 (B7-1 or B7-2) monoclonal antibodies.
  • the soluble CTLA4 molecules have the amino acid sequence of the extracellular domain of CTLA4 as shown in either FIG. 24 or 19 (CTLA4Ig or L104EA29Y, respectively).
  • the invention provides methods for regulating functional CTLA4- and CD28-positive cell interactions with B7-positive cells.
  • the methods comprise contacting the B7-positive cells with a soluble CTLA4 molecule of the invention so as to regulate functional CTLA4- and CD28-positive cell interactions with B7-positive cells, e.g., by interfering with reaction of an endogenous CTLA4 and/or CD28 molecule with a B7 molecule.
  • Suitable amounts of soluble CTLA4 for use in the methods of the invention are described supra.
  • the present invention also provides methods for inhibiting T-cell function but not T-cell depletion in a human by contacting B7-positive cells in the human with a soluble CTLA4.
  • soluble CTLA4 include CTLA4Ig and soluble CTLA4 mutant molecule e.g. L104EA29YIg.
  • the present invention further provides methods for treating immune system diseases and auto-immune diseases such as rheumatic diseases.
  • the methods comprise administering a therapeutic composition, comprising soluble CTLA4 molecules of the invention, to a subject in an amount effective to relieve at least one of the symptoms associated with immune system diseases.
  • the invention may provide long-term therapy for immune system diseases by blocking the T-cell/B7-positive cell interactions, thereby blocking T-cell activation/stimulation by co-stimulatory signals such as B7 binding to CD28, leading to induction of T-cell anergy or tolerance.
  • Immune system diseases include, but are not limited to, autoimmune diseases, immunoproliferative diseases, and graft-related disorders.
  • graft-related diseases include graft versus host disease (GVHD) (e.g., such as may result from bone marrow transplantation, or in the induction of tolerance), immune disorders associated with graft transplantation rejection, chronic rejection, and tissue or cell allo- or xenografts, including allo- or xenografts solid organs (e.g., kidneys), skin, islets, muscles, hepatocytes, neurons.
  • GVHD graft versus host disease
  • immune disorders associated with graft transplantation rejection chronic rejection
  • tissue or cell allo- or xenografts including allo- or xenografts solid organs (e.g., kidneys), skin, islets, muscles, hepatocytes, neurons.
  • immunoproliferative diseases include, but are not limited to, psoriasis; T cell lymphoma; T cell acute lymphoblastic leukemia; testicular angiocentric T cell lymphoma; benign lymphocytic angiitis; and autoimmune diseases such as lupus (e.g., lupus erythematosus, lupus nephritis), Hashimoto's thyroiditis, primary myxedema, Graves' disease, pernicious anemia, autoimmune atrophic gastritis, Addison's disease, diabetes (e.g.
  • insulin dependent diabetes mellitus type I diabetes mellitus, type II diabetes mellitus
  • good pasture's syndrome myasthenia gravis, pemphigus, Crohn's disease, sympathetic ophthalmia, autoimmune uveitis, multiple sclerosis, autoimmune hemolytic anemia, idiopathic thrombocytopenia, primary biliary cirrhosis, chronic action hepatitis, ulcerative colitis, Sjogren's syndrome, rheumatic diseases (e.g., rheumatoid arthritis), polymyositis, scleroderma, and mixed connective tissue disease.
  • rheumatic diseases e.g., rheumatoid arthritis
  • polymyositis scleroderma
  • mixed connective tissue disease mixed connective tissue disease.
  • the soluble CTLA4 molecules of the invention exhibit inhibitory properties in vivo. Under conditions where T-cell/B7-positive cell interactions, for example T cell/B cell interactions, are occurring as a result of contact between T cells and B7-positive cells, binding of introduced CTLA4 molecules to react to B7-positive cells, for example B cells, may interfere, i.e., inhibit, the T cell/B7-positive cell interactions resulting in regulation of immune responses.
  • the invention provides methods for regulating immune responses.
  • Immune responses downregulated (reduced) by the soluble CTLA4 molecules of the invention may be by way of inhibiting or blocking an immune response already in progress or may involve preventing the induction of an immune response.
  • the soluble CTLA4 molecules of the invention may inhibit the functions of activated T cells, such as T lymphocyte proliferation, cytokine secretion and/or cytokine production, by suppressing T cell responses or by inducing specific tolerance in T cells, or both.
  • the soluble CTLA4 molecules of this invention interfering with the CTLA4/CD28/B7 pathway may inhibit T-cell proliferation and/or cytokine secretion, and thus result in reduced tissue destruction and induction of T-cell unresponsiveness or anergy.
  • a preferred embodiment of the invention comprises use of the soluble CTLA4 mutant molecule L104EA29YIg to regulate functional CTLA4- and CD28-positive cell interactions with B7-positive cells, to treat immune system diseases such as rheumatic diseases and/or to downregulate immune responses.
  • the L104EA29YIg of the invention is a soluble CTLA4 mutant molecule comprising at least the two amino acid changes, the leucine (L) to glutamic acid (E) at position +104 and the alanine (A) to tyrosine (Y) change at position +29 (FIG. 19).
  • the L104EA29YIg molecule may encompass further mutations beyond the two specified herein.
  • a preferred embodiment of the invention comprises use of a molecule to block the interaction of B7 with CTLA4 and/or CD28 in conjunction with a DMARD to regulate an immune response in order to treat an immune system disease such as a rheumatic disease.
  • a molecule to block the interaction of B7 with CTLA4 and/or CD28 in conjunction with a DMARD to regulate an immune response in order to treat an immune system disease such as a rheumatic disease.
  • Suitable amounts of the molecule used to block the B7 interaction with CTLA4 and/or CD28 are described supra.
  • the molecule used to block the B7/CTLA4 interaction may be a soluble CTLA4 such as CTLA4Ig, CTLA4Ig/CD28Ig or L104EA29YIg, a soluble CD28 such as CD28Ig, a soluble B7 (B7-1 or B7-2) such as B7Ig, anti-CTLA4 monoclonal antibodies, anti-CD28 monoclonal antibodies or anti-B7 monoclonal antibodies.
  • a soluble CTLA4 such as CTLA4Ig, CTLA4Ig/CD28Ig or L104EA29YIg
  • a soluble CD28 such as CD28Ig
  • a soluble B7 B7-1 or B7-2
  • B7Ig anti-CTLA4 monoclonal antibodies, anti-CD28 monoclonal antibodies or anti-B7 monoclonal antibodies.
  • the DMARD may be a dihydrofolic acid reductase inhibitor such as methotrexate, cyclophosphamide, cyclosporine, cyclosporin A, chloroquine, hydroxychloroquine, sulfasalazine, sulphasalazopyrine, leflunomide, gold salts, D-penicillamine, azathioprine, anakinra, infliximab, etanercept, TNF ⁇ blockers or a biological agent that targets an inflammatory cytokine.
  • a dihydrofolic acid reductase inhibitor such as methotrexate, cyclophosphamide, cyclosporine, cyclosporin A, chloroquine, hydroxychloroquine, sulfasalazine, sulphasalazopyrine, leflunomide, gold salts, D-penicillamine, azathioprine, anakinr
  • a preferred embodiment includes methods for treating a rheumatic disease, such as rheumatoid arthritis, by administering an effective amount of soluble CTLA4 molecules alone, or in conjunction with an effective amount of methotrexate or a molecule that blocks TNF interactions, to a subject.
  • the methods of the invention also may be used to reduce at least one symptom associated with rheumatoid arthritis, including reducing erythrocyte sedimentation rates, serum levels of C-reactive protein, soluble ICAM-1, soluble E-selectin and/or soluble IL-2r.
  • the amount of symptom relief provided by the present invention can be measured using any of the accepted criteria established to measure and document symptom relief in a clinical setting.
  • Acceptable criteria for measuring symptom relief may include scores based on the criteria established by the American College of Rheumatology (e.g., ACR 20), the four measures of symptom relief (in: “CDER Guideline for the Clinical Evaluation of Anti-Inflammatory and Antirheumatic Drugs—FDA 1988), and the Health Assessment Questionnaire (HAQ) (Fries, J. F., et al., 1982 J of Rheumatology 9:789-793).
  • HAQ Health Assessment Questionnaire
  • the present invention provides improving ACR response rates using the methods of the invention.
  • the embodiments of the invention include improving ACR response rates of ACR 20, 50, and/or 70, using the methods of the invention.
  • the subjects treated by the present invention include mammalian subjects, including, human, monkey, ape, dog, cat, cow, horse, goat, pig, rabbit, mouse and rat.
  • the present invention provides various methods, local or systemic, for administering the therapeutic compositions of the invention such as soluble CTLA4 molecule alone or in conjunction with a DMARD, such as methotrexate, a molecule that blocks TNF interactions and/or other therapeutic drug.
  • the methods include intravenous, intramuscular, intraperitoneal, oral, inhalation and subcutaneous methods, as well as implantable pump, continuous infusion, gene therapy, liposomes, suppositories, topical contact, vesicles, capsules, biodegradable polymers, hydrogels, controlled release patch and injection methods.
  • the therapeutic agent, compounded with a carrier is commonly lyophilized for storage and is reconstituted with water or a buffered solution with a neutral pH (about pH 7-8, e.g., pH 7.5) prior to administration.
  • compositions of the invention may be administered to the subject in any pharmaceutically acceptable form.
  • the methods comprise administering to a subject the soluble CTLA4 molecules of the invention to regulate CD28- and/or CTLA4-positive cell interactions with B7-positive cells.
  • the B7-positive cells are contacted with an effective amount of the soluble CTLA4 molecules of the invention, or fragments or derivatives thereof, so as to form soluble CTLA4/B7 complexes. Suitable amounts of soluble CTLA4 are described supra.
  • the complexes interfere with interaction between endogenous CTLA4 and CD28 molecules with B7 family molecules.
  • the soluble CTLA4 molecules may be administered to a subject in an amount and for a time (e.g. length of time and/or multiple times) sufficient to block endogenous B7 molecules from binding their respective ligands, in the subject. Blockage of endogenous B7/ligand binding thereby inhibiting interactions between B7-positive cells with CD28-and/or CTLA4-positive cells.
  • soluble CTLA4 may be administered to a subject daily, weekly, monthly and/or yearly, in single or multiple times per day/week/month/year, depending on need. For example, in one embodiment, the molecule may initially be administered once every two weeks for a month, and then once every month thereafter.
  • soluble CTLA4 molecules may be administered in an amount from about 0.1 to 100 mg/kg weight of the patient/day. Suitable amounts of soluble CTLA4 are described supra. Methotrexate may be administered to a subject in an amount from about 0.1 to 100 mg/week. Suitable amounts of soluble methotrexate are described supra. A molecule that blocks TNF interactions e.g., etanercept, may be administered to a subject in an amount from about 0.1 to 100 mg/week. Suitable amounts of TNF blockers are described supra.
  • the invention also encompasses the use of the compositions of the invention together with other pharmaceutical agents to treat immune system diseases.
  • rheumatic diseases may be treated with molecules of the invention in conjunction with, but not limited to, immunosuppressants such as corticosteroids, cyclosporin (Mathiesen 1989 Cancer Lett. 44(2):151-156), prednisone, azathioprine, (R. Handschumacher, in: “Drugs Used for Immunosuppression” pages 1264-1276), TNF ⁇ blockers or antagonists (New England Journal of Medicine, vol. 340: 253-259, 1999; The Lancet vol. 354: 1932-39, 1999, Annals of Internal Medicine, vol.
  • immunosuppressants such as corticosteroids, cyclosporin (Mathiesen 1989 Cancer Lett. 44(2):151-156), prednisone, azathioprine, (R. Handschumacher, in: “Drugs Used for Immunosuppression” pages 1264-1276
  • the soluble CTLA4 molecules can also be used in combination with one or more of the following agents to regulate an immune response: soluble gp39 (also known as CD40 ligand (CD40L), CD154, T-BAM, TRAP), soluble CD29, soluble CD40, soluble CD80 (e.g. ATCC 68627), soluble CD86, soluble CD28 (e.g. ATCC accession number 68628), soluble CD56, soluble Thy-1, soluble CD3, soluble TCR, soluble VLA-4, soluble VCAM-1, soluble LECAM-1, soluble ELAM-1, soluble CD44, antibodies reactive with gp39 (e.g.
  • ATCC HB-10916, ATCC HB-12055 and ATCC HB-12056 antibodies reactive with CD40 (e.g. ATCC HB-9110), antibodies reactive with B7 (e.g. ATCC HB-253, ATCC CRL-2223, ATCC CRL-2226, ATCC HB-301, ATCC HB-11341, etc), antibodies reactive with CD28 (e.g. ATCC HB-11944 or mAb 9.3 as described by Martin et al (J. Clin. Immun. 4(1):18-22, 1980), antibodies reactive with LFA-1 (e.g.
  • antibodies reactive with LFA-2 antibodies reactive with IL-2, antibodies reactive with IL-12, antibodies reactive with IFN-gamma, antibodies reactive with CD2, antibodies reactive with CD48, antibodies reactive with any ICAM (e.g., ICAM-1 (ATCC CRL-2252), ICAM-2 and ICAM-3), antibodies reactive with CTLA4 (e.g. ATCC HB-304), antibodies reactive with Thy-1, antibodies reactive with CD56, antibodies reactive with CD3, antibodies reactive with CD29, antibodies reactive with TCR, antibodies reactive with VLA-4, antibodies reactive with VCAM-1, antibodies reactive with LECAM-1, antibodies reactive with ELAM-1, antibodies reactive with CD44.
  • monoclonal antibodies are preferred.
  • antibody fragments are preferred.
  • the combination can include: the soluble CTLA4 molecules of the invention and one other immunosuppressive agent; the soluble CTLA4 molecules with two other immunosuppressive agents; the soluble CTLA4 molecules with three other immunosuppressive agents; and the like.
  • the determination of the optimal combination and dosages can be determined and optimized using methods well known in the art.
  • L104EA29YIg and CD80 monoclonal antibodies mAbs
  • L104EA29YIg and CD86 mAbs L104EA29YIg, CD80 mAbs, and CD86 mAbs
  • L104EA29YIg and gp39 mAbs L104EA29YIg and CD40 mAbs
  • L104EA29YIg and CD28 mAbs L104EA29YIg, CD80 and CD86 mAbs, and gp39 mAbs
  • L104EA29YIg, CD80 and CD86 mAbs and CD40 mAbs and L104EA29YIg, anti-LFA1 mAb, and anti-gp39 mAb.
  • a specific example of a gp39 mAb is MR1.
  • Other combinations will be readily appreciated and understood by persons skilled in the art.
  • the soluble CTLA4 molecules of the invention may be administered as the sole active ingredient or together with other drugs in immunomodulating regimens or other anti-inflammatory agents such as DMARDs e.g. for the treatment or prevention of allo- or xenograft acute or chronic rejection or inflammatory or autoimmune disorders, or to induce tolerance.
  • DMARDs e.g. for the treatment or prevention of allo- or xenograft acute or chronic rejection or inflammatory or autoimmune disorders, or to induce tolerance.
  • DMARDs e.g. for the treatment or prevention of allo- or xenograft acute or chronic rejection or inflammatory or autoimmune disorders, or to induce tolerance.
  • a calcineurin inhibitor e.g. cyclosporin A or FK506
  • an immunosuppressive macrolide e.g. rapamycine or a derivative thereof (e.g. 40-O-(2-hydroxy)ethyl-rapamycin)
  • a lymphocyte homing agent e.g
  • CTLA4/CD28-Ig or other adhesion molecule inhibitors, e.g. mAbs or low molecular weight inhibitors including LFA-1 antagonists, Selectin antagonists and VLA-4 antagonists.
  • the compound is particularly useful in combination with a compound that interferes with CD40 and its ligand, e.g. antibodies to CD40 and antibodies to CD40-L.
  • soluble CTLA4 mutant molecules of the invention are administered in conjunction with other immunosuppressive/immunomodulatory or anti-inflammatory therapy, e.g. as hereinabove specified
  • dosages of the co-administered immunosuppressant, immunomodulatory or anti-inflammatory compound will of course vary depending on the type of co-drug employed, e.g. whether it is a steroid or a cyclosporin, on the specific drug employed, on the condition being treated and so forth.
  • the present invention provides in a yet further aspect methods as defined above comprising co-administration, e.g. concomitantly or in sequence, of a therapeutically effective amount of soluble CTLA4 molecules of the invention, e.g. CTLA4Ig and/or L104EA29YIg, in free form or in pharmaceutically acceptable salt form, and a second drug substance, said second drug substance being an immunosuppressant, immunomodulatory or anti-inflammatory drug, e.g. as indicated above.
  • a therapeutically effective amount of soluble CTLA4 molecules of the invention e.g. CTLA4Ig and/or L104EA29YIg
  • a second drug substance being an immunosuppressant, immunomodulatory or anti-inflammatory drug, e.g. as indicated above.
  • kits comprising a soluble CTLA4 molecule, in free form or in pharmaceutically acceptable salt form, to be used concomitantly or in sequence with at least one pharmaceutical composition comprising an immunosuppressant, immunomodulatory or anti-inflammatory drug e.g., a DMARD, NSAID, glucocorticoid or corticosteroid.
  • the kit may comprise instructions for its administration.
  • the kits of the invention can be used in any method of the present invention.
  • These vectors can include commonly used prokaryotic control sequences which are defined herein to include promoters for transcription initiation, optionally with an operator, along with ribosome binding site sequences, including such commonly used promoters as the beta-lactamase (penicillinase) and lactose (lac) promoter systems (Chang, et al., (1977) Nature 198:1056), the tryptophan (trp) promoter system (Goeddel, et al., (1980) Nucleic Acids Res. 8:4057) and the lambda derived P L promoter and N-gene ribosome binding site (Shimatake, et al., (1981) Nature 292:128).
  • promoters as the beta-lactamase (penicillinase) and lactose (lac) promoter systems (Chang, et al., (1977) Nature 198:1056)
  • trp tryptophan
  • trp lambda
  • Such expression vectors will also include origins of replication and selectable markers, such as a beta-lactamase or neomycin phosphotransferase gene conferring resistance to antibiotics, so that the vectors can replicate in bacteria and cells carrying the plasmids can be selected for when grown in the presence of antibiotics, such as ampicillin or kanamycin.
  • origins of replication such as a beta-lactamase or neomycin phosphotransferase gene conferring resistance to antibiotics, so that the vectors can replicate in bacteria and cells carrying the plasmids can be selected for when grown in the presence of antibiotics, such as ampicillin or kanamycin.
  • the expression plasmid can be introduced into prokaryotic cells via a variety of standard methods, including but not limited to CaCl 2 -shock (Cohen, (1972) Proc. Natl. Acad. Sci. USA 69:2110, and Sambrook et al. (eds.), “ Molecular Cloning: A Laboratory Manual”, 2nd Edition, Cold Spring Harbor Press, (1989)) and electroporation.
  • CHO-K1 Tet-On cell line CCL-61
  • CHO designated ECACC 85050302 (CAMR, Salisbury, Wiltshire, UK)
  • CHO clone 13 (GEIMG, Genova, IT)
  • CHO clone B (GEIMG, Genova, IT)
  • CHO-K1/SF designated ECACC 93061607 (CAMR, Salisbury, Wiltshire, UK)
  • RR-CHOK1 designated ECACC 92052129 (CAMR, Salisbury, Wiltshire, UK).
  • Exemplary plant cells include tobacco (whole plants, cell culture, or callus), corn, soybean, and rice cells. Corn, soybean, and rice seeds are also acceptable.
  • Nucleic acid sequences encoding the CTLA4 mutant molecules can also be inserted into a vector designed for expressing foreign sequences in an eukaryotic host.
  • the regulatory elements of the vector can vary according to the particular eukaryotic host.
  • Commonly used eukaryotic control sequences for use in expression vectors include promoters and control sequences compatible with mammalian cells such as, for example, CMV promoter (CDM8 vector) and avian sarcoma virus (ASV) ( ⁇ rLN vector).
  • CMV promoter CDM8 vector
  • ASV avian sarcoma virus
  • Other commonly used promoters include the early and late promoters from Simian Virus 40 (SV40) (Fiers, et al., (1973) Nature 273:113), or other viral promoters such as those derived from polyoma, Adenovirus 2, and bovine papilloma virus.
  • An inducible promoter such as hMTII (Karin, et al., (1982) Nature 299:797-802) may also be used.
  • expression vectors for eukaryotic host cells include, but are not limited to, vectors for mammalian host cells (e.g., BPV-1, pHyg, pRSV, pSV2, pTK2 (Maniatis); pIRES (Clontech); pRc/CMV2, pRc/RSV, pSFV1 (Life Technologies); pVPakc Vectors, pCMV vectors, pSG5 vectors (Stratagene)), retroviral vectors (e.g., pFB vectors (Stratagene)), pcDNA-3 (Invitrogen) or modified forms thereof, adenoviral vectors; Adeno-associated virus vectors, baculovirus vectors, yeast vectors (e.g., pESC vectors (Stratagene)).
  • mammalian host cells e.g., BPV-1, pHyg, pRSV, pSV2, pTK2 (Maniatis); p
  • Nucleic acid sequences encoding CTLA4 mutant molecules can integrate into the genome of the eukaryotic host cell and replicate as the host genome replicates.
  • the vector carrying CTLA4 mutant molecules can contain origins of replication allowing for extrachromosomal replication.
  • the origin of replication from the endogenous yeast plasmid can be used.
  • the 2 ⁇ circle can be used.
  • sequences from the yeast genome capable of promoting autonomous replication can be used (see, for example, Stinchcomb et al., (1979) Nature 282:39); Tschemper et al., (1980) Gene 10:157; and Clarke et al., (1983) Meth. Enz. 101:300).
  • Transcriptional control sequences for yeast vectors include promoters for the synthesis of glycolytic enzymes (Hess et al., (1968) J. Adv. Enzyme Reg. 7:149; Holland et al., (1978) Biochemistry 17:4900). Additional promoters known in the art include the CMV promoter provided in the CDM8 vector (Toyama and Okayama, (1990) FEBS 268:217-221); the promoter for 3-phosphoglycerate kinase (Hitzeman et al., (1980) J. Biol. Chem. 255:2073), and those for other glycolytic enzymes.
  • Regulatory sequences may also be placed at the 3′ end of the coding sequences. These sequences may act to stabilize messenger RNA. Such terminators are found in the 3′ untranslated region following the coding sequences in several yeast-derived and mammalian genes.
  • Exemplary vectors for plants and plant cells include, but are not limited to, Agrobacterium T i plasmids, cauliflower mosaic virus (CaMV), and tomato golden mosaic virus (TGMV).
  • Mammalian cells can be transformed by methods including but not limited to, transfection in the presence of calcium phosphate, microinjection, electroporation, or via transduction with viral vectors.
  • CTLA4 mutant molecules of the inventions can be harvested by methods well known in the art such as cell lysis (e.g. sonication, lysozyme and/or detergents) and protein recovery performed using standard protein purification means, e.g., affinity chromatography or ion-exchange chromatography, to yield substantially pure product (R. Scopes in: “ Protein Purification, Principles and Practice ”, Third Edition, Springer-Verlag (1994); Sambrook et al. (eds.), “ Molecular Cloning: A Laboratory Manual”, 2nd Edition, Cold Spring Harbor Press, (1989)).
  • Expression of CTLA4 mutant molecules can be detected by methods known in the art. For example, the mutant molecules can be detected by Coomassie staining SDS-PAGE gels and immunoblotting using antibodies that bind CTLA4.
  • a CTLA4Ig encoding plasmid was first constructed, and shown to express CTLA4Ig molecules as described in U.S. Pat. Nos. 5,434,131, 5,885,579 and 5,851,795. Then single-site mutant molecules (e.g., L104EIg) were generated from the CTLA4Ig encoding sequence, expressed and tested for binding kinetics for various B7 molecules.
  • the L104EIg nucleotide sequence (as included in the sequence shown in FIG. 18) was used as a template to generate the double-site CTLA4 mutant sequences (as included in the sequences shown in FIGS. 19 - 22 ) which were expressed as proteins and tested for binding kinetics.
  • the double-site CTLA4 mutant sequences include: L104EA29YIg, L104EA29LIg, L104EA29TIg, and L104EA29WIg. Triple-site mutants were also generated.
  • CTLA4Ig comprising the extracellular domain of CTLA4 and an IgCgamma1 domain was constructed as described in U.S. Pat. Nos. 5,434,131, 5,844,095 and 5,851,795, the contents of which are incorporated by reference herein.
  • the extracellular domain of the CTLA4 gene was cloned by PCR using synthetic oligonucleotides corresponding to the published sequence (Dariavach et al., Eur. Journ. Immunol. 18:1901-1905 (1988)).
  • the template for this step was cDNA synthesized from 1 micro g of total RNA from H38 cells (an HTLV II infected T-cell leukemic cell line provided by Drs. Salahudin and Gallo, NCI, Bethesda, Md.).
  • a portion of the PCR product from the first step was reamplified, using an overlapping forward primer, encoding the N terminal portion of the oncostatin M signal peptide and containing a Hind III restriction endonuclease site, CTAGCCACTGAAGCTTCACCAATGGGTGTACTGCTCACACAGAGGACGCTGCTCAGTCTGGTCCTTGCACTC (SEQ ID NO: 3) and the same reverse primer.
  • test molecule can then be injected into the chamber containing the sensor chip and the amount of complementary protein that binds can be assessed based on the change in molecular mass which is physically associated with the dextran-coated side of the sensor chip; the change in molecular mass can be measured by the detector system.
  • single-site mutant nucleotide sequences were generated using non-mutated (e.g., wild-type) DNA encoding CTLA4Ig (U.S. Pat. Nos. 5,434,131, 5,844,095; 5,851,795; and 5,885,796; ATCC Accession No. 68629) as a template.
  • Mutagenic oligonucleotide PCR primers were designed for random mutagenesis of a specific codon by allowing any base at positions 1 and 2 of the codon, but only guanine or thymine at position 3 (XXG/T or also noted as NNG/T).
  • a specific codon encoding an amino acid could be randomly mutated to code for each of the 20 amino acids.
  • XXG/T mutagenesis yields 32 potential codons encoding each of the 20 amino acids.
  • the double-site mutant nucleotide sequences encoding CTLA4 mutant molecules such as L104EA29YIg (deposited on Jun. 19, 2000 with the American Type Culture Collection (ATCC), 10801 University Boulevard., Manassas, Va. 20110-2209 and accorded ATCC accession number PTA-2104), were generated by repeating the mutagenesis procedure described above using L104EIg as a template. This method was used to generate numerous double-site mutants nucleotide sequences such as those encoding CTLA4 molecules L104EA29YIg (as included in the sequence shown in FIG. 19), L104EA29LIg (as included in the sequence shown in FIG.
  • L104EA29TIg (as included in the sequence shown in FIG. 21)
  • L104EA29WIg (as included in the sequence shown in FIG. 22).
  • Triple-site mutants such as those encoding L104EA29YS25KIg, L104EA29YS25NIg and L104EA29YS25RIg, were also generated
  • nucleotide changes do not necessarily translate into amino acid changes as some codons redundantly encode the same amino acid. Any changes of nucleotide from the original or wildtype sequence, silent (i.e. causing no change in the translated amino acid) or otherwise, while not explicitly described herein, are encompassed within the scope of the invention.
  • CTLA4Ig Current in vitro and in vivo studies indicate that CTLA4Ig by itself is unable to completely block the priming of antigen specific activated T cells.
  • anti-CD86 monoclonal antibody did augment the inhibition, indicating that CTLA4Ig was not as effective at blocking CD86 interactions.
  • the soluble CTLA4 mutant molecules described in Example 1 above were screened using a novel screening procedure to identify several mutations in the extracellular domain of CTLA4 that improve binding avidity for CD80 and CD86.
  • This screening strategy provided an effective method to directly identify mutants with apparently slower “off” rates without the need for protein purification or quantitation since “off” rate determination is concentration independent (O'Shannessy et al., (1993) Anal. Biochem., 212:457-468).
  • Senosorgram baselines were normalized to zero response units (RU) prior to analysis. Samples were run over mock-derivatized flow cells to determine background RU values due to bulk refractive index differences between solutions. Equilibrium dissociation constants (K d ) were calculated from plots of R eq versus C, where R eq is the steady-state response minus the response on a mock-derivatized chip, and C is the molar concentration of analyte. Binding curves were analyzed using commercial nonlinear curve-fitting software (Prism, GraphPAD Software).
  • CHO cells (1-10 ⁇ 10 5 ) were first incubated with mAbs or immunoglobulin fusion proteins in DMEM containing 10% fetal bovine serum (FBS), then washed and incubated with fluorescein isothiocyanate-conjugated goat anti-mouse or anti-human immunoglobulin second step reagents (Tago, Burlingame, Calif.). Cells were given a final wash and analyzed on a FACScan (Becton Dickinson).
  • FBS fetal bovine serum
  • CTLA4X C120S and L104EA29YX X120S are identical to CTLA4X C120S and L104EA29YX X120S .
  • Single chain CTLA4X C120S was prepared as previously described (Linsley et al., (1995) J. Biol. Chem., 270:15417-15424). Briefly, an oncostatin M CTLA4 (OMCTLA4) expression plasmid was used as a template, the forward primer, GAGGTGATAAAGCTTCACCAATGGGTGTACTGCTCACACAG (SEQ ID NO: 4) was chosen to match sequences in the vector; and the reverse primer, GTGGTGTATTGGTCTAGATCAATCAGAATCTGGGCACGGTTC (SEQ ID NO: 5) corresponded to the last seven amino acids (i.e.
  • nucleotide sequences AGA, GGA, TGA, CGA, ACT, or GCT are the reversed complementary sequences of the codons for serine.
  • Polymerase chain reaction products were digested with HindIII/XbaI and directionally subcloned into the expression vector 7rLN (Bristol-Myers Squibb Company, Princeton, N.J.).
  • L104EA29YX C120S was prepared in an identical manner. Each construct was verified by DNA sequencing.
  • L104EA29YIg ⁇ 1 ⁇ g; lane 3
  • L104EIg ⁇ 1 ⁇ g; lane 2
  • CTLA4Ig ⁇ 1 ⁇ g; lane 1
  • reducing ⁇ 50 kDa; +BME; plus 2-mercaptoethanol
  • non-reducing ⁇ 100 kDa; ⁇ ME
  • CTLA4Ig did not have the same mobility as dimeric CTLA4Ig (FIG. 25B).
  • the major peaks represent protein dimer while the faster eluting minor peak in FIG. 25B represents higher molecular weight aggregates.
  • Approximately 5.0% of CTLA4Ig was present as higher molecular weight aggregates but there was no evidence of aggregation of L104EA29YIg or L104EIg. Therefore, the stronger binding to CD86Ig seen with L104EIg and LI04EA29YIg could not be attributed to aggregation induced by mutagenesis.
  • the lower K d of L104EA29YIg (3.66 nM) than L104EIg (4.47 nM) or CTLA4Ig (6.51 nM) indicates higher binding avidity of L104EA29YIg to CD80Ig.
  • L104EA29YX C120S bound with approximately 2-fold higher affinity than CTLA4X C120S to both CD80Ig and CD86Ig.
  • the increased affinity was due to approximately 3-fold slower rate of dissociation from both ligands. Therefore, stronger ligand binding by L104EA29Y was most likely due to avidity enhancing structural changes that had been introduced into each monomeric chain rather than alterations in which the molecule dimerized.
  • FACS analysis (FIG. 27) of CTLA4Ig and mutant molecules binding to stably transfected CD80+ and CD86+CHO cells was performed as described herein.
  • CD80-positive and CD86-positive CHO cells were incubated with increasing concentrations of CTLA4Ig, L104EA29YIg, or L104EIg, and then washed.
  • Bound immunoglobulin fusion protein was detected using fluorescein isothiocyanate-conjugated goat anti-human immunoglobulin.
  • CD4-positive T cells were isolated by immunomagnetic negative selection (Linsley et al., (1992) J. Exp. Med. 176:1595-1604). Isolated CD4-positive T cells were stimulated with phorbal myristate acetate (PMA) plus CD80-positive or CD86-positive CHO cells in the presence of titrating concentrations of inhibitor. CD4-positive T cells (8-10 ⁇ 10 4 /well) were cultured in the presence of 1 nM PMA with or without irradiated CHO cell stimulators. Proliferative responses were measured by the addition of 1 ⁇ Ci/well of [3H]thymidine during the final 7 hours of a 72 hour culture.
  • PMA phorbal myristate acetate
  • CD80-positive or CD86-positive CHO cells in the presence of titrating concentrations of inhibitor.
  • CD4-positive T cells (8-10 ⁇ 10 4 /well) were cultured in the presence of 1 nM PMA with or without
  • L104EA29YIg inhibits proliferation of CD80-positive PMA treated CHO cells more than CTLA4Ig (FIG. 28A).
  • L104EA29YIg is also more effective than CTLA4Ig at inhibiting proliferation of CD86-positive PMA treated CHO cells (FIG. 28B). Therefore, L104EA29YIg is a more potent inhibitor of both CD80- and CD86-mediated costimulation of T cells.
  • FIG. 29 shows inhibition by L104EA29YIg and CTLA4Ig of allostimulated human T cells prepared above, and further allostimulated with a human B lymphoblastoid cell line (LCL) called PM that expressed CD80 and CD86 (T cells at 3.0 ⁇ 10 4 /well and PM at 8.0 ⁇ 10 3 /well). Primary allostimulation occurred for 6 days, then the cells were pulsed with 3 H-thymidine for 7 hours, before incorporation of radiolabel was determined.
  • LCL human B lymphoblastoid cell line
  • cytokine production To measure cytokine production (FIG. 30), duplicate secondary allostimulation plates were set up. After 3 days, culture media was assayed using ELISA kits (Biosource, Camarillo, Calif.) using conditions recommended by the manufacturer. L104EA29YIg was found to be more potent than CTLA4Ig at blocking T cell IL-2, IL-4, and ⁇ -IFN (gamma-IFN) cytokine production following a secondary allogeneic stimulus (FIGS. 30 A-C).
  • L104EA29YIg and CTLA4Ig are shown in FIG. 31.
  • Peripheral blood mononuclear cells (PBMC'S; 3.5 ⁇ 10 4 cells/well from each monkey) from 2 monkeys were purified over lymphocyte separation medium (LSM) and mixed with 2 ⁇ g/ml phytohemaglutinin (PHA). The cells were stimulated 3 days then pulsed with radiolabel 16 hours before harvesting.
  • LSM lymphocyte separation medium
  • PHA phytohemaglutinin
  • the cells were stimulated 3 days then pulsed with radiolabel 16 hours before harvesting.
  • L104EA29YIg inhibited monkey T cell proliferation better than CTLA4Ig.
  • CTLA4 mutant molecule L104EA29YIg also known as LEA29Y or LEA
  • CTLA4Ig to relieve at least one symptom associated with rheumatoid arthritis, including reducing: joint swelling, joint tenderness, inflammation, morning stiffness, and pain.
  • the CTLA4Ig molecule used herein begins with methionine at position +1 (or alternatively with alanine at position ⁇ 1) and ends with lysine at position +357 as shown in FIG. 24. DNA encoding an embodiment of the CTLA4Ig molecule has been deposited as ATCC 68629.
  • the L104EA29YIg molecule used herein begins with methionine at position +1 (or alternatively with alanine at position ⁇ 1) and ends with lysine at position +357 as shown in FIG. 19.
  • DNA encoding an embodiment of the L104EA29YIg molecule has been deposited as ATCC PTA 2104.
  • the following provides a description of human patients administered L104EA29YIg or CTLA4Ig to relieve at least one biological surrogate marker associated with rheumatoid arthritis, including reducing erythrocyte sedimentation rates, and serum levels of C-reactive protein and/or IL2 receptor.
  • CTLA4Ig and L104EA29YIg were supplied in single-use glass vials containing 200 mg/vial of CTLA4Ig or 100 mg/vial of L104EA29YIg, respectively. Prior to infusion, the CTLA4Ig and L104EA29YIg were diluted to a final concentration of 25 mg/ml with sterile water for injection (SWFI).
  • SWFI sterile water for injection
  • Group 1 32 patients, CTLA4Ig or LI04EA29YIg matching placebo.
  • Group 2 26 patients; dosage 0.5 mg/kg of CTLA4Ig.
  • Group 3 32 patients; dosage 2.0 mg/kg of CTLA4Ig.
  • Group 4 32 patients; dosage 10.0 mg/kg of CTLA4Ig.
  • Group 5 32 patients; dosage 0.5 mg/kg of L104EA29YIg.
  • Group 6 29 patients; dosage 2.0 mg/kg of L104EA29YIg.
  • Group 7 31 patients; dosage 10.0 mg/kg of L104EA29YIg.
  • Patients were evaluated for baseline symptoms of disease activity prior to receiving any infusions. These baseline evaluations included: joint swelling, joint tenderness, inflammation, morning stiffness, disease activity evaluated by patient and physician as well as disability evaluated by Health Questionnaire Assessment (HAQ) (reported as a physical function score in FIG. 1C), and pain (FIGS. 1A to 1 D). Additionally, the baseline evaluations included erythrocyte sedimentation rates (ESR), and serum levels of C-reactive protein (CRP) and soluble IL-2 receptor (IL-2r) (FIGS. 1C and 1D).
  • ESR erythrocyte sedimentation rates
  • CRP C-reactive protein
  • IL-2r soluble IL-2 receptor
  • ACR American College of Rheumatology
  • a subject satisfied the ACR20 criterion if there was a 20 percent improvement in tender and swollen joint counts and 20 percent improvement in three of the five remaining symptoms measured, such as patient and physician global disease changes, pain, disability, and an acute phase reactant (Felson, D. T., et al., 1993 Arthritis and Rheumatism 36:729-740; Felson, D. T., et al., 1995 Arthritis and Rheumatism 38:1-9).
  • a subject satisfied the ACR50 or ACR70 criterion if there was a 50 or 70 percent improvement, respectively, in tender and swollen joint counts and 50 or 70 percent improvement, respectively, in three of the five remaining symptoms measured, such as patient and physician global disease changes, pain, physical disability, and an acute phase reactant such as CRP or ESR.
  • EIA enzyme immunoassay
  • IL-2sR ⁇ , sICAM-1, and sE-selectin were measured using commercially available calorimetric EIA kits from R&D Systems, Inc. (Minneapolis, Minn.). The lower and upper limits of quantitation were 312-20,000 pg/mL, 40-907 ng/mL and 10-206 ng/mL, respectively. The inter-assay coefficient of variation ranged from 4.48-8.4%, 3.8-5.0% and 5.5-9.0% respectively. According to the kit manufacturer, normal serum values range from 676-2,132 pg/mL, respectively.
  • MMP-3 was measured using a commercially available calorimetric EIA kit from Amersham Pharmacia Biotech (Piscataway, N.J.). The lower and upper limits of quantitation were 30-7,680 ng/mL. The inter-assay coefficient of variation ranged from 6.3-10.6%. According to the kit manufacturer, normal serum values range from 28-99 ng/mL.
  • IL-6 and TNF ⁇ were measured using commercially available chemiluminescent EIA kits from R&D Systems, Inc. (Minneapolis, Minn.). The lower and upper limits of quantitation were 0.3-3,000 pg/mL and 0.7-7,000 pg/mL, respectively. The inter-assay coefficient of variation ranged from 3.1-5.7% and 6.4-20.7%, respectively. According to the kit manufacturer, normal serum values range from ⁇ 0.3-12 pg/mL and ⁇ 0.7-7.5 pg/mL.
  • Serum samples were obtained for assessment of drug-specific antibodies prior to dosing on day 1, and approximately on days 15, 29, 57, 85 and 169. Due to high, preexisting titers directed to the immunoglobulin (Ig) portion of the molecule, specific antibody formation against CTLA4Ig and LEA29Y without Ig constant regions was also assessed.
  • Ig immunoglobulin
  • the plates were then washed and serial dilutions of the test sera or quality control (QC) sera were added to the appropriate wells and incubated for 2 hours at 37° C.
  • Sera was diluted threefold in PBS with 0.25% BSA and 0.05% Tween 20 starting at a 1:10 dilution. Plates were washed and an alkaline-phosphatase-conjugated goat anti-human kappa and lambda (Southern Biotechnology Associates, Inc., Birmingham, Ala.) antibody cocktail was added. Following a 1-hour incubation at 37° C., the plates were washed and 1 mg/ml para-nitrophenyl phosphate in diethanolamine buffer was added to each well.
  • EPT endpoint titer
  • Plate background was determined as the absorbance measurement recorded in the absence of serum. Values were considered positive for seroconversion if they were at least two serial dilutions (ninefold) or greater relative to predose EPT values.
  • Serum QC samples positive for either CTLA4Ig- or LEA29Y-specific antibodies were generated from immunized monkeys. An aliquot of the appropriate QC sample was assayed during each analytical run. Analytical runs were accepted only when the QC samples were within the assay acceptance criteria.
  • CTLA4Ig and L104EA29YIg were generally well-tolerated at all dose-levels. Peri-infusional adverse events were similar across all dose groups, with the exception of headaches. Headache response of patients on day 85 increased dose-dependently 23%, 44%, and 53% in CTLA4Ig-treated patients, and 34%, 45%, and 61% in L104EA29YIg-treated patients, at 0.5, 2.0, and 10.0 mg/kg respectively. In contrast, 31% of the patients administered placebos experienced headaches.
  • FIG. 3A The ACR-20, ⁇ 50, and ⁇ 70 responses of patients treated with CTLA4Ig, L104EA29YIg, or placebo at day 85 are summarized in FIG. 3A.
  • FIGS. 3B and C describe the ACR-20 responses with 95% confidence limits. The responses appear to be dose-dependent with a clear significant response at 10 mg/kg per body weight of the patient.
  • FIGS. 4A and B The percent of patients having reduced swollen and tender joint counts compared to the patients having no response to treatment with CTLA4Ig, L104EA29YIg, or placebo, is shown in FIGS. 4A and B.
  • the therapeutic responses appear to be dose-dependent.
  • a larger percentage of patients show improvement of 20, 50, 70, and even 100% in the 2 and 10 mg/kg groups for both products.
  • FIGS. 5A, B, C, and D The percent of patients having reduced pain, disease activity evaluated by patient and physician mean score units with CTLA4Ig, L104EA29YIg, or placebo, is shown in FIGS. 5A, B, C, and D.
  • the Likert scale is a validated verbal rating scale using adjectives to rank the symptoms (The American College of Rheumatology Preliminary Core Set of Disease Activity Measures for Rheumatoid Arthritis Clinical Trials: Arthritis and Rheumatism, June 1993, 36(6):729-740).
  • FIGS. 6A and B The patient and physician assessments of disease activity change from the baseline by at least 2 units, resulting from treatment with CTLA4Ig, L104EA29YIg, or placebo, are shown in FIGS. 6A and B.
  • the responses appear to be dose-dependent with more marked improvement for the higher doses of active drugs.
  • FIGS. 7A and B The percent reduction in C-reactive protein (CRP) levels in patients treated with CTLA4Ig, L104EA29YIg, or placebo, is shown in FIGS. 7A and B.
  • the responses appear to be dose-dependent with a clear decrease for the 2 and 10 mg/kg active treatment groups.
  • FIG. 7B showed that the difference is quite significant compared to placebo with 95% confidence intervals.
  • FIG. 7C shows the changes in serum level changes from baseline at day 85.
  • FIGS. 9A and B The median and mean tender joint counts in patients treated with CTLA4Ig or placebo over time are shown in FIGS. 9A and B.
  • the change from baseline e.g., reduction in tender joints appears to be more important in the 2 and 10 mg/kg treated groups, than in the placebo or 0.5 mg/kg groups.
  • FIGS. 10A and B The median and mean swollen joint counts in patients treated with CTLA4Ig or placebo over time are shown in FIGS. 10A and B.
  • the change from baseline e.g., reduction in swollen joints
  • FIGS. 12A and B The mean disease activity assessment scores assessed by patient or physician in patients treated with CTLA4Ig or placebo over time are shown in FIGS. 12A and B.
  • the change from baseline e.g., reduction in disease activity appears to be more important in the 2 and 10 mg/kg treated groups than placebo or 0.5 mg/kg groups.
  • FIGS. 14A and B The median and mean swollen joint counts in patients treated with L104EA29YIg (denoted as LEA in the figures) or placebo over time are shown in FIGS. 14A and B.
  • the change from baseline e.g., reduction in swollen joints appears to be more important in the 2 and 10 mg/kg treated groups than placebo or 0.5 mg/kg groups.
  • FIGS. 16A and B The mean disease activity assessment scores evaluated by patient or physician in patients treated with L104EA29YIg (denoted as LEA in the figures) or placebo over time are shown in FIGS. 16A and B.
  • the change from baseline e.g., reduction in disease activity appears to be dose-dependent.
  • FIG. 17 Health Assessment Questionnaire (HAQ); Fries, J. F., et al., 1982 J. of Rheumatology 9:789-793). There is a clear dose dependent improvement with this parameter.
  • the status of the bone structure is monitored in some of the human patients prior to treatment with CTLA4Ig or L104EA29YIg. These patients are administered from 0.5 to 20 mg/kg of CTLA4Ig or L104EA29YIg chronically every two to twelve weeks (alone or in combination with other agents) to maintain their therapeutic improvement over time. Radiographs of patients' hands and feet are taken at predefined intervals: 6 months, and then yearly, as recommended by the FDA guidelines. These patients are monitored in long-term extension after 6 and 12 months to determine if treatment with CTLA4Ig or L104EA29YIg reduces the progression of bone deterioration, and then yearly.
  • the patients are monitored by radiographic methods, including X-ray and/or magnetic resonance imaging (MRI), according to standard practice in the art (Larsen, A. K. and M. Eek 1977 Acta. Radiol. Diag. 18:481-491; Sharp, J. T., et al., 1985 Arthritis and Rheumatism 28:1326-1335).
  • MRI magnetic resonance imaging
  • the results of the radiographic data are evaluated for prevention of structural damage, including slowing the progression of bone erosion and cartilage damage, with joint space narrowing and/or prevention of new erosions.
  • RA Rheumatoid Arthritis
  • Methotrexate remains the cornerstone of the RA treatment. It was the first agent that demonstrated early onset of action, superior efficacy and tolerability compared to the classical DMARDs (e.g. gold, hydroxychloroquine, sulfasalazine) used to treat RA. Clinical benefit may be seen as early as 3 weeks after initiating treatment, and the maximal improvement is generally achieved by 6 months.
  • methotrexate has a number of limitations. For example, despite its increased tolerability, the window between efficacy and liver toxicity is quite narrow. Subjects treated with methotrexate require careful monitoring and unacceptable toxicity is often the reason for discontinuation of treatment.
  • Methotrexate also does not appear to efficiently control disease progression or joint deterioration. For some subjects, practitioners feel compelled to add a second DMARD with the hope of increasing efficacy despite the risk of increased toxicity.
  • co-treatment with methotrexate and a costimulator blocker e.g. CD80 and CD86 blockers such as CTLA4Ig
  • a costimulator blocker e.g. CD80 and CD86 blockers such as CTLA4Ig
  • CTLA4Ig As noted in Example 3, above, significant clinical responses and reductions in surrogate markers of disease activity were observed for CTLA4Ig at doses of 2 and 10 mg/kg with a good tolerability profile. It has also been confirmed that the composition CTLA4Ig, used in Example 3 above, did not induce any side effects. As a result, it was decided to continue the clinical development of CTLA4Ig for rheumatoid arthritis in Phase IIB.
  • This example describes a twelve month study in which primary efficacy was assessed after all subjects completed six months of treatment or discontinue therapy. Efficacy, safety, and disease progression were also assessed throughout the duration of the study.
  • the study utilized a randomized, double blind, placebo controlled, parallel dosing design.
  • the study was designed to evaluate the safety, clinical activity, immunogenicity and pharmacokinetics of two doses of CTLA4Ig: 2 or 10 mg/kg.
  • CTLA4Ig or a placebo were also administered on Day 15. Each dose of study medication was infused intravenously over approximately 30 minutes. The primary efficacy endpoint was the ACR 20 response rate after 6 months.
  • Analgesics that did not contain ASA or NSAIDs were permitted in subjects experiencing pain not adequately controlled by the baseline and study medications, except for 12 hours before a joint evaluation. Decreases in NSAIDs were permitted but only if due to adverse events such as gastrointestinal toxicity.
  • CTLA4Ig at 2 mg/kg or 10 mg/kg was infused every two weeks for the first month, and monthly thereafter for 12 months.
  • the primary endpoint of the first stage of the study was the proportion of subjects meeting the American College of Rheumatology criteria for 20% improvement (ACR 20) on Day 180 (month six).
  • the ACR 20 definition of improvement is a 20% improvement from baseline in the number of tender and swollen joint counts, and a 20% improvement from baseline in 3 of the following 5 core set measurements: subject global assessment of pain, subject global assessment of disease activity, physician global assessment of disease activity, subject assessment of physical function and acute phase reactant value (C-reactive protein (CRP)).
  • C-reactive protein (CRP) C-reactive protein
  • the evaluation for 50% improvement (ACR 50) and 70% improvement (ACR 70) follow similarly. Subjects who discontinued the study due to lack of efficacy (i.e. worsening RA) were considered as ACR non-responders from that time on. For all subjects who dropped out for other reasons, their ACR response at the time of discontinuation was carried forward.
  • CTLA4Ig Two doses of CTLA4Ig (2 mg/kg and 10 mg/kg) were compared with the placebo control group. All subjects were maintained at the same stable entry doses of methotrexate. The primary analysis was the comparison of CTLA4Ig 10 mg/kg with placebo. Sample sizes were based on a 5% level (2-tailed) of significance. Based on published studies, the placebo plus methotrexate control ACR 20 response rate at 6 months is about 25%. A sample of 107 subjects (adjusted for a possible 15% dropout) per treatment arm was determined to yield a 94% power to detect a difference of 25% at the 5% level (two-tailed).
  • the sample was determined to yield a power of 95% and 90% to detect differences of 20% and 14% in ACR 50 and ACR 70, respectively. If the comparison between CTLA4Ig 10 mg/kg and placebo was significant with regards to ACR 20, then the comparison between CTLA4Ig 2 mg/kg and placebo was carried out. This second testing should have a power of 88%. This sequentially rejective procedure based on Chi-square tests was also used to test for differences in ACR 50 and ACR 70 responses.
  • FIG. 39 shows the proportion of new tender and swollen joints at day 180 of the study after therapy with methotrexate alone or in combination with CTLA4Ig (administered at 2 or 10 mg/kg body weight of subject).
  • FIG. 46 shows the mean percent improvement in physical function from baseline as measured by HAQ.
  • CTLA4Ig health-related quality of life
  • HRQOL health-related quality of life
  • the SF-36 was administered to all subjects at baseline, 90 and 180 days.
  • the SF-36 consists of 36 items which covers eight domains (physical function, role-physical, bodily pain, general health, vitality, social function, role emotional, and mental health). These individual domains are used to derive the physical and mental component summary scores which range from 0 to 100, with higher scores indicating better quality of life. Absolute differences of 5 or more in the SF-36 scores were considered clinically meaningful.
  • CRP levels decreased from baseline in both CTLA4Ig-treated groups more than in the control group, with greater reduction observed in the 10 mg/kg dosing group (see FIGS. 47, 48 and 52 ).
  • Soluble IL-2r levels decreased from baseline in both CTLA4Ig-treated groups more than in the control group, with greater reduction observed in the 10 mg/kg dosing group (see FIG. 54).
  • Serum IL-6 levels decreased from in both CTLA4Ig-treated groups more than in the control group (see FIG. 55).
  • CTLA4Ig The effects of CTLA4Ig on serum TNF ⁇ levels were inconclusive. The 2 mg/kg group increased and the 10 mg/kg group decreased relative to the control group (see FIG. 56).
  • CTLA4Ig was well tolerated at all doses. There were no deaths, malignancies or opportunistic infections in any subjects receiving CTLA4Ig. Serious adverse events (SAEs) and non-serious adverse events (NSAEs) were similar or less frequent in the active-treatment groups compared to the control group.
  • SAEs Serious adverse events
  • NSAEs non-serious adverse events
  • CTLA4Ig significantly reduced the signs and symptoms of rheumatoid arthritis in subjects receiving methotrexate as assessed by ACR response criteria.
  • the effects of CTLA4Ig appear to increase in proportion to dose level.
  • the improvement from baseline in all ACR core components is higher in the 10 mg/kg group than the 2 mg/kg group.
  • CTLA4Ig at 10 mg/kg doses demonstrated clinically and statistically significant improvements in all 8 domains of the SF-36. All pharmacodynamic biomarkers assayed appeared to decrease in proportion to CTLA4Ig dose level except for TNF ⁇ .
  • CTLA4Ig was safe and well tolerated in subjects with rheumatoid arthritis receiving methotrexate.
  • the adverse event profile for both CTLA4Ig doses was similar to the control group.
  • CTLA4Ig in combination with etanercept, to treat patients with active Rheumatoid Arthritis.
  • Etanercept along with infliximab, comprises a new generation of Rheumatoid Arthritis drugs which targets Tumor Necrosis Factor (TNF.
  • Etanercept is a dimeric fusion protein having an extracellular portion of the TNF receptor linked to the Fc portion of human immunoglobulin (IgG1). This fusion protein binds to TNF, blocks its interactions with cell surface TNF receptors and render TNF molecules biologically inactive.
  • This example describes a twelve month study in which efficacy was assessed after all subjects completed six months of treatment or discontinued therapy. Efficacy, safety and disease progression were also assessed throughout the duration of the study.
  • CTLA4Ig Infusions of CTLA4Ig were given on Days 1, 15, 30, and monthly thereafter, for 6 months (primary treatment phase). Each dose of study medication was infused intravenously for approximately 30 minutes.
  • the primary treatment phase of the study took place during the first six months of treatment. During this period, subjects were required to remain on stable doses of etanercept (25 mg twice weekly). DMARDs other than etanercept were not permitted. Low-dose stable corticosteroid (at 10 mg daily or less) and/or stable non-steroidal anti-inflammatory drug (NSAID), including acetyl salicylic acid (ASA), use was allowed. Analgesics (that do not contain ASA or NSAIDs) were permitted in subjects experiencing pain that was not adequately controlled by the baseline and study medications, except for 12 hours before a joint evaluation.
  • NSAID stable non-steroidal anti-inflammatory drug
  • Analgesics that do not contain ASA or NSAIDs were permitted in subjects experiencing pain that was not adequately controlled by the baseline and study medications, except for 12 hours before a joint evaluation.
  • the primary endpoint of this study was to collect data regarding the proportion of subjects meeting modified American College of Rheumatology (ACR) criteria for 20% improvement (ACR 20) after six months.
  • the modified ACR 20 criteria were used to accommodate the low CRP levels in this study's subject population.
  • the modified ACR criteria were defined as 1) a greater than 20% improvement in tender and swollen joint count and 2) a greater than 20% improvement in 2 of the remaining 4 core data set measures (global pain, physician, subject, functional assessment).
  • CRP which is normally a part of the standard ACR core data sets, was not included in the modified ACR criteria due to the low levels of CRP in subjects using TNF blockers, such as etanercept.
  • the standard ACR criteria, and two alternative criteria were also evaluated as secondary endpoints.
  • sample was determined to yield a power of 91% and 83% to detect differences of 30 and 25% in ACR 50 and 70, respectively.
  • due to slow enrollment only 122 subjects were randomized and 121 treated and analyzed (one subject was randomized but never received treatment).
  • CTLA4Ig and etanercept had a dermatological malignancy.
  • One subject had a basal cell carcinoma that was excised after the Day 150 visit.
  • the other subject had a squamous cell carcinoma which was a pre-existing lesion that the subject decided to have removed after the Day 120 visit.
  • Another subject experienced angioedema that was considered by the investigator to be a drug reaction to azithromycin.
  • CTLA4Ig plus etanercept at 2 mg/kg was similar to that observed in subjects receiving the same dose of CTLA4Ig plus methotrexate therapy (Example 5).
  • the criteria for evaluation in the methotrexate (Example 5) trial was the standard ACR, that includes CRP among the core components, while in the etanercept trial (Example 6) the criteria for evaluation was the modified ACR, that excludes CRP.
  • CTLA4Ig (2 mg/kg) in combination with etanercept reduced the signs and symptoms of rheumatoid arthritis, as compared with etanercept alone.
  • the increases in the modified ACR20 and ACR 70 assays were statistically significant.
  • Efficacy of CTLA4Ig plus etanercept therapy was observed within one month of the start of treatment.
  • CTLA4Ig was generally safe and well tolerated when administered in combination with etanercept with the safety profile similar to etanercept therapy alone.
  • CTLA4Ig was not immunogenic during the six month trial period.
  • the efficacy of CTLA4Ig therapy in combination with etanercept (Example 6) was similar to the same dose of CTLA4Ig with methotrexate (Example 5).
  • the following example provides the one-year results from a Phase IIB, multi-center, randomized, double-blind, placebo controlled clinical study to evaluate the safety and clinical efficacy of administering two different doses of CTLA4Ig in combination with methotrexate to treat patients with active Rheumatoid Arthritis (RA).
  • the study presented in this example is a continuance of the six-month study presented in Example 5.
  • results presented in this clinical study report are based on data from an analysis performed after all subjects completed 6 months of treatment and again after all subjects completed 12 months of treatment.
  • the 10 mg/kg CTLA4Ig plus MTX group may be referred to as the 10 mg/kg group
  • the 2 mg/kg plus MTX group is referred to as the 2 mg/kg group
  • the CTLA4Ig (BMS-188667) placebo plus MTX group is referred to as the placebo group.
  • Treatment Groups Subjects were randomized 1:1:1 to one of three treatment groups:
  • Group 1 CTLA4Ig (BMS-188667) 10 mg/kg by intravenous infusion
  • Infusions were to occur at approximately the same time of day throughout the study. All doses of study medication were administered in a fixed volume of 75 mL at a constant rate over approximately 30 minutes. The intravenous bag and line were flushed with 25 mL of dextrose 5% in water (D5W) solution at the end of each infusion. All intravenous infusions were administered with the subject in the seated position. Subjects were observed for Adverse Events (Aes) and changes in vital signs (blood pressure, heart-rate, body temperature) from the start of each infusion (pre-dose, 15, 30, 45, 60, 75, 90, 120 minutes) and for a minimum of 2 hours after the start of the infusion. The observation period could be extended, if clinically indicated.
  • Adverse Events Aes
  • vital signs blood pressure, heart-rate, body temperature
  • Systemic (non-topical) corticosteroids Provided that the dose was stable and the total dose was less than or equal to the equivalent of prednisone 10 mg/day. Intra-articular injections were to be avoided; however, if necessary, up to two intra-articular injections were permitted. NOTE: A joint that received an intra-articular injection was counted as “active” in ALL subsequent assessments/evaluations.
  • NSAIDs including ASA: Provided the dose was stable
  • Acetaminophen, combination products including acetaminophen and narcotic analgesics i.e., acetaminophen with codeine phosphate, acetaminophen with propoxyphene napsylate, acetaminophen with oxycodone hydrochloride, acetaminophen with oxycodone bitartrate, etc.
  • narcotic analgesics i.e., acetaminophen with codeine phosphate, acetaminophen with propoxyphene napsylate, acetaminophen with oxycodone hydrochloride, acetaminophen with oxycodone bitartrate, etc.
  • tramadol For subjects experiencing pain not adequately controlled by baseline or study medication (except for 12 hours before a joint evaluation)
  • Table 1 is a schedule of study procedures and evaluations. TABLE 1 Schedule of Study Procedures and Evaluations Treatment Period Pretreatment (Day) Treatment Day e,f,j,h Screen Visit Day ( ⁇ 28 to ⁇ 2) ( ⁇ 2) 1 15 30 60 90 120 150 180 210 240 270 300 330 360 Screening assessments Informed consent X Complete History and Physical X X i CXR X a ECG X a X Stabilize/Withdraw prohibited X medications (if necessary) b Enroll Subject X X m Randomize Subject k X Dosing h X X X X X X X X X X X Interim Assessments f Duration of morning stiffness X X X X X X X X X X X X X X X X Tender joint count X X X X X X X X X X X X X X X Tender joint count X X X X
  • the ACR 20, ACR 50, and ACR 70 definition of response corresponds to a 20%, 50%, or 70% improvement, respectively, over baseline in tender and swollen joints (components 1 and 2) and a 20%, 50%, and 70% improvement, respectively, in three of the five remaining core data set measures (components 3 to 7).
  • a Major Clinical Response is defined as maintenance of an ACR 70 response over a continuous 6-month period. See Table 1 for the days that data for each component was collected.
  • ACR 20, ACR 50, and ACR 70 response rates on Day 360 were compared between each CTLA4Ig (BMS-188667) treatment group and placebo at the Dunnett-adjusted 0.027 (two-tailed) level of significance.
  • a Major Clinical Response was defined as the maintenance of an ACR 70 response over a continuous 6-month period. At the 12-month analysis, the proportion of subjects who achieved a Major Clinical Response among the three groups was summarized.
  • ACR-N The cumulative index, ACR-N was evaluated at each follow-up assessment, and the AUC was assessed for up to 6 months and up to 12 months.
  • the trapezoidal rule was used to compute the AUC.
  • the ACR-N AUC was compared between the two CTLA4Ig (BMS-188667) treatment groups and the placebo group using an analysis of variance (ANOVA) for 6- and 12-month data. This allowed for the assessment of subject response throughout the study.
  • Biomarkers were also used to assess the efficacy of the CTLA4Ig+MTX or placebo+MTX treatment regimens.
  • Potential biomarkers for immunomodulation or inflammation in RA include CRP, soluble IL-2r, RF, soluble ICAM-1, E-selectin, serum IL-6, and TNF ⁇ . These parameters were summarized by treatment group, using frequencies and mean change from baseline to Day 180 and Day 360.
  • An Adverse Event was defined as any new or worsening illness, sign symptom or clinically significant laboratory test abnormality noted by the Investigator during the course of the study, regardless of causality.
  • a serious adverse event was defined as an AE that met any of the following criteria: was fatal; was life-threatening; resulted in or prolonged hospitalization; resulted in persistent or significant disability or incapacity, was cancer, was a congenital anomaly/birth defect, resulted in an overdose, resulted in the development of drug dependency or drug abuse, or was an important medical event.
  • Vital sign measurements were obtained at screening and at each study visit during and following study drug administration. Vital sign measurements (seated blood pressure, heart rate, and body temperature) were summarized by treatment group using means.
  • a Kaplan-Meier plot of the cumulative proportion of subjects who discontinued for any reason during the first 12 months is presented in FIG. 69; the cumulative proportion of subjects who discontinued due to lack of efficacy in presented in FIG. 70. Note that in both graphs after approximately 30 days of therapy, discontinuation rates with placebo were consistently higher compared with both CTLA4Ig (BMS-188667) groups. Additionally, after approximately 150 days of therapy, discontinuation rates for 2 mg/kg CTLA4Ig group were higher than those for 10 mg/kg.
  • the 10 mg/kg CTLA4Ig group had the longest mean duration of exposure for both study phases and the placebo group had the shortest mean duration of exposure for both study phases (Day 180: 163 days, 156 days, 140 days; Day 360: 286 days, 268 days, and 234 days; 10 mg/kg, 2 mg/kg, and placebo, respectively).
  • Subjects were to have been treated with a “stable” dose of MTX (10-30 mg weekly) for at least 6 months, for 28 days prior to Day 1. With the exception of 4 subjects, all subjects received between 10 and 30 mg of MTX weekly in addition to CTLA4Ig (BMS-188667) during the primary phase (Day 1-180). During the secondary phase (Day 181-360), the dose of MTX could have been adjusted provided it remained between 10 and 30 mg
  • CTLA4Ig (BMS-188667) 10 mg/kg group had superior efficacy compared to the placebo group at Day 180 and Day 360.
  • results for some efficacy parameters were significantly better compared to the placebo group, results for most other efficacy parameters were numerically higher compared to placebo.
  • the ACR50 and ACR70 responses at Day 180 for the 10 mg/kg CTLA4Ig group were also significantly higher compared to the placebo group (Table 12, FIG. 71A and FIG. 71B).
  • the ACR50 and the ACR70 responses at Day 180 for the 2 mg/kg CTLA4Ig group were significantly higher compared to the placebo group.
  • the ACR20 response at Day 180 for the 2 mg/kg CTLA4Ig group was slightly higher compared to the placebo group; however, no statistically significant differences were observed.
  • Major Clinical Response was defined as maintenance of an ACR 70 response over a continuous 6-month period.
  • ACR-N numeric ACR
  • mean ACR-N increased slightly with 10 mg/kg CTLA4Ig, but remained relatively unchanged with 2 mg/kg CTLA4Ig and placebo.
  • the ACR-N was consistently higher for the 10 mg/kg CTLA4Ig group compared to the 2 mg/kg CTLA4Ig and placebo groups.
  • CTLA4Ig BMS-188667
  • CTLA4Ig BMS-188667
  • SF-36 summary scores range from 0 to 100 with higher scores indicating a better quality of life.
  • Analyses were performed on the LOCF (last observation carried forward) data set as well as the as the observed data set.
  • Results of the Health Outcomes at Day 360 were similar to those seen at Day 180.
  • For the 10 mg/kg CTLA4Ig group statistically significant improvements from baseline compared to the placebo group were observed in all four mental and all four physical domains of the SF-36 at Day 360, using the LOCF analysis (i.e., 95% CIs did not include 0) (FIGS. 80 A, and 80 B).
  • For the 2 mg/kg CTLA4Ig group a statistically significant difference in three of four physical domains at Day 360 and one of four mental domains at Day 360 compared to the placebo group was observed.
  • a comparison of the postbaseline means for the biomarker/PD parameters at Day 180 to those at Day 360 reveals important trends.
  • all biomarkers/PD measures continued to decrease, with the exception of soluble IL-2r which increased slightly.
  • mean values for 3 of the biomarkers/PD parameters either decreased slightly (ICAM-1, serum IL-6) or remained relatively constant (E-selectin) and mean values for the other 3 biomarkers/PD measures increased slightly (soluble IL-2r, RF, TNF ⁇ ).
  • mean values for all of the biomarkers/PD parameters increased slightly at Day 360, with the exception of TNF ⁇ which remained relatively unchanged.
  • CTLA4Ig (BMS-188667) administered at 10 mg/kg (+MTX) had superior efficacy compared to placebo (+MTX) at Day 180 and Day 360.
  • administration of 10 mg/kg CTLA4Ig was significantly better than placebo:
  • the 10 mg/kg CTLA4Ig group had a lower number of new active joints and a higher number of subjects reporting no new active tender and swollen joints compared with the placebo group at Day 180 and at Day 360.

Abstract

The present invention relates to compositions and methods for treating immune system diseases such as rheumatic disease, by administering to a subject soluble CTLA4 molecules that block endogenous B7 molecules from binding their ligands, alone, or in conjunction with other agents including Disease Modifying Anti-Rheumatic Drugs (DMARDs).

Description

  • This application is a continuation-in-part (CIP) of U.S. Ser. No. 09/898,195, filed Jul. 2, 2001, and claims the priorities of provisional applications, U.S. Serial No. 60/215,913, filed Jul. 3, 2000, U.S. Serial No. 60/373,852, filed Apr. 19, 2002 and U.S. Serial No. 60/407,246, filed Aug. 30, 2002, the contents of which are hereby incorporated by reference in their entirety into this application. [0001]
  • Throughout this application various publications are referenced. The disclosures of these publications in their entireties are hereby incorporated by reference into this application in order to more fully describe the state of the art to which this invention pertains.[0002]
  • FIELD OF THE INVENTION
  • The present invention relates generally to the field of immune system diseases, e.g., rheumatic diseases. In particular, the invention relates to methods and compositions for treating immune system diseases, e.g., rheumatic diseases, such as rheumatoid arthritis, by administering to a subject an effective amount of soluble CTLA4 molecules alone, or in conjunction with a Disease Modifying Anti-Rheumatic Drug (DMARD). [0003]
  • BACKGROUND OF THE INVENTION
  • No cure currently exists for rheumatic diseases. Rather, therapeutic agents are used to treat the symptoms. Typically, the therapeutic agents are administered over long periods of time and the therapeutic value is often diminished by adverse side effects. [0004]
  • Rheumatic diseases encompass a group of diseases that affect the musculo-skeletal and connective tissues of the body. These diseases are characterized by chronic inflammation that often leads to permanent tissue damage, deformity, atrophy and disability. Rheumatic diseases affect the joints, bone, soft tissue, or spinal cord (Mathies, H. 1983 Rheuma) and are classified as inflammatory rheumatism, degenerative rheumatism, extra-articular rheumatism, or collagen diseases. Some rheumatic diseases are known to be autoimmune diseases caused by a subject's altered immune response. [0005]
  • Rheumatoid arthritis is a progressive rheumatic disease, affecting approximately 2% of the adult population of developed countries (Utsinger, P. D., et al., 1985 [0006] Rheumatoid Arthritis, p. 140). This disease is characterized by persistent inflammatory synovitis that causes destruction of cartilage and bone erosion, leading to structural deformities in the peripheral joints. The symptoms associated with rheumatoid arthritis include joint swelling, joint tenderness, inflammation, morning stiffness, and pain, especially upon flexing. Subjects having advanced stages of arthritis suffer from structural damage, including joint destruction with bone erosion (in: “Principals of Internal Medicine, Harrison, 13th edition, pages 1648-1655). In addition, patients can present other clinical symptoms of various organic lesions, including lesions of the skin, kidney, heart, lung, central nervous system, and eyes due to vasculitis related to the autoimmune process.
  • Other symptoms that correlate with rheumatoid arthritis include elevated erythrocyte sedimentation rates, and elevated levels of serum C-reactive protein (CRP) and/or soluble IL-2 receptor (IL-2r). The erythrocyte sedimentation rate is increased in nearly all patients with active rheumatoid arthritis. The level of serum C-reactive protein is also elevated and correlates with disease activity and the likelihood of progressive joint damage. Additionally, the level of soluble IL-2r, a product of activated T-cells, is elevated in blood serum and synovial fluid of patients with active rheumatoid arthritis (see: “Principals of Internal Medicine, Harrison, 13[0007] th edition, page 1650).
  • Rheumatoid arthritis is believed to be a T-cell-mediated autoimmune disease, involving antigen-nonspecific intercellular interactions between T-lymphocytes and antigen-presenting cells. In general, the magnitude of the T-cell response is determined by the co-stimulatory response elicited by the interaction between T-cell surface molecules and their ligands (Mueller, et al., 1989 [0008] Ann. Rev. Immunol. 7:445-480). Key co-stimulatory signals are provided by the interaction between T-cell surface receptors, CD28 and CTLA4, and their ligands, such as B7-related molecules CD80 (i.e., B7-1) and CD86 (i.e., B7-2), on antigen presenting cells (Linsley, P. and Ledbetter, J. 1993 Ann. Rev. Immunol. 11:191-212).
  • T-cell activation in the absence of co-stimulation results in anergic T-cell response (Schwartz, R. H., 1992 [0009] Cell 71:1065-1068) wherein the immune system becomes nonresponsive to stimulation.
  • Since rheumatoid arthritis is thought to be a T-cell-mediated immune system disease, one strategy to develop new agents to treat rheumatoid arthritis is to identify molecules that block co-stimulatory signals between T-lymphocytes and antigen presenting cells, by blocking the interaction between endogenous CD28 or CTLA4 and B7. Potential molecules include soluble CTLA4 molecules that are modified (i.e. CTLA4 mutant molecules) to bind to B7 with higher avidity than wildtype CTLA4 (the sequence of which is shown in FIG. 23) or CD28, thereby blocking the co-stimulatory signals. [0010]
  • Soluble forms of CD28 and CTLA4 have been constructed by fusing variable (V)-like extracellular domains of CD28 and CTLA4 to immunoglobulin (Ig) constant domains resulting in CD28Ig and CTLA4Ig. A nucleotide and amino acid sequence of CTLA4Ig is shown in FIG. 24 with the protein beginning with methionine at position +1 or alanine at position −1 and ending with lysine at position +357. CTLA4Ig binds both CD80-positive and CD86-positive cells more strongly than CD28Ig (Linsley, P., et al., 1994 [0011] Immunity 1:793-80). Many T-cell-dependent immune responses have been found to be blocked by CTLA4Ig both in vitro and in vivo. (Linsley, P., et al., 1991b, supra; Linsley, P., et al., 1992a Science 257:792-795; Linsley, P., et al., 1992b J. Exp. Med. 176:1595-1604; Lenschow, D. J., et al. 1992 Science 257:789-792; Tan, P., et al., 1992 J. Exp. Med. 177:165-173; Turka, L. A., 1992 Proc. Natl. Acad. Sci. USA 89:11102-11105).
  • To alter binding affinity to natural ligands, such as B7, soluble CTLA4Ig fusion molecules were modified by mutation of amino acids in the CTLA4 portion of the molecules. Regions of CTLA4 that, when mutated, alter the binding affinity or avidity for B7 ligands include the complementarity determining region 1 (CDR-1 as described in U.S. Pat. Nos. 6,090,914, 5,773,253, 5,844,095; in copending U.S. Patent Application Serial No. 60/214,065; and by Peach et al, 1994. J. Exp. Med., 180:2049-2058) and complementarity determining region 3 (CDR-3)-like regions (CDR-3 is the conserved region of the CTLA4 extracellular domain as described in U.S. Pat. Nos. 6,090,914, 5,773,253 and 5,844,095; in copending U.S. Patent Application Serial No. 60/214,065; and by Peach, R. J., et al [0012] J Exp Med 1994 180:2049-2058; the CDR-3-like region encompasses the CDR-3 region and extends, by several amino acids, upstream and/or downstream of the CDR-3 motif). The CDR-3-like region includes a hexapeptide motif MYPPPY (SEQ ID NO.: 20) that is highly conserved in all CD28 and CTLA4 family members. Alanine scanning mutagenesis through the hexapeptide motif in CTLA4, and at selected residues in CD28Ig, reduced or abolished binding to CD80 (Peach, R. J., et al J Exp Med 1994 180:2049-2058; U.S. Pat. No. 5,434,131; U.S. Pat. No. 6,090,914; U.S. Pat. No. 5,773,253.
  • Further modifications were made to soluble CTLA4Ig molecules by interchanging homologous regions of CTLA4 and CD28. These chimeric CTLA4/CD28 homologue mutant molecules identified the MYPPPY hexapeptide motif common to CTLA4 and CD28, as well as certain non-conserved amino acid residues in the CDR-1- and CDR-3-like regions of CTLA4, as regions responsible for increasing the binding avidity of CTLA4 with CD80 (Peach, R. J., et al., 1994 [0013] J Exp Med 180:2049-2058).
  • Soluble CTLA4 molecules, such as CTLA4Ig, CTLA4 mutant molecules or chimeric CTLA4/CD28 homologue mutants as described supra, introduce a new group of therapeutic drugs to treat rheumatic diseases. [0014]
  • Present treatments for rheumatic diseases, such as rheumatoid arthritis, include administering nonspecific cytotoxic immunosuppressive drugs known as Disease Modifying Anti-Rheumatic Drugs (DMARDs), such as methotrexate, infliximab, cyclophosphamide, azathioprine, cyclosporin A, sulfasalazine, hydroxychloroquine, leflunomide, etanercept, and tumor necrosis factor-alpha (TNFα) or other cytokine blockers or antagonists. These immunosuppressive drugs suppress the entire immune system of the subject, and long-term use increases the risk of infection and oncogenesis. Moreover, these drugs merely slow down the progress of the rheumatoid arthritis, which resumes at an accelerated pace after the therapy is discontinued. Additionally, prolonged therapy with these nonspecific drugs produces toxic side effects, including a tendency towards development of certain malignancies, kidney failure, bone marrow suppression, pulmonary fibrosis, malignancy, diabetes, and liver function disorders. These drugs may also gradually cease being effective after about 2-5 years (Kelley's Textbook of Rheumatology, 6[0015] th Edition, pages 1001-1022). Newer, biologically based, DMARDs such as cytokine blockers may be more potent and may have longer lasting effects than older DMARDS such as hydrochloroquine, however, the long term safety of these newer drugs is still unknown. Reports of multiple sclerosis and lupus exist with the use of TNF blockers.
  • Alternatively, therapeutic agents that are non-specific immunosuppressive and anti-inflammatory drugs have been used to obtain symptomatic relief. These drugs are dose-dependent and do not protect from disease progression. These drugs include Non-Steroidal Anti-Inflammatory Drugs (NSAIDS) as well as steroid compounds (e.g., corticosteroids or glucocorticoids), such as prednisone and methylprednisolone. Steroids also have significant toxic side effects associated with their long-term use. (Kelley's Textbook of Rheumatology, 6[0016] th Edition, pages 829-833).
  • Thus, current treatments for rheumatoid arthritis are of limited efficacy, involve significant toxic side effects, and cannot be used continuously for prolonged periods of time. [0017]
  • Accordingly, there exists a need for treatments that are effective and more potent for treating rheumatic diseases, such as rheumatoid arthritis, and avoids the disadvantages of old conventional methods and agents, by targeting a pathophysiological mechanism of auto-immunity. [0018]
  • SUMMARY OF THE INVENTION
  • The present invention provides compositions and methods for treating immune system diseases, by administering to a subject soluble CTLA4 molecules, which bind to B7 molecules on B7-positive cells, thereby inhibiting endogenous B7 molecules from binding CTLA4 and/or CD28 on T-cells. Soluble CTLA4 molecules used in the methods of the invention include CTLA4Ig and soluble CTLA4 mutant molecule L104EA29YIg. [0019]
  • Alternatively, the present invention provides compositions and methods for treating immune system diseases, by administering to a subject a combination of a DMARD and a molecule that blocks B7 interaction with CTLA4 and/or CD28. [0020]
  • The present invention also provides methods for inhibiting T-cell function, but not causing T-cell depletion, in a human by contacting B7-positive cells in the human with soluble CTLA4. Examples of soluble CTLA4 include CTLA4Ig and soluble CTLA4 mutant molecules, such as L104EA29YIg. [0021]
  • The present invention also provides methods for treating (e.g. reducing symptoms of) rheumatic diseases, such as rheumatoid arthritis, by administering to a subject suffering from symptoms of arthritis, soluble CTLA4 molecules such as CTLA4Ig and/or soluble CTLA4 mutant molecule L104EA29YIg and/or a mix of any soluble CTLA molecule. The CTLA4 mutant molecule L104EA29YIg e.g. beginning with methionine at position +1 or alanine at position −1 and ending with lysine at position +357, as shown in FIG. 19, is preferred for use in the methods of the invention. [0022]
  • The present invention also provides methods for treating (e.g. reducing symptoms of) rheumatic diseases, such as rheumatoid arthritis, by administering to the subject a combination of 1) a DMARD, such as methotrexate or a molecule that blocks TNF interactions, and 2) soluble CTLA4 molecules, such as CTLA4Ig. [0023]
  • The present invention also provides methods for reducing pathophysiological changes associated with an immune system disease (e.g., rheumatic disease), such as structural damage, by administering to the subject diagnosed with the immune system disease (e.g., rheumatoid arthritis), soluble CTLA4 molecules alone or in conjunction with other therapeutic drugs, such as a DMARD. [0024]
  • The present invention also provides a pharmaceutical composition for treating immune system diseases, such as rheumatic diseases, comprising a pharmaceutically acceptable carrier and a biologically effective agent, such as soluble CTLA4 molecules, alone or in conjunction with other therapeutic drugs, such as a DMARD, a NSAID, a corticosteroid and/or a glucocorticoid. [0025]
  • Kits comprising pharmaceutical compositions therapeutic for immune system disease are also encompassed by the invention. In one embodiment, a kit comprising one or more of the pharmaceutical compositions of the invention is used to treat an immune system disease, e.g. rheumatoid arthritis. For example, the pharmaceutical composition comprises an effective amount of soluble CTLA4 molecules that bind to B7 molecules on B7-positive cells, thereby blocking the B7 molecules from binding CTLA4 and/or CD28 on T-cells. Further, the kit may contain one or more immunosuppressive agents used in conjunction with the pharmaceutical compositions of the invention. Potential immunosuppressive agents include, but are not limited to, corticosteroids, nonsteroidal antiinflammatory drugs (e.g. Cox-2 inhibitors), prednisone, cyclosporine, cyclosporin A, azathioprine, methotrexate, TNFα blockers or antagonists, hydroxychloroquine, sulphasalazopyrine (sulfasalazine), gold salts, infliximab, etanercept, anakinra and any biological agent targeting an inflammatory cytokine. [0026]
  • The present invention also provides methods for reducing the erythrocyte sedimentation rate that is associated with rheumatoid arthritis. [0027]
  • Additionally, the present invention provides methods for reducing the levels of certain components of blood serum which are associated with rheumatoid arthritis, including C-reactive protein, IL-6, TNF-α, soluble ICAM-1, soluble E-selectin and/or soluble IL-2r.[0028]
  • BRIEF DESCRIPTION OF THE FIGURES
  • FIG. 1A: Demographic data of patient cohorts. Demographic data including gender, race, and disease duration as described in Example 3, infra. [0029]
  • FIG. 1B: Demographic data of patient cohorts. Demographic data including gender, age, weight, and disease activity, evaluated by the patient and by the physician, as described in Example 3, infra. [0030]
  • FIG. 1C: Demographic data of patient cohorts as described in Example 3, infra. Demographic data including disease activity, erythrocyte sedimentation rate (ESR), physical function (disability evaluated by health questionnaire), and C-reactive protein (CRP). [0031]
  • FIG. 1D: Demographic data of patient cohorts as described in Example 3, infra. Demographic data including joint swelling, joint tenderness, morning stiffness, and pain. [0032]
  • FIG. 1E: Demographic data of patient cohorts as described in Example 3, infra. Demographic data including prior treatments. [0033]
  • FIG. 2: Summary of discontinuations at [0034] day 85 by reason as described in Example 3, infra.
  • FIG. 3A: ACR responses at [0035] Day 85 as described in Example 3, infra: ACR-20, −50, and −70 responses.
  • FIG. 3B: ACR-20 responses at [0036] Day 85, including placebo response, as described in Example 3, infra: ACR-20 response with 95% confidence limits.
  • FIG. 3C: ACR-20 responses at [0037] Day 85 as described in Example 3, infra: Difference in ACR-20 response with respect to 95% confidence intervals.
  • FIG. 4A: Basic (20% improvement) clinical responses in swollen and tender joint count in percentage of patients at [0038] Day 85 as described in Example 3, infra: basic clinical response, ACR-20.
  • FIG. 4B: Clinical responses (in percentage improvement) in swollen and tender joint count in percentage of patients at [0039] Day 85 as described in Example 3, infra: change in clinical response in percentage improvement.
  • FIG. 5A: Pain response (by Likert scale by mean unit change from baseline) in percentage of patients at [0040] Day 85 as described in Example 3, infra: pain score changes from baseline.
  • FIG. 5B: Patient global disease changes (by Likert scale by mean unit change from baseline) in percentage of patients at [0041] Day 85 as described in Example 3, infra: patient global disease activity changes.
  • FIG. 5C: Physician global disease changes (by Likert scale by mean unit change from baseline) in percentage of patients at [0042] Day 85 as described in Example 3, infra: physician global disease activity changes.
  • FIG. 5D: Pain (by Likert scale by mean unit change from baseline) in percentage of patients at [0043] Day 85 as described in Example 3, infra: pain changes from baseline.
  • FIG. 6A: Patient global assessment of disease activity change from baseline by range of 2 units at [0044] Day 85 as described in Example 3, infra; disease activity improvement.
  • FIG. 6B: Physician global assessment of disease activity change from baseline by range of 2 units at [0045] Day 85 as described in Example 3, infra; disease activity improvement.
  • FIG. 7A: Percent reduction in C-reactive protein (CRP) levels at [0046] Day 85 as described in Example 3, infra: percentage reduction in CRP levels from baseline.
  • FIG. 7B: Difference in reduction in C-reactive protein (CRP) levels at [0047] Day 85 as described in Example 3, infra: percent reduction difference in CRP levels with 95% confidence intervals.
  • FIG. 7C: Mean reduction in C-reactive protein (CRP) levels at [0048] Day 85 as described in Example 3, infra: mean change from baseline.
  • FIG. 8: Reduction in soluble IL-2 receptor levels mean change from baseline at [0049] Day 85 as described in Example 3, infra.
  • FIG. 9A: The effect of CTLA4Ig on tender joints over time as described in Example 3, infra: median difference from baseline. [0050]
  • FIG. 9B: The effect of CTLA4Ig on tender joints over time as described in Example 3, infra: mean difference from baseline. [0051]
  • FIG. 10A: The effect of CTLA4Ig on swollen joints over time as described in Example 3, infra: median difference from baseline. [0052]
  • FIG. 10B: The effect of CTLA4Ig on swollen joints over time as described in Example 3, infra: mean difference from baseline. [0053]
  • FIG. 11: The effect of CTLA4Ig on pain assessment mean difference from baseline over time as described in Example 3, infra. [0054]
  • FIG. 12A: The effect of CTLA4Ig on patient assessment of disease activity mean difference from baseline over time as described in Example 3, infra. [0055]
  • FIG. 12B: The effect of CTLA4Ig on physician assessment of disease activity mean difference from baseline over time as described in Example 3, infra. [0056]
  • FIG. 13A: The effect of L104EA29YIg on tender joints over time as described in Example 3, infra: median difference from baseline. [0057]
  • FIG. 13B: The effect of L104EA29YIg on tender joints over time as described in Example 3, infra: mean change from baseline. [0058]
  • FIG. 14A: The effect of L104EA29YIg on swollen joints over time as described in Example 3, infra: median difference from baseline. [0059]
  • FIG. 14B: The effect of L104EA29YIg on swollen joints over time as described in Example 3, infra: mean change from baseline. [0060]
  • FIG. 15: The effect of L104EA29YIg on pain assessment over time as described in Example 3, infra: mean change from baseline over time. [0061]
  • FIG. 16A: The effect of L104EA29YIg on patient assessment of disease activity mean difference from baseline over time as described in Example 3, infra. [0062]
  • FIG. 16B: The effect of L104EA29YIg on physician assessment of disease activity mean difference from baseline over time as described in Example 3, infra. [0063]
  • FIG. 17: Percent improvement in patient disability assessed by Health Assessment Questionnaire (HAQ) compared to the baseline at [0064] Day 85 with CTLA4Ig and L104EA29YIg treatment as described in Example 3, infra.
  • FIG. 18: Nucleotide and amino acid sequence of L104EIg (SEQ ID NOs: 6-7) as described in Example 1, infra. [0065]
  • FIG. 19: Nucleotide and amino acid sequence of L104EA29YIg (SEQ ID NOs: 8-9) as described in Example 1, infra. [0066]
  • FIG. 20: Nucleotide and amino acid sequence of L104EA29LIg (SEQ ID NOs: 10-11) as described in Example 1, infra. [0067]
  • FIG. 21: Nucleotide and amino acid sequence of L104EA29TIg (SEQ ID NOs: 12-13) as described in Example 1, infra. [0068]
  • FIG. 22: Nucleotide and amino acid sequence of L104EA29WIg (SEQ ID NOs: 14-15) as described in Example 1, infra. [0069]
  • FIG. 23: Nucleotide and amino acid sequence of CTLA4 receptor (SEQ ID NOs: 16-17). [0070]
  • FIG. 24: Nucleotide and amino acid sequence of CTLA4Ig (SEQ ID NOs: 18-19). FIG. 25: SDS gel (FIG. 25A) for CTLA4Ig (lane 1), L104EIg (lane 2), and L104EA29YIg (lane 3A); and size exclusion chromatographs of CTLA4Ig (FIG. 25B) and L104EA29YIg (FIG. 25C). [0071]
  • FIG. 26 (left and right depictions): A ribbon diagram of the CTLA4 extracellular Ig V-like fold generated from the solution structure determined by NMR spectroscopy. FIG. 26 (right depiction) shows an expanded view of the CDR-1 (S25-R33) region and the MYPPPY region indicating the location and side-chain orientation of the avidity enhancing mutations, L104 and A29. [0072]
  • FIGS. [0073] 27A & 27B: FACS assays showing binding of L104EA29YIg, L104EIg, and CTLA4Ig to human CD80- or CD86-transfected CHO cells as described in Example 2, infra.
  • FIGS. [0074] 28A & 28B: Graphs showing inhibition of proliferation of CD80-positive and CD86-positive CHO cells as described in Example 2, infra.
  • FIGS. [0075] 29A & 29B: Graphs showing that L104EA29YIg is more effective than CTLA4Ig at inhibiting proliferation of primary and secondary allostimulated T cells as described in Example 2, infra.
  • FIGS. [0076] 30A-C: Graphs illustrating that L104EA29YIg is more effective than CTLA4Ig at inhibiting IL-2 (FIG. 30A), IL-4 (FIG. 30B), and gamma (γ)-interferon (FIG. 30C) cytokine production of allostimulated human T cells as described in Example 2, infra.
  • FIG. 31: A graph demonstrating that L104EA29YIg is more effective than CTLA4Ig at inhibiting proliferation of phytohemaglutinin-(PHA) stimulated monkey T cells as described in Example 2, infra. [0077]
  • FIG. 32: A graph showing the equilibrium binding analysis of L104EA29YIg, L104EIg, and wild-type CTLA4Ig to CD86Ig. [0078]
  • FIGS. [0079] 33A & B: Reduction in soluble ICAM-1 and soluble E-selectin levels mean change from baseline at Day 85 as described in Example 3, infra.
  • FIG. 34: A graph showing the summary of ACR20 response by visit day in response to methotrexate and CTLA4Ig (2 and 10 mg/kg) therapy, as described in Example 5, infra. [0080]
  • FIG. 35: A graph showing the summary of ACR50 response by visit day in response to methotrexate alone or methotrexate and CTLA4Ig (2 and 10 mg/kg) therapy, as described in Example 5, infra. [0081]
  • FIG. 36: A graph showing the summary of ACR70 response by visit day in response to methotrexate alone or methotrexate and CTLA4Ig (2 and 10 mg/kg) therapy, as described in Example 5, infra. [0082]
  • FIG. 37: A graph showing the mean ACR-N over time in response to methotrexate alone or methotrexate and CTLA4Ig (2 and 10 mg/kg) therapy, as described in Example 5, infra. [0083]
  • FIG. 38: A bar graph showing the ACR response in response to methotrexate alone or methotrexate and CTLA4Ig (2 and 10 mg/kg) therapy on [0084] day 180 with a 95% confidence interval, as described in Example 5, infra.
  • FIG. 39: A bar graph showing the proportion of New Active Joints in response to methotrexate alone or methotrexate and CTLA4Ig (2 and 10 mg/kg) therapy on [0085] day 180, as described in Example 5, infra.
  • FIG. 40: A bar graph showing ACR response after therapy with methotrexate alone or methotrexate and CTLA4Ig (2 and 10 mg/kg) on [0086] day 180, as described in Example 5, infra.
  • FIG. 41: A graph showing percent improvement in tender joints after therapy with methotrexate alone or methotrexate and CTLA4Ig (2 and 10 mg/kg)—mean percent improvement from baseline, as described in Example 5, infra. [0087]
  • FIG. 42: A graph showing percent improvement in swollen joints after therapy with methotrexate alone or methotrexate and CTLA4Ig (2 and 10 mg/kg)—mean percent improvement from baseline, as described in Example 5, infra. [0088]
  • FIG. 43: A graph showing percent improvement in pain after therapy with methotrexate alone or methotrexate and CTLA4Ig (2 and 10 mg/kg)—mean percent improvement from baseline, as described in Example 5, infra. [0089]
  • FIG. 44: A graph showing percent improvement in regard to disease activity as reported by the subject after therapy with methotrexate alone or methotrexate and CTLA4Ig (2 and 10 mg/kg)—mean percent improvement from baseline, as described in Example 5, infra. [0090]
  • FIG. 45: A graph showing percent improvement in regard to disease activity as reported by the physician after therapy with methotrexate alone or methotrexate and CTLA4Ig (2 and 10 mg/kg)—mean percent improvement from baseline, as described in Example 5, infra. [0091]
  • FIG. 46: A graph showing percent improvement regarding physical function after therapy with methotrexate alone or methotrexate and CTLA4Ig (2 and 10 mg/kg)—mean percent improvement from baseline as measured by HAQ, as described in Example 5, infra. [0092]
  • FIG. 47: A graph showing percent improvement in CRP levels function after therapy with methotrexate alone or methotrexate and CTLA4Ig (2 and 10 mg/kg)—mean percent improvement from baseline, as described in Example 5, infra. [0093]
  • FIG. 48: A graph showing percent improvement in CRP levels function after therapy with methotrexate alone or methotrexate and CTLA4Ig (2 and 10 mg/kg)—median percent improvement from baseline, as described in Example 5, infra. [0094]
  • FIG. 49: A graph showing the difference in ACR response rate on [0095] day 180 in two groups after therapy with CTLA4Ig (2 and 10 mg/kg) in comparison to a group treated with methotrexate (MTX) only (95% Confidence Limits), as described in Example 5, infra.
  • FIG. 50: A graph showing the change from baseline for SF-36 Physical Health Component on [0096] day 180, in two groups after therapy with CTLA4Ig (2 and 10 mg/kg) compared to a group treated with methotrexate only (95% Confidence Limits), as described in Example 5, infra.
  • FIG. 51: A graph showing the change from baseline for SF-36 Mental Health Component on [0097] Day 180, in two groups after therapy with CTLA4Ig (2 and 10 mg/kg) compared to a group treated with methotrexate only (95% Confidence Limits), as described in Example 5, infra.
  • FIG. 52: A bar graph showing CRP levels at [0098] day 180 after therapy with methotrexate alone or methotrexate and CTLA4Ig (2 and 10 mg/kg), as described in Example 5, infra.
  • FIG. 53: A bar graph showing Rheumatoid Factor levels on [0099] day 180 after therapy with methotrexate alone or methotrexate and CTLA4Ig (2 and 10 mg/kg), as described in Example 5, infra.
  • FIG. 54: A bar graph showing IL-2r levels on [0100] day 180 after therapy with methotrexate alone or methotrexate and CTLA4Ig (2 and 10 mg/kg), as described in Example 5, infra.
  • FIG. 55: A bar graph showing IL-6 levels on [0101] day 180 after therapy with methotrexate alone or methotrexate and CTLA4Ig (2 and 10 mg/kg), as described in Example 5, infra.
  • FIG. 56: A bar graph showing TNFα levels on [0102] day 180 after therapy with methotrexate alone or methotrexate and CTLA4Ig (2 and 10 mg/kg), as described in Example 5, infra.
  • FIG. 57: A table of the univariate methotrexate dose at screening/enrollment for treatment group BMS 10-treated with CTLA4Ig at 10 mg/kg body weight as described in Example 5, infra. [0103]
  • FIG. 58: A table of the univariate methotrexate dose at screening/enrollment for treatment group BMS 2-treated with CTLA4Ig at 2 mg/kg body weight as described in Example 5, infra. [0104]
  • FIG. 59: A table of the univariate methotrexate dose at screening/enrollment for the placebo group, as described in Example 5, infra. [0105]
  • FIG. 60: A table of the univariate methotrexate dose up to and including [0106] day 180 of the study for treatment group BMS 10-treated with CTLA4Ig at 10 mg/kg body weight as described in Example 5, infra.
  • FIG. 61: A table of the univariate methotrexate dose up to and including [0107] day 180 of the study for treatment group BMS 2-treated with CTLA4Ig at 2 mg/kg body weight as described in Example 5, infra.
  • FIG. 62: A table of the univariate methotrexate dose up to and including [0108] day 180 of the study for the placebo group, as described in Example 5, infra.
  • FIG. 63: A bar graph showing the difference in modified ACR response rates on [0109] day 180 in two groups after therapy with etanercept alone (25 mg twice weekly) or in combination with CTLA4Ig (2 mg/kg), as described in Example 6, infra.
  • FIGS. [0110] 64A-C: Graphs showing percentage improvement of individual components of the modified ACR criteria as assessed on each visit day after therapy with etanercept alone (25 mg twice weekly) or in combination with CTLA4Ig (2 mg/kg) as described in Example 6, infra. A. Tender Joint Count. B. Swollen Joint Count. C. Pain Assessment.
  • FIG. 65: A. A graph showing the change from baseline for SF-36 Physical Health Component on [0111] day 180, in two groups after therapy with etanercept (25 mg biweekly) alone or in combination with CTLA4Ig (2 mg/kg) (95% Confidence Limits), as described in Example 6, infra. B. A graph showing the change from baseline for SF-36 Mental Health Component on day 180, in two groups after therapy with etanercept (25 mg biweekly) alone or in combination with CTLA4Ig (2 mg/kg) (95% Confidence Limits), as described in Example 6, infra.
  • FIG. 66: Nucleotide sequence of a CTLA4Ig encoding a signal peptide; a wild type amino acid sequence of the extracellular domain of CTLA4 starting at methionine at position +1 to aspartic acid at position +124, or starting at alanine at position −1 to aspartic acid at position +124; and an Ig region (SEQ ID NO.: 21). [0112]
  • FIG. 67: Amino acid sequence of a CTLA4Ig having a signal peptide; a wild type amino acid sequence of the extracellular domain of CTLA4 starting at methionine at position +1 to aspartic acid at position +124, or starting at alanine at position −1 to aspartic acid at position +124; and an Ig region (SEQ ID NO.: 22). [0113]
  • FIG. 68: A schematic diagram showing the disposition of subjects into three cohorts as described in Example 7, infra. [0114]
  • FIG. 69: A Kaplan-Meier plot of the cumulative proportion of subjects who discontinued for any reason during the first 12 months of the study, as described in Example 7, infra. [0115]
  • FIG. 70: A Kaplan-Meier plot of the cumulative proportion of subjects who discontinued due to lack of efficacy during the first 12 months of study, as described in Example 7, infra. [0116]
  • FIG. 71A: A graph showing the ACR Responses on [0117] Day 180 for patients administered methotrexate alone or methotrexate and CTLA4Ig (2 or 10 mg/kg body weight) as described in Example 7, infra.
  • FIG. 71B: A graph showing the 95 Percent Confidence Intervals for Differences in ACR Responses on [0118] Day 180 for patients administered methotrexate alone or methotrexate and CTLA4Ig (2 or 10 mg/kg body weight) as described in Example 7, infra.
  • FIG. 72A: A graph showing the ACR Responses on [0119] Day 360 for patients administered methotrexate alone or methotrexate and CTLA4Ig (2 or 10 mg/kg body weight) as described in Example 7, infra.
  • FIG. 72B: A graph showing the 95 Percent Confidence Intervals for Differences in ACR Responses on [0120] Day 360 for patients administered methotrexate alone or methotrexate and CTLA4Ig (2 or 10 mg/kg body weight) as described in Example 7, infra.
  • FIG. 73A: A graph summarizing the [0121] ACR 20 Response by Visit during a one year interval for patients administered methotrexate alone or methotrexate and CTLA4Ig (2 or 10 mg/kg body weight) as described in Example 7, infra.
  • FIG. 73B: A graph summarizing the [0122] ACR 50 Response by Visit during a one year interval for patients administered methotrexate alone or methotrexate and CTLA4Ig (2 or 10 mg/kg body weight) as described in Example 7, infra.
  • FIG. 73C: A graph summarizing the [0123] ACR 70 Response by Visit during a one year interval for patients administered methotrexate alone or methotrexate and CTLA4Ig (2 or 10 mg/kg body weight) as described in Example 7, infra.
  • FIG. 74: A graph showing the Mean ACR-N over a one year time interval for patients administered methotrexate alone or methotrexate and CTLA4Ig (2 or 10 mg/kg body weight) as described in Example 7, infra. [0124]
  • FIG. 75: A graph showing the Proportion of New Active Joints at [0125] Day 180 for patients administered methotrexate alone or methotrexate and CTLA4Ig (2 or 10 mg/kg body weight) as described in Example 7, infra.
  • FIG. 76A: A graph showing the Frequency of New Tender Joints per Subject at [0126] Day 180 for patients administered methotrexate alone or methotrexate and CTLA4Ig (2 or 10 mg/kg body weight) as described in Example 7, infra.
  • FIG. 76B: A graph showing the Frequency of New Tender Joints per Subject at [0127] Day 360 for patients administered methotrexate alone or methotrexate and CTLA4Ig (2 or 10 mg/kg body weight) as described in Example 7, infra.
  • FIG. 77A: A graph showing the Frequency of New Swollen Joints per Subject at [0128] Day 180 for patients administered methotrexate alone or methotrexate and CTLA4Ig (2 or 10 mg/kg body weight) as described in Example 7, infra.
  • FIG. 77B: A graph showing the Frequency of New Swollen Joints per Subject at [0129] Day 360 for patients administered methotrexate alone or methotrexate and CTLA4Ig (2 or 10 mg/kg body weight) as described in Example 7, infra.
  • FIG. 78: A graph showing the Proportion of New Active Joints at [0130] Day 360 for patients administered methotrexate alone or methotrexate and CTLA4Ig (2 or 10 mg/kg body weight) as described in Example 7, infra.
  • FIG. 79: Graphs showing the: A) Change from Baseline in the Physical Health Domains on [0131] Day 180, and B) Change from Baseline in the Mental Health Domains on 180, for patients administered methotrexate alone or methotrexate and CTLA4Ig (2 or 10 mg/kg body weight) as described in Example 7, infra.
  • FIG. 80: Graphs showing the: A) Change from Baseline in the Physical Health Domains on [0132] Day 360, and B) Change from Baseline in the Mental Health Domains on Day 360, for patients administered methotrexate alone or methotrexate and CTLA4Ig (2 or 10 mg/kg body weight) as described in Example 7, infra.
  • FIG. 81: A graph showing the Soluble IL-2r Levels at Baseline, [0133] Days 180 and 360 for patients administered methotrexate alone or methotrexate and CTLA4Ig (2 or 10 mg/kg body weight) as described in Example 7, infra.
  • FIG. 82: A graph showing the Rheumatoid Factor Levels at Baseline, [0134] Days 180 and 360 for patients administered methotrexate alone or methotrexate and CTLA4Ig (2 or 10 mg/kg body weight) as described in Example 7, infra.
  • FIG. 83: A graph showing the ICAM-1 Levels at Baseline, [0135] Days 180 and 360 for patients administered methotrexate alone or methotrexate and CTLA4Ig (2 or 10 mg/kg body weight) as described in Example 7, infra.
  • FIG. 84: A graph showing the e-Selectin Levels at Baseline, [0136] Days 180 and 360 for patients administered methotrexate alone or methotrexate and CTLA4Ig (2 or 10 mg/kg body weight) as described in Example 7, infra.
  • FIG. 85: A graph showing the Serum IL-6 at Baseline, [0137] Days 180 and 360 for patients administered methotrexate alone or methotrexate and CTLA4Ig (2 or 10 mg/kg body weight) as described in Example 7, infra.
  • FIG. 86A: A graph showing the CRP Levels at Baseline, [0138] Days 180 and 360 for patients administered methotrexate alone or methotrexate and CTLA4Ig (2 or 10 mg/kg body weight) as described in Example 7, infra.
  • FIG. 86B: A graph showing the TNFα Levels at Baseline, [0139] Days 180 and 360 for patients administered methotrexate alone or methotrexate and CTLA4Ig (2 or 10 mg/kg body weight) as described in Example 7, infra.
  • DETAILED DESCRIPTION OF THE INVENTION
  • Definitions [0140]
  • All scientific and technical terms used in this application have meanings commonly used in the art unless otherwise specified. As used in this application, the following words or phrases have the meanings specified. [0141]
  • As used herein, DMARD refers to a Disease Modifying Anti-Rheumatic Drug. A DMARD is any agent that modifies the symptoms and/or progression associated with an immune system disease, including autoimmune diseases (e.g. rheumatic diseases), graft-related disorders and immunoproliferative diseases. DMARDs modify one or more of the symptoms and/or disease progression associated with rheumatic disease. Symptoms of rheumatic diseases, include the following: joint swelling, pain, tenderness, morning stiffness, structural damage, an elevated level of serum C-reactive protein (CRP), an elevated level of soluble IL-2r, an elevated level of soluble ICAM-1, an elevated level of soluble E-selectin, an elevated level of rheumatoid factor, an elevated level of IL-6 or an elevated erythrocyte sedimentation rate (ESR). These symptoms and the reduction of these symptoms can be evaluated by any well known evaluation methods including: Health Questionnaire Assessments; [0142] ACR 20, 50, 70; and/or Medical Outcomes Study Short Form-36.
  • DMARDs include, but are not limited to, dihydrofolic acid reductase inhibitors e.g., methotrexate; cyclophosphamide; cyclosporine; cyclosporin A; chloroquine; hydroxychloroquine; sulfasalazine (sulphasalazopyrine) gold salts D-penicillamine; leflunomide; azathioprine; anakinra; TNF blockers e.g., infliximab (REMICADER) or etanercept; and a biological agent that targets an inflammatory cytokine. [0143]
  • As used herein, NSAID refers to a Non-Steroidal Anti-Inflammatory Drug. NSAIDs reduce inflammatory reactions in a subject. NSAIDs include, but are not limited to acetyl salicylic acid, choline magnesium salicylate, diflunisal, magnesium salicylate, salsalate, sodium salicylate, diclofenac, etodolac, fenoprofen, flurbiprofen, indomethacin, ketoprofen, ketorolac, meclofenamate, naproxen, nabumetone, phenylbutazone, piroxicam, sulindac, tolmetin, acetaminophen, ibuprofen, Cox-2 inhibitors, meloxicam and tramadol. [0144]
  • As used herein, “ligand” refers to a molecule that specifically recognizes and binds another molecule, for example, a ligand for CTLA4 is a B7 molecule. In a further example, a ligand for the B7 molecule is a CTLA4 and/or CD28 molecule. The interaction of a molecule and its ligand can be regulated by compositions of the invention. For example, CTLA4 interaction with its ligand B7 can be blocked by administration of CTLA4Ig molecules. Alternatively, Tumor Necrosis Factor (TNF) interaction with its ligand, TNF receptor (TNFR), can be blocked by administration of etanercept or other TNF/TNFR blocking molecules. [0145]
  • As used herein “wild type CTLA4” or “non-mutated CTLA4” has the amino acid sequence of naturally occurring, full length CTLA4 as shown in FIG. 23 (also as described in U.S. Pat. Nos. 5,434,131, 5,844,095, and 5,851,795 herein incorporated by reference in their entirety), or any portion or derivative thereof, that recognizes and binds a B7 or interferes with a B7 so that it blocks binding to CD28 and/or CTLA4 (e.g., endogenous CD28 and/or CTLA4). In particular embodiments, the extracellular domain of wild type CTLA4 begins with methionine at position +1 and ends at aspartic acid at position +124, or the extracellular domain of wild type CTLA4 begins with alanine at position −1 and ends at aspartic acid at position +124 as shown in FIG. 23. Wild type CTLA4 is a cell surface protein, having an N-terminal extracellular domain, a transmembrane domain, and a C-terminal cytoplasmic domain. The extracellular domain binds to target molecules, such as a B7 molecule. In a cell, the naturally occurring, wild type CTLA4 protein is translated as an immature polypeptide, which includes a signal peptide at the N-terminal end. The immature polypeptide undergoes post-translational processing, which includes cleavage and removal of the signal peptide to generate a CTLA4 cleavage product having a newly generated N-terminal end that differs from the N-terminal end in the immature form. One skilled in the art will appreciate that additional post-translational processing may occur, which removes one or more of the amino acids from the newly generated N-terminal end of the CTLA4 cleavage product. Alternatively, the signal peptide may not be removed completely, generating molecules that begin before the common starting amino acid methionine. Thus, the mature CTLA4 protein may start at methionine at position +1 or alanine at position −1. The mature form of the CTLA4 molecule includes the extracellular domain or any portion thereof, which binds to B7. [0146]
  • As used herein, a “CTLA4 mutant molecule” means wildtype CTLA4 as shown in FIG. 23 or any portion or derivative thereof, that has a mutation or multiple mutations (preferably in the extracellular domain of wildtype CTLA4). A CTLA4 mutant molecule has a sequence that it is similar but not identical to the sequence of wild type CTLA4 molecule, but still binds a B7. The mutations may include one or more amino acid residues substituted with an amino acid having conservative (e.g., substitute a leucine with an isoleucine) or non-conservative (e.g., substitute a glycine with a tryptophan) structure or chemical properties, amino acid deletions, additions, frameshifts, or truncations. CTLA4 mutant molecules may include a non-CTLA4 molecule therein or attached thereto. The mutant molecules may be soluble (i.e., circulating) or bound to a cell surface. Additional CTLA4 mutant molecules include those described in U.S. patent application Ser. Nos. 09/865,321, 60/214,065 and 60/287,576; in U.S. Pat. Nos. 6,090,914 5,844,095 and 5,773,253; and as described by Peach, R. J., et al., in [0147] J Exp Med 180:2049-2058 (1994)). CTLA4 mutant molecules can be made synthetically or recombinantly.
  • “CTLA4Ig” is a soluble fusion protein comprising an extracellular domain of wildtype CTLA4 that binds B7, or a portion thereof, joined to an immunoglobulin constant region (Ig), or a portion thereof. A particular embodiment comprises the extracellular domain of wild type CTLA4 (as shown in FIG. 23) starting at methionine at position +1 and ending at aspartic acid at position +124, or starting at alanine at position −1 to aspartic acid at position +124; a junction amino acid residue glutamine at position +125; and an immunoglobulin portion encompassing glutamic acid at position +126 through lysine at position +357 (DNA encoding CTLA4Ig was deposited on May 31, 1991 with the American Type Culture Collection (ATCC), 10801 University Blvd., Manassas, Va. 20110-2209 under the provisions of the Budapest Treaty, and has been accorded ATCC accession number ATCC 68629; Linsley, P., et al., 1994 [0148] Immunity 1:793-80). CTLA4Ig-24, a Chinese Hamster Ovary (CHO) cell line expressing CTLA4Ig was deposited on May 31, 1991 with ATCC identification number CRL-10762). The soluble CTLA4Ig molecules used in the methods and/or kits of the invention may or may not include a signal (leader) peptide sequence. Typically, in the methods and/or kits of the invention, the molecules do not include a signal peptide sequence.
  • “L104EA29YIg” is a fusion protein that is a soluble CTLA4 mutant molecule comprising an extracellular domain of wildtype CTLA4 with amino acid changes A29Y (a tyrosine amino acid residue substituting for an alanine at position 29) and L104E (a glutamic acid amino acid residue substituting for a leucine at position +104), or a portion thereof that binds a B7 molecule, joined to an Ig tail (included in FIG. 19; DNA encoding L104EA29YIg was deposited on Jun. 20, 2000 with ATCC number PTA-2104; copending in U.S. patent application Ser. Nos. 09/579,927, 60/287,576 and 60/214,065, incorporated by reference herein). The soluble L104EA29YIg molecules used in the methods and/or kits of the invention may or may not include a signal (leader) peptide sequence. Typically, in the methods and/or kits of the invention, the molecules do not include a signal peptide sequence. [0149]
  • As used herein, “soluble” refers to any molecule, or fragments and derivatives thereof, not bound or attached to a cell, i.e., circulating. For example, CTLA4, B7 or CD28 can be made soluble by attaching an immunoglobulin (Ig) moiety to the extracellular domain of CTLA4, B7 or CD28, respectively. Alternatively, a molecule such as CTLA4 can be rendered soluble by removing its transmembrane domain. Typically, the soluble molecules used in the methods, compositions and/or kits of the invention do not include a signal (or leader) sequence. [0150]
  • As used herein, “soluble CTLA4 molecules” means non-cell-surface-bound (i.e. circulating) CTLA4 molecules or any functional portion of a CTLA4 molecule that binds B7 including, but not limited to: CTLA4Ig fusion proteins (e.g. encoded by DNA deposited with ATCC accession number 68629), wherein the extracellular domain of CTLA4 is fused to an immunoglobulin (Ig) moiety such as IgCγ1 (IgCgamma1), IgCγ2 (IgCgamma2), IgCγ3 (IgCgamma3), IgCγ4 (IgCgamma4), IgCμ (IgCmu), IgCα1 (IgCalpha1), IgCα2 (IgCalpha2), IgCδ (IgCdelta) or IgCε (IgCepsilon), rendering the fusion molecule soluble, or fragments and derivatives thereof; proteins with the extracellular domain of CTLA4 fused or joined with a portion of a biologically active or chemically active protein such as the papillomavirus E7 gene product (CTLA4-E7), melanoma-associated antigen p97 (CTLA4-p97) or HIV env protein (CTLA4-env gp 120) (as described in U.S. Pat. No. 5,844,095, herein incorporated by reference in its entirety), or fragments and derivatives thereof; hybrid (chimeric) fusion proteins such as CD28/CTLA4Ig (as described in U.S. Pat. No. 5,434,131, herein incorporated by reference in its entirety), or fragments and derivatives thereof; CTLA4 molecules with the transmembrane domain removed to render the protein soluble (Oaks, M. K., et al., 2000 [0151] Cellular Immunology 201:144-153, herein incorporated by reference in its entirety), or fragments and derivatives thereof. “Soluble CTLA4 molecules” also include fragments, portions or derivatives thereof, and soluble CTLA4 mutant molecules, having CTLA4 binding activity. The soluble CTLA4 molecules used in the methods of the invention may or may not include a signal (leader) peptide sequence. Typically, in the methods, compositions and/or kits of the invention, the molecules do not include a signal peptide sequence.
  • As used herein “the extracellular domain of CTLA4” is the portion of CTLA4 that recognizes and binds CTLA4 ligands, such as B7 molecules. For example, an extracellular domain of CTLA4 comprises methionine at position +1 to aspartic acid at position +124 (FIG. 23). Alternatively, an extracellular domain of CTLA4 comprises alanine at position −1 to aspartic acid at position +124 (FIG. 23). The extracellular domain includes fragments or derivatives of CTLA4 that bind a B7 molecule. The extracellular domain of CTLA4 as shown in FIG. 23 may also include mutations that change the binding avidity of the CTLA4 molecule for a B7 molecule. [0152]
  • As used herein, the term “mutation” means a change in the nucleotide or amino acid sequence of a wildtype molecule, for example, a change in the DNA and/or amino acid sequences of the wild-type CTLA4 extracellular domain. A mutation in DNA may change a codon leading to a change in the amino acid sequence. A DNA change may include substitutions, deletions, insertions, alternative splicing, or truncations. An amino acid change may include substitutions, deletions, insertions, additions, truncations, or processing or cleavage errors of the protein. Alternatively, mutations in a nucleotide sequence may result in a silent mutation in the amino acid sequence as is well understood in the art. In that regard, certain nucleotide codons encode the same amino acid. Examples include nucleotide codons CGU, CGG, CGC, and CGA encoding the amino acid, arginine (R); or codons GAU, and GAC encoding the amino acid, aspartic acid (D). Thus, a protein can be encoded by one or more nucleic acid molecules that differ in their specific nucleotide sequence, but still encode protein molecules having identical sequences. The amino acid coding sequence is as follows: [0153]
    One Letter
    Amino Acid Symbol Symbol Codons
    Alanine Ala A GCU, GCC, GCA, GCG
    Cysteine Cys C UGU, UGC
    Aspartic Acid Asp D GAU, GAC
    Glutamic Acid Glu E GAA, GAG
    Phenylalanine Phe F UUU, UUC
    Glycine Gly G GGU, GGC, GGA, GGG
    Histidine His H CAU, CAC
    Isoleucine Ile I AUU, AUC, AUA
    Lysine Lys K AAA, AAG
    Leucine Leu L UUA, UUG, CUU, CUC, CUA, CUG
    Methionine Met M AUG
    Asparagine Asn N AAU, AAC
    Proline Pro P CCU, CCC, CCA, CCG
    Glutamine Gln Q CAA, CAG
    Arginine Arg R CGU, CGC, CGA, CGG, AGA, AGG
    Serine Ser S UCU, UCC, UCA, UCG, AGU, AGC
    Threonine Thr T ACU, ACC, ACA, ACG
    Valine Val V GUU, GUC, GUA, GUG
    Tryptophan Trp W UGG
    Tyrosine Tyr Y UAU, UAC
  • The mutant molecule may have one or more mutations. As used herein, a “non-CTLA4 protein sequence” or “non-CTLA4 molecule” means any protein molecule that does not bind B7 and does not interfere with the binding of CTLA4 to its target. The non-CTLA4 molecule, attached to the extracellular domain of a CTLA4 molecule can alter the solubility or affinity of the CTLA4 molecule. An example includes, but is not limited to, an immunoglobulin (Ig) constant region or portion thereof. Preferably, the Ig constant region is a human or monkey Ig constant region, e.g., human C(gamma)1, including the hinge, CH2 and CH3 regions. The Ig constant region can be mutated to reduce its effector functions (U.S. Pat. Nos. 5,637,481, 5,844,095 and 5,434,131). [0154]
  • As used herein, a “fragment” or “portion” is any part or segment of a molecule e.g. CTLA4 or CD28, preferably the extracellular domain of CTLA4 or CD28 or a part or segment thereof, that recognizes and binds its target, e.g., a B7 molecule. [0155]
  • As used herein, “B7” refers to the B7 family of molecules including, but not limited to, B7-1 (CD80) (Freeman et al, 1989, J. Immunol. 143:2714-2722, herein incorporated by reference in its entirety), B7-2 (CD86) (Freeman et al, 1993, Science 262:909-911 herein incorporated by reference in its entirety; Azuma et al, 1993, Nature 366:76-79 herein incorporated by reference in its entirety) that may recognize and bind CTLA4 and/or CD28. A B7 molecule can be expressed on an activated B cell. [0156]
  • As used herein, “CD28” refers to the molecule that recognizes and binds B7 as described in U.S. Pat. Nos. 5,580,756 and 5,521,288 (herein incorporated by reference in their entirety). [0157]
  • As used herein, “B7-positive cells” are any cells with one or more types of B7 molecules expressed on the cell surface. [0158]
  • As used herein, a “derivative” is a molecule that shares sequence similarity and activity of its parent molecule. For example, a derivative of CTLA4 includes a soluble CTLA4 molecule having an amino acid sequence at least 70% similar to the extracellular domain of wildtype CTLA4, and which recognizes and binds B7 e.g. CTLA4Ig or soluble CTLA4 mutant molecule L104EA29YIg. A derivative means any change to the amino acid sequence and/or chemical quality of the amino acid e.g., amino acid analogs. [0159]
  • As used herein, to “regulate” an immune response is to activate, stimulate, up-regulate, inhibit, block, down-regulate or modify the immune response. The auto-immune diseases described herein, may be treated by regulating an immune response e.g., by regulating functional CTLA4- and/or CD28-positive cell interactions with B7-positive cells. For example, a method for regulating an immune response comprises contacting the B7-positive cells with a soluble CTLA4 molecule of the invention so as to form soluble CTLA4/B7 complexes, the soluble CTLA4 molecule interfering with reaction of an endogenous CTLA4 and/or CD28 molecule with said B7 molecule. [0160]
  • As used herein, to “block” or “inhibit” a receptor, signal or molecule means to interfere with the activation of the receptor, signal or molecule, as detected by an art-recognized test. For example, blockage of a cell-mediated immune response can be detected by determining reduction of Rheumatic Disease associated symptoms. Blockage or inhibition may be partial or total. [0161]
  • As used herein, “blocking B7 interaction” means to interfere with the binding of B7 to its ligands, such as CD28 and/or CTLA4, thereby obstructing T-cell and B7-positive cell interactions. Examples of agents that block B7 interactions include, but are not limited to, molecules such as an antibody (or portion or derivative thereof) that recognizes and binds to the any of CTLA4, CD28 or B7 molecules (e.g. B7-1, B7-2); a soluble form (or portion or derivative thereof) of the molecules such as soluble CTLA4; a peptide fragment or other small molecule designed to interfere with the cell signal through the CTLA4/CD28/B7-mediated interaction. In a preferred embodiment, the blocking agent is a soluble CTLA4 molecule, such as CTLA4Ig (ATCC 68629) or L104EA29YIg (ATCC PTA-2104), a soluble CD28 molecule such as CD28Ig (ATCC 68628), a soluble B7 molecule such as B7Ig (ATCC 68627), an anti-B7 monoclonal antibody (e.g. ATCC HB-253, ATCC CRL-2223, ATCC CRL-2226, ATCC HB-301, ATCC HB-11341 and monoclonal antibodies as described in by Anderson et al in U.S. Pat. No. 6,113,898 or Yokochi et al., 1982. J. Immun., 128(2)823-827), an anti-CTLA4 monoclonal antibody (e.g. ATCC HB-304, and monoclonal antibodies as described in references 82-83) and/or an anti-CD28 monoclonal antibody (e.g. ATCC HB 11944 and mAb 9.3 as described by Hansen (Hansen et al., 1980. Immunogenetics 10: 247-260) or Martin (Martin et al., 1984. J. Clin. Immun., 4(1):18-22)). Blocking B7 interactions can be detected by art-recognized tests such as determining reduction of immune disease (e.g., rheumatic disease) associated symptoms, by determining reduction in T-cell/B7-cell interactions or by determining reduction in B7 interaction with CTLA4 and/or CD28. Blockage may be partial or total. [0162]
  • As used herein, an “effective amount” of a molecule is defined as an amount that blocks the interaction of the molecule with its ligand. For example, an effective amount of a molecule that blocks B7 interaction with CTLA4 and/or CD28 may be defined as the amount of the molecule that, when bound to B7 molecules on B7-positive cells, inhibit B7 molecules from binding endogenous ligands such as CTLA4 and CD28. Alternatively, an effective amount of a molecule that blocks B7 interaction with CTLA4 and/or CD28 may be defined as the amount of the molecule that, when bound to CTLA4 and/or CD28 molecules on T cells, inhibit B7 molecules from binding endogenous ligands such as CTLA4 and CD28. The inhibition or blockage may be partial or complete. [0163]
  • As used herein, “treating” a disease means to manage a disease by medicinal or other therapies. Treatment of a disease may ameliorate the symptoms of a disease, reduce the severity of a disease, alter the course of disease progression and/or ameliorate or cure the basic disease problem. For example, to treat an auto-immune disease may be accomplished by regulating an immune response e.g., by regulating functional CTLA4- and/or CD28-positive cell interactions with B7-positive cells. Alternatively, treating an auto-immune disease may be accomplished by preventing the disease from occurring or progressing through the use of the compositions described herein. [0164]
  • As used herein, “immune system disease” means any disease mediated by T-cell interactions with B7-positive cells including, but not limited to, autoimmune diseases, graft related disorders and immunoproliferative diseases. Examples of immune system diseases include graft versus host disease (GVHD) (e.g., such as may result from bone marrow transplantation, or in the induction of tolerance), immune disorders associated with graft transplantation rejection, chronic rejection, and tissue or cell allo- or xenografts, including solid organs (e.g., kidney transplants), skin, islets, muscles, hepatocytes, neurons. Examples of immunoproliferative diseases include, but are not limited to, psoriasis, T-cell lymphoma, T-cell acute lymphoblastic leukemia, testicular angiocentric T-cell lymphoma, benign lymphocytic angiitis, lupus (e.g. lupus erythematosus, lupus nephritis), Hashimoto's thyroiditis, primary myxedema, Graves' disease, pernicious anemia, autoimmune atrophic gastritis, Addison's disease, diabetes (e.g. insulin dependent diabetes mellitis, type I diabetes mellitis, type II diabetes mellitis), good pasture's syndrome, myasthenia gravis, pemphigus, Crohn's disease, sympathetic ophthalmia, autoimmune uveitis, multiple sclerosis, autoimmune hemolytic anemia, idiopathic thrombocytopenia, primary biliary cirrhosis, chronic action hepatitis, ulceratis colitis, Sjogren's syndrome, rheumatic diseases (e.g. rheumatoid arthritis), polymyositis, scleroderma, and mixed connective tissue disease. [0165]
  • As used herein, “rheumatic diseases” means any disease that affects the joints, bone, soft tissue, or spinal cord (Mathies, H. 1983 [0166] Rheuma) and comprises inflammatory rheumatism, degenerative rheumatism, extra-articular rheumatism, and collagen diseases. Additionally, rheumatic diseases include, but are not limited to, chronic polyarthritis, psoriasis arthropathica, ankylosing spondylitis, rheumatoid arthritis, panarteriitis nodosa, systemic lupus erythematosus, progressive systemic scleroderma, periarthritis humeroscapularis, arthritis uratica, chondrocalcinosis, dermatomyositis, muscular rheumatism, myositis, and myogelosis. Some rheumatic diseases are known to be autoimmune diseases caused by a subject's altered immune response.
  • As used herein, “gene therapy” is a process to treat a disease by genetic manipulation. Gene therapy involves introducing a nucleic acid molecule into a cell and the cell expressing a gene product encoded by the nucleic acid molecule. For example, as is well known by those skilled in the art, introducing the nucleic acid molecule into a cell may be performed by introducing an expression vector containing the nucleic acid molecule of interest into cells ex vivo or in vitro by a variety of methods including, for example, calcium phosphate precipitation, diethyaminoethyl dextran, polyethylene glycol (PEG), electroporation, direct injection, lipofection or viral infection (Sambrook et al., [0167] Molecular Cloning: A Laboratory Manual (Cold Spring Harbor Laboratory Press 1989); Kriegler M. Gene Transfer ad Expression: A Laboratory Manual (W. H. Freeman and Co, New York, N.Y., 1993) and Wu, Methods in Enzymology (Academic Press, New York, 1993), each of which is incorporated herein by reference). Alternatively, nucleotide sequences of interest may be introduced into a cell in vivo using a variety of vectors and by a variety of methods including, for example: direct administration of the nucleic acid into a subject (Williams et al, 1991 PNAS 88:2726-2730); or insertion of the nucleic acid molecule into a viral vector, production of the recombinant virus or viral particle, and infection of the subject with the recombinant virus (Battleman et al, 1993 J Neurosci 13:94-951; Carroll et al, 1993 J Cell Biochem 17E:241; Lebkowski et al, U.S. Pat. No. 5,354,678; Davison and Elliott, Molecular Virology: A Practical Approach (IRL Press, New York, 1993)). Other methods used for in vivo transfer include encapsulation of the nucleic acid into liposomes, and direct introduction of the liposomes, or liposomes combined with a hemagglutinating Sendai virus, into a subject (U.S. Pat. No. 5,824,655, incorporated by reference herein). The transfected or infected cells express the protein products encoded by the nucleic acid in order to ameliorate a disease or the symptoms of a disease.
  • As used herein, “Health Questionnaire Assessments (HAQs)” refers to a set of questions used to evaluate patients for symptoms of disease activity. These symptoms included: joint swelling, joint tenderness, inflammation, morning stiffness, disease activity and disability evaluated by each patient in a self-administered questionnaire regarding their physical well-being and function, disease activity and disability as evaluated a physician, and pain (Fries, J. F., et al., 1982 [0168] J. of Rheumatology 9:789-793).
  • As used herein, “ACR” refers to clinical response studies based on criteria established by the American College of Rheumatology. A subject satisfied the “ACR20” criterion if there was about a 20 percent improvement in tender and swollen joint counts and 20 percent improvement in three of five remaining symptoms measured, such as patient and physician global disease changes, pain, physical disability, and an acute phase reactant such as CRP or ESR (Felson, D. T., et al., 1993 [0169] Arthritis and Rheumatism 36:729-740; Felson, D. T., et al., 1995 Arthritis and Rheumatism 38:1-9). Similarly, a subject satisfied the “ACR50” or “ACR70” criterion if there was about a 50 or 70 percent improvement, respectively, in tender and swollen joint counts and about 50 or 70 percent improvement, respectively, in three of five remaining symptoms measured, such as patient and physician global disease changes, pain, physical disability, and an acute phase reactant such as CRP or ESR.
  • As used herein, the “Medical Outcomes Study Short Form-36 (SF-36)” refers to forms used to evaluate the impact of a DMARD (e.g., methotrexate or etanercept) and CTLA4Ig therapy on health-related quality of life (HRQOL). The SF-36 consists of 36 items which covers four physical and four mental domains (physical function, role-physical, bodily pain, general health, vitality, social function, role emotional, and mental health). These individual domains are used to derive the physical and mental component summary scores which range from about 0 to 100, with higher scores indicating better quality of life. Absolute differences of 5 or more in the SF-36 scores were considered clinically meaningful. [0170]
  • As used herein, “alleviate” refers to lessening or making less severe, one or more of the symptoms of an immune disease (e.g., rheumatic disease) including, but not limited to, joint swelling, pain, tenderness, morning stiffness, structural damage, an elevated level of serum C-reactive protein (CRP), an elevated level of soluble IL-2r, an elevated level of soluble ICAM-1, an elevated level of soluble E-selectin, an elevated level of rheumatoid factor, an elevated level of IL-6 or an elevated erythrocyte sedimentation rate. [0171]
  • In order that the invention herein described may be more fully understood the following description is set forth. [0172]
  • Compositions and Methods of the Invention [0173]
  • The present invention provides compositions and methods for treating immune system diseases, such as rheumatic diseases, by administering to a subject an effective amount of a ligand that blocks B7 interactions with CTLA4 and/or CD28. For example, such ligands include: soluble CTLA4 molecules (such as CTLA4Ig, CTLA4-E7, CTLA4-p97, CTLA4-env gp120, and mutant CTLA4 molecules such as, CTLA4/CD28Ig, L104EA29YIg, L104EA29LIg, L104EA29TIg and/or L104EA29WIg), soluble CD28 molecules, soluble B7-1 molecules, soluble B7-2 molecules, and monoclonal antibodies that recognize and bind B7, CD28 and/or CTLA4 (e.g., an anti-CTLA4 monoclonal antibody, an anti-CD28 monoclonal antibody, an anti-B7-1 monoclonal antibody or an anti-B7-2 monoclonal antibody. [0174]
  • Further, the present invention provides compositions and methods for treating immune system diseases, such as rheumatic diseases, by administering to a subject a combination of an effective amount of 1) a DMARD (such as methotrexate or a molecule that blocks TNF interactions, e.g., blocks TNF interactions with its ligand) or other therapeutic agent, plus 2) an effective amount of a molecule that blocks B7 interaction with CTLA4 and/or CD28 such as soluble CTLA4 molecules (e.g., CTLA4Ig, CTLA4Ig/CD28Ig, CTLA4-E7, CTLA4-p97, CTLA4-env gp120, L104EA29YIg, L104EA29LIg, L104EA29TIg and/or L104EA29WIg), soluble CD28 molecules, soluble B7-1 molecules, soluble B7-2 molecules, and monoclonal antibodies that recognize and bind B7, CD28 and/or CTLA4 (e.g., an anti-CTLA4 monoclonal antibody, an anti-CD28 monoclonal antibody, an anti-B7-1 monoclonal antibody or an anti-B7-2 monoclonal antibody). [0175]
  • An effective amount of a molecule that blocks B7 interaction with CTLA4 and/or CD28 may be defined as the amount of anti-B7 monoclonal antibodies, soluble CTLA4 and/or soluble CD28 molecules that, when bound to B7 molecules on B7-positive cells, inhibit B7 molecules from binding endogenous ligands such as CTLA4 and CD28. The inhibition may be partial or complete. [0176]
  • Alternatively, an effective amount of a molecule that blocks B7 interaction with CTLA4 and/or CD28 may be defined as the amount of anti-CTLA4 monoclonal antibody, anti-CD28 monoclonal antibody or soluble B7 (B7-1 or B7-2) molecules that, when bound to CTLA4 and/or CD28 molecules on T cells, inhibit B7 molecules from binding endogenous ligands such as CTLA4 and CD28. The inhibition may be partial or complete. [0177]
  • An effective amount of a molecule that blocks B7 interaction with CTLA4 and/or CD28 is an amount about 0.1 to 100 mg/kg weight of a subject. In another embodiment, the effective amount is an amount about 0.5 to 5 mg/kg weight of a subject, 0.1 to 5 mg/kg weight of a subject, about 5 to 10 mg/kg weight of a subject, about 10 to 15 mg/kg weight of a subject, about 15 to 20 mg/kg weight of a subject, about 20 to 25 mg/kg weight of a subject, about 25 to 30 mg/kg weight of a subject, about 30 to 35 mg/kg weight of a subject, about 35 to 40 mg/kg weight of a subject, about 40 to 45 mg/kg of a subject, about 45 to 50 mg/kg weight of a subject, about 50 to 55 mg/kg weight of a subject, about 55 to 60 mg/kg weight of a subject, about 60 to 65 mg/kg weight of a subject, about 65 to 70 mg/kg weight of a subject, about 70 to 75 mg/kg weight of a subject, about 75 to 80 mg/kg weight of a subject, about 80 to 85 mg/kg weight of a subject, about 85 to 90 mg/kg weight of a subject, about 90 to 95 mg/kg weight of a subject, or about 95 to 100 mg/kg weight of a subject. [0178]
  • In an embodiment, the effective amount of a molecule that blocks B7 interaction with CTLA4 and/or CD28 is an amount about 2 mg/kg to about 10 mg/kg weight of a subject. The preferred amount is 10 mg/kg weight of a subject. In another embodiment, the effective amount is an amount about 0.1 to 4 mg/kg weight of a subject. In another embodiment the effective amount is an amount about 0.1 to 0.5 mg/kg weight of a subject, about 0.5 to 1.0 mg/kg weight of a subject, about 1.0 to 1.5 mg/kg weight of a subject, about 1.5 to 2.0 mg/kg weight of a subject, about 2.0 to 2.5 mg/kg weight of a subject, about 2.5 to 3.0 mg/kg weight of a subject, about 3.0 to 3.5 mg/kg weight of a subject, about 3.5 to 4.0 mg/kg weight of a subject, about 4.0 to 4.5 mg/kg weight of a subject, about 4.5 to 5.0 mg/kg weight of a subject, about 5.0 to 5.5 mg/kg weight of a subject, about 5.5 to 6.0 mg/kg weight of a subject, about 6.0 to 6.5 mg/kg weight of a subject, about 6.5 to 7.0 mg/kg weight of a subject, about 7.0 to 7.5 mg/kg weight of a subject, about 7.5 to 8.0 mg/kg weight of a subject, about 8.0 to 8.5 mg/kg weight of a subject, about 8.5 to 9.0 mg/kg weight of a subject, about 9.0 to 9.5 mg/kg weight of a subject, about 9.5 to 10.0 mg/kg weight of a subject. [0179]
  • In another embodiment, the effective amount is an amount about 0.1 to 20 mg/kg weight of a subject. In another embodiment, the effective amount is an amount about 0.1 to 2 mg/kg weight of a subject, about 2 to 4 mg/kg weight of a subject, about 4 to 6 mg/kg weight of a subject, about 6 to 8 mg/kg weight of a subject, about 8 to 10 mg/kg weight of a subject, about 10 to 12 mg/kg weight of a subject, about 12 to 14 mg/kg weight of a subject, about 14 to 16 mg/kg weight of a subject, about 16 to 18 mg/kg weight of a subject or about 18 to 20 mg/kg weight of a subject. [0180]
  • In another embodiment, the effective amount is about 2 mg/kg weight of a subject. In yet another embodiment, the effective amount is about 10 mg/kg weight of a subject. [0181]
  • In a specific embodiment, the molecule that blocks B7 interaction with CTLA4 and/or CD28 is soluble CTLA4 and the effective amount of a soluble CTLA4 molecule is about 2 mg/kg weight of a subject. In another specific embodiment, the effective amount of a soluble CTLA4 molecule is about 10 mg/kg weight of a subject. In another specific embodiment, an effective amount of a soluble CTLA4 is 500 mg for a subject weighing less than 60 kg, 750 mg for a subject weighing between 60-100 kg and 1000 mg for a subject weighing more than 100 kg. [0182]
  • An effective amount of the molecule that blocks B7 interaction with CTLA4 and/or CD28 is soluble CTLA4 may be administered to a subject daily, weekly, monthly and/or yearly, in single or multiple times per hour/day/week/month/year, depending on need. For example, in one embodiment, the molecule may initially be administered once every two weeks for a month, and then once every month thereafter. [0183]
  • In a preferred embodiment, the immune disease is a rheumatic disease. Rheumatic diseases are any diseases which are characterized by (i) inflammation or degeneration of musculo-skeletal or connective tissue structures of the body, particularly the joints, and including muscles, tendons, cartilage, synovial and fibrous tissues, (ii) accompanied by joint swelling, joint tenderness, inflammation, morning stiffness, and/or pain, or impairment of locomotion or function of those structures and, in some cases, (iii) often accompanied by serological evidence of rheumatoid factor and other inflammatory surrogate markers. [0184]
  • Rheumatic diseases include, but are not limited to, rheumatoid arthritis. The symptoms of rheumatoid arthritis include joint swelling, joint tenderness, inflammation, morning stiffness, and pain leading to physical disability. Subjects afflicted with the advanced stages of arthritis suffer from symptoms of structural damage and debilitating pain. Other organs also can be impaired by the autoimmune mechanism. [0185]
  • In an embodiment of the invention used to treat an immune system disease, the DMARD is methotrexate or a molecule that blocks TNF interactions such as etanercept, and the molecule that blocks B7 interaction with CTLA4 and/or CD28 is a soluble CTLA4. In a further embodiment, the methods of the invention comprise administering to a subject an effective amount of methotrexate or a molecule that blocks TNF interactions in combination with an effective amount of soluble CTLA4 in order to treat rheumatic diseases such as rheumatoid arthritis. [0186]
  • Effective amounts of methotrexate range about 0.1 to 40 mg/week. In one embodiment, the effective amount includes ranges of about 0.1 to 5 mg/week, about 5 to 10 mg/week, about 10 to 15 mg/week, about 15 to 20 mg/week, about 20 to 25 mg/week, about 25 to 30 mg/week, about 30 to 35 mg/week, or about 35 to 40 mg/week. In one embodiment, methotrexate is administered in an amount ranging about 5 to 30 mg/week. [0187]
  • In one embodiment, the effective amount of a soluble CTLA4 molecule is about 2 mg/kg weight subject and the effective amount of methotrexate is about 10 to 30 mg/week. In another embodiment, the effective amount of a soluble CTLA4 molecule is about 10 mg/kg weight subject and the effective amount of methotrexate is about 10 to 30 mg/week. [0188]
  • In an embodiment of the invention used to treat an immune system disease, the DMARD is etanercept and the molecule that blocks B7 interaction with CTLA4 and/or CD28 is a soluble CTLA4. In a further embodiment, the methods of the invention comprise administering to a subject an effective amount of etanercept in combination with an effective amount of soluble CTLA4 in order to treat rheumatic diseases such as rheumatoid arthritis. [0189]
  • Effective amounts of etanercept range about 0.1 to 100 mg/week. In one embodiment, the effective amount includes ranges of about 10 to 100 mg/week, about 0.1 to 50 mg/week, about 0.1 to 5 mg/week, about 5 to 10 mg/week, about 10 to 15 mg/week, about 15 to 20 mg/week, about 20 to 25 mg/week, about 25 to 30 mg/week, about 30 to 35 mg/week, about 35 to 40 mg/week, about 40 to 45 mg/week, about 45 to 50 mg/week, about 50 to 55 mg/week, about 55 to 60 mg/week, about 60 to 65 mg/week, about 65 to 70 mg/week, about 70 to 75 mg/week, about 75 to 80 mg/week, about 80 to 85 mg/week, about 85 to 90 mg/week, about 90 to 95 mg/week or about 95 to 100 mg/week. In one embodiment, etanercept is administered in an amount of about 50 mg/week, alternatively etanercept may be administered in an amount of about 25 mg twice weekly. [0190]
  • In one embodiment, the effective amount of a soluble CTLA4 molecule is about 2 mg/kg weight subject and the effective amount of etanercept is about 25 mg twice a week. In another embodiment, the effective amount of a soluble CTLA4 molecule is about 10 mg/kg weight subject and the effective amount of etanercept is about 25 mg twice a week. [0191]
  • The invention also provides compositions and methods for treating immune system diseases, such as rheumatic diseases, by administering to a subject a combination of an effective amount of an NSAID and/or other therapeutic agent plus an effective amount of a molecule that blocks B7 interaction with CTLA4 and/or CD28. [0192]
  • The invention also provides compositions and methods for treating immune system diseases, such as rheumatic diseases, by administering to a subject a an effective amount of a glucocorticoid, corticosteroid and/or other therapeutic agent plus an effective amount of a molecule that blocks B7 interaction with CTLA4 and/or CD28. [0193]
  • Compositions [0194]
  • The present invention provides compositions for treating immune diseases, such as rheumatic diseases, comprising soluble CTLA4 molecules. Further, the present invention provides compositions comprising a biological agent that inhibits T-cell function but not T-cell depletion in a human by contacting B7-positive cells in the human with a soluble CTLA4. Examples of soluble CTLA4 include CTLA4Ig and soluble CTLA4 mutant molecules such as L104EA29YIg (FIG. 19), L104EA29LIg (FIG. 20), L104EA29TIg (FIG. 21), and L104EA29WIg (FIG. 22). [0195]
  • CTLA4 molecules, with mutant or wildtype sequences, may be rendered soluble by deleting the CTLA4 transmembrane segment (Oaks, M. K., et al., 2000 [0196] Cellular Immunology 201:144-153).
  • Alternatively, soluble CTLA4 molecules, with mutant or wildtype sequences, may be fusion proteins, wherein the CTLA4 molecules are fused to non-CTLA4 moieties such as immunoglobulin (Ig) molecules that render the CTLA4 molecules soluble. For example, a CTLA4 fusion protein may include the extracellular domain of CTLA4 fused to an immunoglobulin constant domain, resulting in the CTLA4Ig molecule (FIG. 24) (Linsley, P. S., et al., 1994 [0197] Immunity 1:793-80). Examples of immunoglobulin domains that may be fused to CTLA4 include, but are not limited to IgCγ1 (IgCgamma1), IgCγ2 (IgCgamma2), IgCγ3 (IgCgamma3), IgCγ4 (IgCgamma4), IgCμ (IgCmu), IgCα1 (IgCalpha1), IgCα2 (IgCalpha2), IgCδ (IgCdelta) or IgCε (IgCepsilon).
  • For clinical protocols, it is preferred that the immunoglobulin moiety does not elicit a detrimental immune response in a subject. The preferred moiety is the immunoglobulin constant region, including the human or monkey immunoglobulin constant regions. One example of a suitable immunoglobulin region is human Cγ1, including the hinge, CH2 and CH3 regions which can mediate effector functions such as binding to Fc receptors, mediating complement-dependent cytotoxicity (CDC), or mediate antibody-dependent cell-mediated cytotoxicity (ADCC). The immunoglobulin moiety may have one or more mutations therein, (e.g., in the CH2 domain, to reduce effector functions such as CDC or ADCC) where the mutation modulates the binding capability of the immunoglobulin to its ligand, by increasing or decreasing the binding capability of the immunoglobulin to Fc receptors. For example, mutations in the immunoglobulin moiety may include changes in any or all its cysteine residues within the hinge domain, for example, the cysteines at positions +130, +136, and +139 are substituted with serine (FIG. 24). The immunoglobulin moiety may also include the proline at position +148 substituted with a serine, as shown in FIG. 24. Further, the mutations in the immunoglobulin moiety may include having the leucine at position +144 substituted with phenylalanine, leucine at position +145 substituted with glutamic acid, or glycine at position +147 substituted with alanine. [0198]
  • Additional non-CTLA4 moieties for use in the soluble CTLA4 molecules or soluble CTLA4 mutant molecules include, but are not limited to, p97 molecule, env gp120 molecule, E7 molecule, and ova molecule (Dash, B. et al. 1994 [0199] J. Gen. Virol. 75 (Pt 6):1389-97; Ikeda, T., et al. 1994 Gene 138(1-2):193-6; Falk, K., et al. 1993 Cell. Immunol. 150(2):447-52; Fujisaka, K. et al. 1994 Virology 204(2):789-93). Other molecules are also possible (Gerard, C. et al. 1994 Neuroscience 62(3):721; Byrn, R. et al. 1989 63(10):4370; Smith, D. et al. 1987 Science 238:1704; Lasky, L. 1996 Science 233:209).
  • The soluble CTLA4 molecule of the invention can include a signal peptide sequence linked to the N-terminal end of the extracellular domain of the CTLA4 portion of the molecule. The signal peptide can be any sequence that will permit secretion of the molecule, including the signal peptide from oncostatin M (Malik, et al., (1989) [0200] Molec. Cell. Biol. 9: 2847-2853), or CD5 (Jones, N. H. et al., (1986) Nature 323:346-349), or the signal peptide from any extracellular protein. The soluble CTLA4 molecule of the invention can include the oncostatin M signal peptide linked at the N-terminal end of the extracellular domain of CTLA4, and the human immunoglobulin molecule (e.g., hinge, CH2 and CH3) linked to the C-terminal end of the extracellular domain (wildtype or mutated) of CTLA4. This molecule includes the oncostatin M signal peptide encompassing an amino acid sequence having methionine at position −26 through alanine at position −1, the CTLA4 portion encompassing an amino acid sequence having methionine at position +1 through aspartic acid at position +124, a junction amino acid residue glutamine at position +125, and the immunoglobulin portion encompassing an amino acid sequence having glutamic acid at position +126 through lysine at position +357.
  • Specifically, the soluble CTLA4 mutant molecules of the invention, comprising the mutated CTLA4 sequences described infra, can be fusion molecules comprising human Ig, e.g., IgC(gamma)1 (i.e. IgCγ1) moieties fused to the mutated CTLA4 fragments. [0201]
  • In one embodiment, the soluble CTLA4 mutant molecules comprise IgCγ1 (IgCgamma1) fused to an extracellular domain of CTLA4 comprising a single-site mutation in the extracellular domain. The extracellular domain of CTLA4 comprises methionine at position +1 through aspartic acid at position +124 (e.g., FIG. 23). The extracellular domain of the CTLA4 can comprise alanine at position −1 through aspartic acid at position +124 (e.g., FIG. 23). Examples of single-site mutations-include the following wherein the leucine at position +104 is changed to any other amino acid. [0202]
    Single-site mutant: Codon change:
    L104EIg Glutamic acid GAG
    L104SIg Serine AGT
    L104TIg Threonine ACG
    L104AIg Alanine GCG
    L104WIg Tryptophan TGG
    L104QIg Glutamine CAG
    L104KIg Lysine AAG
    L104RIg Arginine CGG
    L104GIg Glycine GGG
  • Further, the invention provides mutant molecules having the extracellular domain of CTLA4 with two mutations, fused to an Ig Cγ1 (IgCgamma1) moiety. Examples include the following wherein the leucine at position +104 is changed to another amino acid (e.g. glutamic acid) and the glycine at position +105, the serine at position +25, the threonine at position +30 or the alanine at position +29 is changed to any other amino acid: [0203]
    Double-site mutants: Codon change:
    L104EG105FIg Phenylalanine TTC
    L104EG105WIg Tryptophan TGG
    L104EG105LIg Leucine CTT
    L104ES25RIg Arginine CGG
    L104ET30GIg Glycine GGG
    L104ET30NIg Asparagine AAT
    L104EA29YIg Tyrosine TAT
    L104EA29LIg Leucine TTG
    L104EA29TIg Threonine ACT
    L104EA29WIg Tryptophan TGG
  • Further still, the invention provides mutant molecules having the extracellular domain of CTLA4 comprising three mutations, fused to an IgCγ1 (IgCgamma1) moiety. Examples include the following wherein the leucine at position +104 is changed to another amino acid (e.g. glutamic acid), the alanine at position +29 is changed to another amino acid (e.g. tyrosine) and the serine at position +25 is changed to another amino acid: [0204]
    Triple-site Mutants: Codon changes:
    L104EA29YS25KIg Lysine AAA
    L104EA29YS25KIg Lysine AAG
    L104EA29YS25NIg Asparagine AAC
    L104EA29YS25RIg Arginine CGG
  • Soluble CTLA4 mutant molecules may have a junction amino acid residue which is located between the CTLA4 portion and the Ig portion of the molecule. The junction amino acid can be any amino acid, including glutamine. The junction amino acid can be introduced by molecular or chemical synthesis methods known in the art. [0205]
  • The soluble CTLA4 proteins of the invention, and fragments thereof, can be generated by chemical synthesis methods. The principles of solid phase chemical synthesis of polypeptides are well known in the art and may be found in general texts relating to this area (Dugas, H. and Penney, C. 1981 [0206] Bioorganic Chemistry, pp 54-92, Springer-Verlag, New York). The soluble CTLA4 proteins may be synthesized by solid-phase methodology utilizing an Applied Biosystems 430A peptide synthesizer (Applied Biosystems, Foster City, Calif.) and synthesis cycles supplied by Applied Biosystems. Protected amino acids, such as t-butoxycarbonyl-protected amino acids, and other reagents are commercially available from many chemical supply houses.
  • The present invention provides CTLA4 mutant molecules including a signal peptide sequence linked to the N-terminal end of the extracellular domain of the CTLA4 portion of the mutant molecule. The signal peptide can be any sequence that will permit secretion of the mutant molecule, including the signal peptide from oncostatin M (Malik, et al., 1989 [0207] Molec. Cell. Biol. 9: 2847-2853), or CD5 (Jones, N. H. et al., 1986 Nature 323:346-349), or the signal peptide from any extracellular protein.
  • The invention provides soluble CTLA4 mutant molecules comprising a single-site mutation in the extracellular domain of CTLA4 such as L104EIg (as included in FIG. 18) or L104SIg, wherein L104EIg and L104SIg are mutated in their CTLA4 sequences so that leucine at position +104 is substituted with glutamic acid or serine, respectively. The single-site mutant molecules further include CTLA4 portions encompassing methionine at position +1 through aspartic acid at position +124, a junction amino acid residue glutamine at position +125, and an immunoglobulin portion encompassing glutamic acid at position +126 through lysine at position +357. The immunoglobulin portion of the mutant molecule may also be mutated so that the cysteines at positions +130, +136, and +139 are substituted with serine, and the proline at position +148 is substituted with serine. Alternatively, the single-site soluble CTLA4 mutant molecule may have a CTLA4 portion encompassing alanine at position −1 through aspartic acid at position +124. [0208]
  • The invention provides soluble CTLA4 mutant molecules comprising a double-site mutation in the extracellular domain of CTLA4, such as L104EA29YIg, L104EA29LIg, L104EA29TIg or L104EA29WIg, wherein leucine at position +104 is substituted with a glutamic acid and alanine at position +29 is changed to tyrosine, leucine, threonine and tryptophan, respectively. The sequences for L104EA29YIg, L104EA29LIg, L104EA29TIg and L104EA29WIg, starting at methionine at position +1 and ending with lysine at position +357, plus a signal (leader) peptide sequence are shown in FIGS. [0209] 19-22 respectively. The double-site mutant molecules further comprise CTLA4 portions encompassing methionine at position +1 through aspartic acid at position +124, a junction amino acid residue glutamine at position +125, and an immunoglobulin portion encompassing glutamic acid at position +126 through lysine at position +357. The immunoglobulin portion of the mutant molecule may also be mutated, so that the cysteines at positions +130, +136, and +139 are substituted with serine, and the proline at position +148 is substituted with serine. Alternatively, these mutant molecules can have a CTLA4 portion encompassing alanine at position −1 through aspartic acid at position +124.
  • The invention provides soluble CTLA4 mutant molecules comprising a double-site mutation in the extracellular domain of CTLA4, such as L104EG105FIg, L104EG105WIg and L104EG105LIg, wherein leucine at position +104 is substituted with a glutamic acid and glycine at position +105 is substituted with phenylalanine, tryptophan and leucine, respectively. The double-site mutant molecules further comprise CTLA4 portions encompassing methionine at position +1 through aspartic acid at position +124, a junction amino acid residue glutamine at position +125, and an immunoglobulin portion encompassing glutamic acid at position +126 through lysine at position +357. The immunoglobulin portion of the may also be mutated, so that the cysteines at positions +130, +136, and +139 are substituted with serine, and the proline at position +148 is substituted with serine. Alternatively, these mutant molecules can have a CTLA4 portion encompassing alanine at position −1 through aspartic acid at position +124. [0210]
  • The invention provides L104ES25RIg which is a double-site mutant molecule comprising a CTLA4 portion encompassing methionine at position +1 through aspartic acid at position +124, a junction amino acid residue glutamine at position +125, and the immunoglobulin portion encompassing glutamic acid at position +126 through lysine at position +357. The portion having the extracellular domain of CTLA4 is mutated so that serine at position +25 is substituted with arginine, and leucine at position +104 is substituted with glutamic acid. Alternatively, L104ES25RIg can have a CTLA4 portion encompassing alanine at position −1 through aspartic acid at position +124. [0211]
  • The invention provides soluble CTLA4 mutant molecules comprising a double-site mutation in the extracellular domain of CTLA4, such as L104ET30GIg and L104ET30NIg, wherein leucine at position +104 is substituted with a glutamic acid and threonine at position +30 is substituted with glycine and asparagine, respectively. The double-site mutant molecules further comprise CTLA4 portions encompassing methionine at position +1 through aspartic acid at position +124, a junction amino acid residue glutamine at position +125, and an immunoglobulin portion encompassing glutamic <acid at position +126 through lysine at position +357. The immunoglobulin portion of the mutant molecule may also be mutated, so that the cysteines at positions +130, +136, and +139 are substituted with serine, and the proline at position +148 is substituted with serine. Alternatively, these mutant molecules can have a CTLA4 portion encompassing alanine at position −1 through aspartic acid at position +124. [0212]
  • The invention provides soluble CTLA4 mutant molecules comprising a triple-site mutation in the extracellular domain of CTLA4, such as L104EA29YS25KIg, L104EA29YS25NIg, L104EA29YS25RIg, wherein leucine at position +104 is substituted with a glutamic acid, alanine at position +29 is substituted with tyrosine, and serine at position +25 is substituted with lysine, asparagine and arginine, respectively. The triple-site mutant molecules further comprise CTLA4 portions encompassing methionine at position +1 through aspartic acid at position +124, a junction amino acid residue glutamine at position +125, and an immunoglobulin portion encompassing glutamic acid at position +126 through lysine at position +357. The immunoglobulin portion of the mutant molecule may also be mutated, so that the cysteines at positions +130, +136, and +139 are substituted with serine, and the proline at position +148 is substituted with serine. Alternatively, these mutant molecules can have a CTLA4 portion encompassing alanine at position −1 through aspartic acid at position +124. [0213]
  • Additional embodiments of soluble CTLA4 mutant molecules include chimeric CTLA4/CD28 homologue mutant molecules that bind a B7 (Peach, R. J., et al., 1994 [0214] J Exp Med 180:2049-2058). Examples of these chimeric CTLA4/CD28 mutant molecules include HS1, HS2, HS3, HS4, HS5, HS6, HS4A, HS4B, HS7, HS8, HS9, HS10, HS11, HS12, HS13 and HS14 (U.S. Pat. No. 5,773,253) Preferred embodiments of the invention are soluble CTLA4 molecules such as CTLA4Ig (as shown in FIG. 24, starting at methionine at position +1 and ending at lysine at position +357) and soluble CTLA4 mutant L104EA29YIg (as shown in FIG. 19, starting at methionine at position +1 and ending at lysine at position +357).
  • The invention further provides nucleic acid molecules comprising nucleotide sequences encoding the amino acid sequences corresponding to the soluble CTLA4 molecules of the invention. In one embodiment, the nucleic acid molecule is a DNA (e.g., cDNA) or a hybrid thereof. For example, a CTLA4Ig molecule can comprise a GCT or GCC codon, encoding alanine, at nucleotide position +49 to +51 as shown in FIG. 24. In another example, a CTLA4Ig molecule can comprise a GGT or GGG codon, encoding glycine, at nucleotide position +436 to +438 as shown in FIG. 24. In yet another example, a CTLA4Ig molecule can comprise a CGG or CGT codon, encoding arginine, at nucleotide position +631 to +633 as shown in FIG. 24. DNA encoding CTLA4Ig (FIG. 24) was deposited on May 31, 1991 with the American Type Culture Collection (ATCC), 10801 University Blvd., Manassas, Va. 20110-2209 and has been accorded ATCC accession number ATCC 68629. DNA encoding L104EA29YIg (sequence included in FIG. 19) was deposited on Jun. 19, 2000 with ATCC and has been accorded ATCC accession number PTA-2104. Alternatively, the nucleic acid molecules are RNA or a hybrid thereof. [0215]
  • The nucleic acid molecules of the invention also include derivative nucleic acid molecules which differ from DNA or RNA molecules, and anti-sense molecules. Derivative molecules include peptide nucleic acids (PNAs), and non-nucleic acid molecules including phosphorothioate, phosphotriester, phosphoramidate, and methylphosphonate molecules, that bind to single-stranded DNA or RNA in a base pair-dependent manner (Zamecnik, P. C., et al., 1978 [0216] Proc. Natl. Acad. Sci. 75:280284; Goodchild, P. C., et al., 1986 Proc. Natl. Acad. Sci. 83:4143-4146). Peptide nucleic acid molecules comprise a nucleic acid oligomer to which an amino acid residue, such as lysine, and an amino group have been added. These small molecules, also designated anti-gene agents, stop transcript elongation by binding to their complementary (template) strand of nucleic acid (Nielsen, P. E., et al., 1993 Anticancer Drug Des 8:53-63). Reviews of methods for synthesis of DNA, RNA, and their analogues can be found in: Oligonucleotides and Analogues, eds. F. Eckstein, 1991, IRL Press, New York; Oligonucleotide Synthesis, ed. M. J. Gait, 1984, IRL Press, Oxford, England. Additionally, methods for antisense RNA technology are described in U.S. Pat. Nos. 5,194,428 and 5,110,802. A skilled artisan can readily obtain these classes of nucleic acid molecules using the herein described soluble CTLA4 polynucleotide sequences, see for example Innovative and Perspectives in Solid Phase Synthesis (1992) Egholm, et al. pp 325-328 or U.S. Pat. No. 5,539,082.
  • Additionally, the invention provides a vector, which comprises the nucleotide sequences of the invention. The term vector includes, but is not limited to, plasmids, cosmids, and phagemids. In one embodiment, the vector can be an autonomously replicating vector comprising a replicon that directs the replication of the rDNA within the appropriate host cell. Alternatively, the vector can direct integration of the recombinant vector into the host cell. Various viral vectors may also be used, such as, for example, a number of well known retroviral and adenoviral vectors (Berkner 1988 [0217] Biotechniques 6:616-629).
  • The vectors can permit expression of the soluble CTLA4 transcript or polypeptide sequences in prokaryotic or eukaryotic host cells. The vectors include expression vectors, comprising an expression control element, such as a promoter sequence, which enables transcription of the inserted soluble CTLA4 nucleic acid sequences and can be used for regulating the expression (e.g., transcription and/or translation) of an operably linked soluble CTLA4 sequence in an appropriate host cell. Expression control elements are known in the art and include, but are not limited to, inducible promoters, constitutive promoters, secretion signals, enhancers, transcription terminators, and other transcriptional regulatory elements. Other expression control elements that are involved in translation are known in the art, and include the Shine-Dalgarno sequence (e.g., prokaryotic host cells), and initiation and termination codons. [0218]
  • Specific initiation signals may also be required for efficient translation of a soluble CTLA4 sequence. These signals include the ATG-initiation codon and adjacent sequences. In cases where the soluble CTLA4 initiation codon and upstream sequences are inserted into the appropriate expression vector, no additional translational control signals may be needed. However, in cases where only the coding sequence, or a portion thereof, is inserted, exogenous transcriptional control signals including the ATG-initiation codon may be provided. Furthermore, the initiation codon should be in the correct reading-frame to ensure translation of the entire insert. Exogenous transcriptional elements and initiation codons can be of various origins, both natural and synthetic. The efficiency of expression may be enhanced by the inclusion of enhancers appropriate to the cell system in use (Scharf, D., et al, 1994 [0219] Results Probl. Cell. Differ. 20:125-62; Bittner, et al., 1987 Methods in Enzymol. 153:516-544).
  • The preferred vectors for expression of the soluble CTLA4 sequences in eukaryote host cells include expression control elements, such as the baculovirus polyhedrin promoter for expression in insect cells. Other expression control elements include promoters or enhancers derived from the genomes of plant cells (e.g., heat shock, RUBISCO, storage protein genes), viral promoters or leader sequences or from plant viruses, and promoters or enhancers from the mammalian genes or from mammalian viruses. [0220]
  • The preferred vector includes at least one selectable marker gene that encodes a gene product that confers drug resistance such as resistance to ampicillin or tetracyline. The vector also comprises multiple endonuclease restriction sites that enable convenient insertion of exogenous DNA sequences. Methods for generating a recombinant expression vector encoding the soluble CTLA4 proteins of the invention are well known in the art, and can be found in Sambrook et al., ([0221] Molecular Cloning; A Laboratory Manual, 2nd edition, Sambrook, Fritch, and Maniatis 1989, Cold Spring Harbor Press) and Ausubel et al. (1989 Current Protocols in Molecular Biology, John Wiley & Sons, New York N.Y.).
  • The preferred vectors for generating soluble CTLA4 transcripts and/or the encoded soluble CTLA4 polypeptides are expression vectors which are compatible with prokaryotic host cells. Prokaryotic cell expression vectors are well known in the art and are available from several commercial sources. For example, pET vectors (e.g., pET-21, Novagen Corp.), BLUESCRIPT phagemid (Stratagene, LaJolla, Calif.), pSPORT (Gibco BRL, Rockville, Md.), or ptrp-lac hybrids may be used to express soluble CTLA4 polypeptides in bacterial host cells. [0222]
  • Alternatively, the preferred expression vectors for generating soluble CTLA4 transcripts and/or the encoded soluble CTLA4 polypeptides are expression vectors which are compatible with eukaryotic host cells. The more preferred vectors are those compatible with vertebrate cells. Eukaryotic cell expression vectors are well known in the art and are available from several commercial sources. Typically, such vectors are provided containing convenient restriction sites for insertion of the desired DNA segment. Typical of such vectors are PSVL and pKSV-10 (Pharmacia), pBPV-1/pML2d (International Biotechnologies, Inc.), pTDT1 (ATCC, #31255), and similar eukaryotic expression vectors. [0223]
  • Examples of expression vectors for include, but are not limited to, vectors for mammalian host cells (e.g., BPV-1, pHyg, pRSV, pSV2, pTK2 (Maniatis); pIRES (Clontech); pRc/CMV2, pRc/RSV, pSFV1 (Life Technologies); pVPakc Vectors, pCMV vectors, pSG5 vectors (Stratagene)), retroviral vectors (e.g., pFB vectors (Stratagene)), pcDNA-3 (Invitrogen) or modified forms thereof, adenoviral vectors; adeno-associated virus vectors, baculovirus vectors, yeast vectors (e.g., pESC vectors (Stratagene)). [0224]
  • A host vector system is also provided. The host vector system comprises the vector of the invention in a suitable host cell. Examples of suitable host cells include, but are not limited to, prokaryotic and eukaryotic cells. In accordance with the practice of the invention, eukaryotic cells are also suitable host cells. Examples of eukaryotic cells include any animal cell, whether primary or immortalized, yeast (e.g., [0225] Saccharomyces cerevisiae, Schizosaccharomyces pombe, and Pichia pastoris), and plant cells. Exemplary animal cells include cells from bovine, ovine, porcine, murine, equine, monkey and ape. Myeloma, COS and CHO cells are examples of animal cells that may be used as hosts. Particular CHO cells include, but are not limited to, DG44 (Chasin, et la., 1986 Som. Cell. Molec. Genet. 12:555-556; Kolkekar 1997 Biochemistry 36:10901-10909), CHO-K1 (ATCC No. CCL-61), CHO-K1 Tet-On cell line (Clontech), CHO designated ECACC 85050302 (CAMR, Salisbury, Wiltshire, UK), CHO clone 13 (GEIMG, Genova, IT), CHO clone B (GEIMG, Genova, IT), CHO-K1/SF designated ECACC 93061607 (CAMR, Salisbury, Wiltshire, UK), and RR-CHOK1 designated ECACC 92052129 (CAMR, Salisbury, Wiltshire, UK). Exemplary plant cells include whole plants, cell culture, or callus, from tobacco, corn, soybean, and rice cells. Corn, soybean, and rice seeds are also acceptable.
  • The CTLA4 mutant molecules of the invention may be isolated as naturally-occurring polypeptides, or from any source whether natural, synthetic, semi-synthetic or recombinant. Accordingly, the CTLA4 mutant polypeptide molecules may be isolated as naturally-occurring proteins from any species, particularly mammalian, including bovine, ovine, porcine, murine, equine, and preferably human. Alternatively, the CTLA4 mutant polypeptide molecules may be isolated as recombinant polypeptides that are expressed in prokaryote or eukaryote host cells, or isolated as a chemically synthesized polypeptide. [0226]
  • A skilled artisan can readily employ standard isolation methods to obtain isolated CTLA4 mutant molecules. The nature and degree of isolation will depend on the source and the intended use of the isolated molecules. [0227]
  • CTLA4 mutant molecules and fragments or derivatives thereof, can be produced by recombinant methods. Accordingly, an isolated nucleotide sequence encoding wild-type CTLA4 molecules may be manipulated to introduce mutations, resulting in nucleotide sequences that encode the CTLA4 mutant polypeptide molecules. For example, the nucleotide sequences encoding the CTLA4 mutant molecules may be generated by site-directed mutagenesis methods, using primers and PCR amplification. The primers can include specific sequences designed to introduce desired mutations. Alternatively, the primers can be designed to include randomized or semi-randomized sequences to introduce random mutations. Standard recombinant methods ([0228] Molecular Cloning; A Laboratory Manual, 2nd edition, Sambrook, Fritch, and Maniatis 1989, Cold Spring Harbor Press) and PCR technology (U.S. Pat. No. 4,603,102) can be employed for generating and isolating CTLA4 mutant polynucleotides encoding CTLA4 mutant polypeptides.
  • The invention includes pharmaceutical compositions comprising pharmaceutically effective amounts of a molecule that blocks B7 interaction with CTLA4 and/or CD28 such as soluble CTLA4 molecules, CD28 molecules, B7 (B7-1 or B7-2) molecules, anti-CTLA4 monoclonal antibodies, anti-CD28 monoclonal antibodies or anti-B7 (B7-1 or B7-2) monoclonal antibodies. The pharmaceutical compositions of the invention are useful for treatment of immune system diseases. In certain embodiments, immune system diseases are mediated by CD28/CTLA4/B7 interactions. The soluble CTLA4 molecules are preferably soluble CTLA4 molecules with wildtype sequence and/or soluble CTLA4 molecules having one or more mutations in the extracellular domain of CTLA4. The pharmaceutical composition can include soluble CTLA4 protein molecules and/or nucleic acid molecules, and/or vectors encoding the molecules. In preferred embodiments, the soluble CTLA4 molecules have the amino acid sequence of the extracellular domain of CTLA4 as shown in either FIG. 24 or [0229] 19 (CTLA4Ig or L104EA29Y, respectively). Even more preferably, the soluble CTLA4 mutant molecule is L104EA29YIg as disclosed herein. The compositions may additionally include other therapeutic agents, including, but not limited to, DMARDs, NSAIDs, corticosteroids, glucocorticoids, drug toxins, alkylating agents, anti-neoplastic drugs, enzymes, antibodies, or conjugates.
  • An embodiment of the pharmaceutical composition of the invention comprises an effective amount of a molecule that blocks B7 interaction with CTLA4 and/or CD28, such as the molecules and the suitable amounts of the molecules described supra, and an effective amount of a DMARD. [0230]
  • The amount of DMARDS administered to a subject varies depending on several factors including the efficacy of the drug on a specific subject and the toxicity (i.e. the tolerability) of a drug to a specific subject (Guidelines for the Management of Rheumatoid Arthritis, Arthritis and Rheumatism Vol. 39, No. 5, May 1996, pages 713-711; Physician's Desk Reference 2002, Medical Economics Company, Inc. Montvale, N.J. 07645). The following provides a range of drug dosages for each DMARD. An attending physician will determine specific dosages for each subject. [0231]
  • Depending on the DMARD, an effective amount can be in a range of about 1 to about 5000 mg/day. This range can be modified to an amount of about 1 to 10 mg/day, about 10 to 50 mg/day, about 50 to 100 mg/day, about 100 to 150 mg/day, about 150 to 200 mg/day, about 200 to 250 mg/day, about 250 to 300 mg/day, about 300 to 350 mg/day, about 350 to 400 mg/day, about 400 to 450 mg/day, about 450 to 500 mg/day, about 500 to 550 mg/day, about 550 to 600 mg/day, about 600 to 650 mg/day, about 650 to 700 mg/day, about 700 to 750 mg/day, about 750 to 800 mg/day, about 800 to 850 mg/day, about 850 to 900 mg/day, about 900 to 950 mg/day, about 950 to 1000 mg/day, about 1000 to 1100 mg/day, about 1100 to 1200 mg/day, about 1200 to 1300 mg/day, about 1300 to 1400 mg/day, about 1400 to 1500 mg/day, about 1500 to 1600 mg/day, about 1600 to 1700 mg/day, about 1700 to 1800 mg/day, about 1800 to 1900 mg/day, about 1900 to 2000 mg/day, about 2000 to 2500 mg/day, about 2500 to 3000 mg/day, about 3000 to 3500 mg/day, about 3500 to 4000 mg/day, about 4000 to 4500 mg/day or about 4500 to 5000 mg/day. It would be clear to one skilled in the art that dosage will vary depending on the particular DMARD being used. Specific examples of appropriate dosages, depending on the DMARD, are described below. [0232]
  • In another embodiment, an effective amount of a DMARD can be in a range of about 0.1 mg/week to 40 mg/week; 0.1 mg/week to 5 mg/week; 5 mg/week to 10 mg/week; 10 mg/week to 30 mg/week; 30 mg/week to 35 mg/week; 0.1 mg/week to 100 mg/week; or 30 mg/week to 50 mg/week. In another embodiment, a DMARD can be administered in an amount of about 50 mg/week or 25 mg twice weekly. It would be clear to one skilled in the art that dosage range will vary depending on the particular DMARD being used, for example see below. [0233]
  • Methotrexate is an antimetabolite molecule that interferes with DNA synthesis, repair and cellular replication. Methotrexate functions as an inhibitor of dihydrofolic acid reductase i.e. it is a folic acid antagonist. Methotrexate is commonly administered in an amount about 0.1 to 40 mg per week with a common dosage ranging about 5 to 30 mg per week. Methotrexate may be administered to a subject in various increments: about 0.1 to 5 mg/week, about 5 to 10 mg/week, about 10 to 15 mg/week, about 15 to 20 mg/week, about 20 to 25 mg/week, about 25 to 30 mg/week, about 30 to 35 mg/week, or about 35 to 40 mg/week. In one embodiment, an effective amount of a DMARD, including methotrexate, is an amount about 10 to 30 mg/week. [0234]
  • Cyclophosphamide, an alkylating agent, may be administered in dosages ranging about 1 to 10 mg/kg body weight per day. [0235]
  • Cyclosporine (e.g. NEORAL[0236] R) also known as Cyclosporin A, is commonly administered in dosages ranging from about 1 to 10 mg/kg body weight per day. Dosages ranging about 2.5 to 4 mg per body weight per day are commonly used.
  • Chloroquine or hydroxychloroquine (e.g. PLAQUENIL[0237] R), is commonly administered in dosages ranging about 100 to 1000 mg daily. Preferred dosages range about 200-600 mg administered daily.
  • Sulfasalazine (e.g., AZULFIDINE EN-tabs[0238] R) is commonly administered in amounts ranging about 50 to 5000 mg per day, with a common dosage of about 2000 to 3000 mg per day for adults. Dosages for children are commonly about 5 to 100 mg/kg of body weight, up to 2 grams per day.
  • Gold salts are formulated for two types of administration: injection or oral. Injectable gold salts are commonly prescribed in dosages about 5 to 100 mg doses every two to four weeks. Orally administered gold salts are commonly prescribed in doses ranging about 1 to 10 mg per day. [0239]
  • D-penicillamine or penicillamine (CUPRIMINE[0240] R) is commonly administered in dosages about 50 to 2000 mg per day, with preferred dosages about 125 mg per day up to 1500 mg per day.
  • Azathioprine is commonly administered in dosages of about 10 to 250 mg per day. Preferred dosages range about 25 to 200 mg per day. [0241]
  • Anakinra (e.g. KINERET[0242] R) is an interleukin-1 receptor antagonist. A common dosage range for anakinra is about 10 to 250 mg per day, with a recommended dosage of about 100 mg per day.
  • Infliximab (REMICADE[0243] R) is a chimeric monoclonal antibody that binds to tumor necrosis factor alpha (TNFα) and inhibits the activity of TNFα. Infliximab is commonly administered in dosages about 1 to 20 mg/kg body weight every four to eight weeks. Dosages of about 3 to 10 mg/kg body weight may be administered every four to eight weeks depending on the subject.
  • Etanercept (e.g. ENBREL[0244] R) is a dimeric fusion protein that binds the tumor necrosis factor (TNF) and blocks its interactions with TNF receptors. Commonly administered dosages of etanercept are about 10 to 100 mg per week for adults with a preferred dosage of about 50 mg per week. Dosages for juvenile subjects range about 0.1 to 50 mg/kg body weight per week with a maximum of about 50 mg per week. For adult patients, etanercept is commonly administered e.g., injected, in 25 mg doses twice weekly e.g., 72-96 hours apart in time.
  • Leflunomide (ARAVA[0245] R) is commonly administered at dosages about 1 and 100 mg per day. A common daily dosage is about 10 to 20 mg per day.
  • A further embodiment of the invention is a pharmaceutical composition comprising an effective amount of a soluble CTLA4, such as CTLA4Ig, and an effective amount of a DMARD, such as methotrexate or etanercept. [0246]
  • A pharmaceutical composition comprising soluble CTLA4 can be used for methods for blocking B7 interaction with CTLA4 and/or CD28; or for treating immune system diseases. Effective amounts of soluble CTLA4 in the pharmaceutical composition range about 0.1 to 100 mg/kg weight of the subject. In another embodiment, the effective amount is an amount about 0.5 to 5 mg/kg weight of a subject, about 5 to 10 mg/kg weight of a subject, about 10 to 15 mg/kg weight of a subject, about 15 to 20 mg/kg weight of a subject, about 20 to 25 mg/kg weight of a subject, about 25 to 30 mg/kg weight of a subject, about 30 to 35 mg/kg weight of a subject, about 35 to 40 mg/kg weight of a subject, about 40 to 45 mg/kg of a subject, about 45 to 50 mg/kg weight of a subject, about 50 to 55 mg/kg weight of a subject, about 55 to 60 mg/kg weight of a subject, about 60 to 65 mg/kg weight of a subject, about 65 to 70 mg/kg weight of a subject, about 70 to 75 mg/kg weight of a subject, about 75 to 80 mg/kg weight of a subject, about 80 to 85 mg/kg weight of a subject, about 85 to 90 mg/kg weight of a subject, about 90 to 95 mg/kg weight of a subject, or about 95 to 100 mg/kg weight of a subject. [0247]
  • In an embodiment, the effective amount of soluble CTLA4 is an amount about 2 mg/kg to about 10 mg/kg weight of a subject. In another embodiment, the effective amount is an amount about 0.1 to 4 mg/kg weight of a subject. In another embodiment the effective amount is an amount about 0.1 to 0.5 mg/kg weight of a subject, about 0.5 to 1.0 mg/kg weight of a subject, about 1.0 to 1.5 mg/kg weight of a subject, about 1.5 to 2.0 mg/kg weight of a subject, about 2.0 to 2.5 mg/kg weight of a subject, about 2.5 to 3.0 mg/kg weight of a subject, about 3.0 to 3.5 mg/kg weight of a subject or about 3.5 to 4.0 mg/kg weight of a subject. In another embodiment, the effective amount is an amount about 0.1 to 20 mg/kg weight of a subject. In another embodiment, the effective amount is an amount about 0.1 to 2 mg/kg weight of a subject, about 2 to 4 mg/kg weight of a subject, about 4 to 6 mg/kg weight of a subject, about 6 to 8 mg/kg weight of a subject, about 8 to 10 mg/kg weight of a subject, about 10 to 12 mg/kg weight of a subject, about 12 to 14 mg/kg weight of a subject, about 14 to 16 mg/kg weight of a subject, about 16 to 18 mg/kg weight of a subject or about 18 to 20 mg/kg weight of a subject. In an embodiment, the effective amount is 2 mg/kg weight of a subject. In another embodiment, the effective amount is about 10 mg/kg weight of a subject. [0248]
  • In a specific embodiment, an effective amount of soluble CTLA4 is 500 mg for a subject weighing less than 60 kg, 750 mg for a subject weighing between 60-100 kg and 1000 mg for a subject weighing more than 100 kg. [0249]
  • Effective amounts of methotrexate in the pharmaceutical composition range about 0.1 to 40 mg/week. In one embodiment, the effective amount is an amount about 0.1 to 5 mg/week, about 5 to 10 mg/week, about 10 to 15 mg/week, about 15 to 20 mg/week, about 20 to 25 mg/week, about 25 to 30 mg/week, about 30 to 35 mg/week, or about 35 to 40 mg/week. In one embodiment, an effective amount of a DMARD, including methotrexate, is an amount about 10 to 30 mg/week. [0250]
  • In one embodiment, the effective amount of a soluble CTLA4 molecule is about 2 mg/kg weight subject and the effective amount of methotrexate is about 10 to 30 mg/week. In another embodiment, the effective amount of a soluble CTLA4 molecule is about 10 mg/kg weight subject and the effective amount of methotrexate is about 10 to 30 mg/week. [0251]
  • Effective amounts of etanercept in the pharmaceutical composition range about 0.1 to 100 mg/week. In one embodiment, the effective amount is ranges about 0.1 to 5 mg/week, about 5 to 10 mg/week, about 10 to 15 mg/week, about 15 to 20 mg/week, about 20 to 25 mg/week, about 25 to 30 mg/week, about 30 to 35 mg/week, about 35 to 40 mg/week, about 40 to 45 mg/week, about 45 to 50 mg/week, about 50 to 55 mg/week, about 55 to 60 mg/week, about 60 to 65 mg/week, about 65 to 70 mg/week, about 70 to 75 mg/week, about 75 to 80 mg/week, about 80 to 85 mg/week, about 85 to 90 mg/week, about 90 to 95 mg/week or about 95 to 100 mg/week. In one embodiment, etanercept is administered in an amount ranging about 50 mg/week e.g., 25 mg administered twice weekly. [0252]
  • In one embodiment, the effective amount of a soluble CTLA4 molecule is about 2 mg/kg weight subject and the effective amount of etanercept is about 25 mg twice a week. In another embodiment, the effective amount of a soluble CTLA4 molecule is about 10 mg/kg weight subject and the effective amount of etanercept is about 25 mg twice a week. [0253]
  • The compositions of the invention further encompass a pharmaceutical composition comprising soluble CTLA4 in combination with other treatments for rheumatic disease including, but not limited to: collagen, dnaJ, molecules that block TNF function (e.g., pegsunercept), molecules that block cytokine function (e.g., AMG719), molecules that block LFA-1 function (e.g., efalizumab) and stem cell transplants. These other treatments are currently being studied in clinical trials (www.clinicaltrials.gov) to determine their effect on rheumatoid arthritis. [0254]
  • Collagen, for example in the form of bovine II collagen, may be orally administered to a patient suffering from rheumatoid arthritis in order to alleviate one or more symptoms of rheumatoid arthritis. [0255]
  • DnaJ is a small peptide which mimics a protein contained in a gene in many patients with rheumatoid arthritis. The peptide is derived from [0256] E. coli bacteria heat shock protein. DnaJ may be orally administered to a patient suffering from rheumatoid arthritis in order to alleviate one or more symptoms of rheumatoid arthritis.
  • TNF is a molecule involved in the inflammatory response of patients with rheumatoid arthritis. Conceivably, any molecule that blocks TNF function e.g., by blocking TNF binding to the TNF receptor (TNFR), may help modify the progression of rheumatoid arthritis and alleviate some of its symptoms. Several TNF blockers such as infliximab and etanercept, have been shown to be efficacious in treating rheumatoid arthritis. Other TNF blockers such as pegsunercept are being developed and tested (Phase II clinical trial) for their efficacy in treating rheumatoid arthritis. [0257]
  • Cytokines e.g., Interleukin-1 (IL-1), are cell secreted molecules involved in mediating immune responses. Conceivably, any molecule that blocks cytokine function e.g., by blocking IL-1 interaction with its receptor, may help modify the progression of rheumatoid arthritis and alleviate one or more of its symptoms. Anakinra, a recombinant protein that blocks IL-1 interaction with its receptor (IL-IR) has been shown to be efficacious in treating rheumatoid arthritis. An IL-1 inhibitor, AMG719, is being developed and tested (Phase II clinical trial) for its efficacy in treating rheumatoid arthritis. [0258]
  • Lymphocyte function associated molecule 1 (LFA-1) is a molecule composed of two subunits, CD11a and CD18, which functions by mediating lymphocyte adhesion to various cell types such as endothelium. Conceivably, interference of LFA-1 function may help modify the progression of rheumatoid arthritis and alleviate one or more of its symptoms. An anti-LFA-1 antibody, efalizumab, is being developed and tested (Phase II clinical trial) for its efficacy in treating rheumatoid arthritis. [0259]
  • Blockage of TNF, cytokine or LFA-1 interaction to their ligands by a potentially therapeutic molecule can be determined by any number of assays known to those skilled in the art. For example, competition assays may be used to test blockage by the molecule of interest e.g., a molecule can be exposed to a TNF/TNFR binding pair in order to compete with TNF to bind to TNFR. Alternatively, functional assays can be performed to test blockage e.g., a molecule can be tested for its ability to inhibit an inflammatory cascade, or any part of an inflammatory reaction such as swelling, redness or pain, caused by a cytokine. [0260]
  • The present invention also provides pharmaceutical compositions comprising the molecules of the invention e.g., CTLA4Ig and an acceptable carrier or adjuvant which is known to those of skill of the art. The pharmaceutical compositions preferably include suitable carriers and adjuvants which include any material which when combined with the molecules of the invention (e.g., a soluble CTLA4 molecule, such as, CTLA4Ig or L104EA29Y) retain the molecule's activity, and is non-reactive with the subject's immune system. These carriers and adjuvants include, but are not limited to, ion exchangers, alumina, aluminum stearate, lecithin, serum proteins, such as human serum albumin, buffer substances such as phosphates, glycine, sorbic acid, potassium sorbate, partial glyceride mixtures of saturated vegetable fatty acids, phosphate buffered saline solution, water, emulsions (e.g. oil-water emulsion), salts or electrolytes such as protamine sulfate, disodium hydrogen phosphate, potassium hydrogen phosphate, sodium chloride, zinc salts, colloidal silica, magnesium trisilicate, polyvinyl pyrrolidone, cellulose-based substances and polyethylene glycol. Other carriers may also include sterile solutions; tablets, including coated tablets and capsules. Typically such carriers contain excipients such as starch, milk, sugar (e.g. sucrose, glucose, maltose), certain types of clay, gelatin, stearic acid or salts thereof, magnesium or calcium stearate, talc, vegetable fats or oils, gums, glycols, or other known excipients. Such carriers may also include flavor and color additives or other ingredients. Compositions comprising such carriers are formulated by well known conventional methods. Such compositions may also be formulated within various lipid compositions, such as, for example, liposomes as well as in various polymeric compositions, such as polymer microspheres. [0261]
  • In a further embodiment of the invention, the present invention provides kits (i.e., a packaged combination of reagents with instructions) containing the molecules of the invention useful for blocking B7 interactions with its ligands and/or for treating an immune system disease. [0262]
  • The kit can contain a pharmaceutical composition that includes one or more agents, for example, a soluble CTLA4 molecule alone, or with a second agent, and an acceptable carrier or adjuvant, e.g., pharmaceutically acceptable buffer, such as phosphate-buffered saline, Ringer's solution and dextrose solution. It may further include other materials desirable from a commercial and user standpoint, including other buffers, diluents, filters, needles, syringes, and package inserts with instructions for use. The agents may be provided as dry powders, usually lyophilized, including excipients that upon dissolving will provide a reagent solution having the appropriate concentration. [0263]
  • Second agents can include the following: steroids, glucocorticoids, drug toxins, alkylating agents, anti-neoplastic drugs, enzymes, antibodies, conjugates, immunosuppressive agents, corticosteroids, DMARDs, nonsteroidal antiinflammatory drugs (NSAIDs), prednisone, azathioprine, methotrexate, TNFα blockers or antagonists, infliximab, any biological agent targeting an inflammatory cytokine, chloroquine, hydroxychloroquine, sulfasalazine (sulphasalazopryine), gold salts, etanercept, anakinra, cyclophosphamide, leflunomide, collagen, dnaJ, a molecule that blocks TNF receptors (e.g., pegsunercept), a molecule that blocks cytokine function (e.g., AMG719), a molecule that blocks LFA-1 function (e.g., efalizumab), acetyl salicylic acid, choline magnesium salicylate, diflunisal, magnesium salicylate, salsalate, sodium salicylate, diclofenac, etodolac, fenoprofen, flurbiprofen, indomethacin, ketoprofen, ketorolac, meclofenamate, naproxen, nabumetone, phenylbutazone, piroxicam, sulindac, tolmetin, acetaminophen, ibuprofen, Cox-2 inhibitors, meloxicam, codeine phosphate, propoxyphene napsylate, oxycodone hydrochloride, oxycodone bitartrate, tramadol, dihydrofolic acid reductase inhibitor, cyclosporine, cyclosporin A or D-penicillamine. [0264]
  • The kit comprises a container with a label and/or instructions. Suitable containers include, for example, bottles, vials, and test tubes. The containers can be formed from a variety of materials such as glass or plastic. The container can have a sterile access port (for example the container can be an intravenous solution bag or a vial having a stopper pierceable by a needle such as a hypodermic injection needle). The container can hold a pharmaceutical composition such as a pharmaceutical composition having an agent that is effective for blocking B7 interactions with its ligand and/or treating an immune system disease. [0265]
  • The kit can also comprise a second container comprising one or more second agents as described herein (e.g., any of the DMARDS or NSAIDS) and/or a pharmaceutically acceptable buffer, such as phosphate-buffered saline, Ringer's solution and dextrose solution. It may further include other materials desirable from a commercial and user standpoint, including other buffers, diluents, filters, needles, syringes, and package inserts with instructions for use. [0266]
  • The kit may also suitably include a label and/or instructions on, or associated with the container. The label can provide directions for carrying out the preparation of the agents for example, dissolving of the dry powders, and/or treatment for a specific immune system disease. [0267]
  • The label and/or the instructions can indicate directions for either in vivo or in vitro use of the pharmaceutical composition. The label and/or the instructions can indicate that the pharmaceutical composition is used alone, or in combination with a second agent. [0268]
  • The label can indicate appropriate dosages for the molecules of the invention. For example, the label can indicate that dosages for a molecule that is effective for blocking B7 interactions with its ligand and/or treating an immune system disease is about 0.1 to 100 mg/kg weight of the subject, about 0.5 to 5 mg/kg weight of a subject, about 5 to 10 mg/kg weight of a subject, about 10 to 15 mg/kg weight of a subject, about 15 to 20 mg/kg weight of a subject, about 20 to 25 mg/kg weight of a subject, about 25 to 30 mg/kg weight of a subject, about 30 to 35 mg/kg weight of a subject, about 35 to 40 mg/kg weight of a subject, about 40 to 45 mg/kg of a subject, about 45 to 50 mg/kg weight of a subject, about 50 to 55 mg/kg weight of a subject, about 55 to 60 mg/kg weight of a subject, about 60 to 65 mg/kg weight of a subject, about 65 to 70 mg/kg weight of a subject, about 70 to 75 mg/kg weight of a subject, about 75 to 80 mg/kg weight of a subject, about 80 to 85 mg/kg weight of a subject, about 85 to 90 mg/kg weight of a subject, about 90 to 95 mg/kg weight of a subject, about 95 to 100 mg/kg weight of a subject, about 2 to 10 mg/kg weight of a subject, about 0.1 to 4 mg/kg weight of a subject, about 0.1 to 0.5 mg/kg weight of a subject, about 0.5 to 1.0 mg/kg weight of a subject, about 1.0 to 1.5 mg/kg weight of a subject, about 1.5 to 2.0 mg/kg weight of a subject, about 2.0 to 2.5 mg/kg weight of a subject, about 2.5 to 3.0 mg/kg weight of a subject, about 3.0 to 3.5 mg/kg weight of a subject, about 3.5 to 4.0 mg/kg weight of a subject, about 4.0 to 4.5 mg/kg weight of a subject, about 4.5 to 5.0 mg/kg weight of a subject, about 5.0 to 5.5 mg/kg weight of a subject, about 5.5 to 6.0 mg/kg weight of a subject, about 6.0 to 6.5 mg/kg weight of a subject, about 6.5 to 7.0 mg/kg weight of a subject, about 7.0 to 7.5 mg/kg weight of a subject, about 7.5 to 8.0 mg/kg weight of a subject, about 8.0 to 8.5 mg/kg weight of a subject, about 8.5 to 9.0 mg/kg weight of a subject, about 9.0 to 9.5 mg/kg weight of a subject, about 9.5 to 10.0 mg/kg weight of a subject, about 0.1 to 2 mg/kg weight of a subject, about 2 to 4 mg/kg weight of a subject, about 4 to 6 mg/kg weight of a subject, about 6 to 8 mg/kg weight of a subject, about 8 to 10 mg/kg weight of a subject, about 10 to 12 mg/kg weight of a subject, about 12 to 14 mg/kg weight of a subject, about 14 to 16 mg/kg weight of a subject, about 16 to 18 mg/kg weight of a subject, about 18 to 20 mg/kg weight of a subject, about 0.5 mg/kg weight of the subject, 2 mg/kg weight of the subject, 10 mg/kg weight of the subject, about 0.5 mg/kg to 100 weight of the subject, about 0.5 to 10 mg/kg weight of a subject, about 0.1 to 20 mg/kg weight of a subject, about 500 mg for a subject weighing less than 60 kg, 750 mg for a subject weighing between 60-100 kg or 1000 mg for a subject weighing more than 100 kg [0269]
  • The label and/or instructions can also indicate dosages for a second agent, such as a DMARD, is about 1 to about 5000 mg/day, about 1 to 10 mg/day, about 10 to 50 mg/day, about 50 to 100 mg/day, about 100 to 150 mg/day, about 150 to 200 mg/day, about 200 to 250 mg/day, about 250 to 300 mg/day, about 300 to 350 mg/day, about 350 to 400 mg/day, about 400 to 450 mg/day, about 450 to 500 mg/day, about 500 to 550 mg/day, about 550 to 600 mg/day, about 600 to 650 mg/day, about 650 to 700 mg/day, about 700 to 750 mg/day, about 750 to 800 mg/day, about 800 to 850 mg/day, about 850 to 900 mg/day, about 900 to 950 mg/day, about 950 to 1000 mg/day, about 1000 to 1100 mg/day, about 1100 to 1200 mg/day, about 1200 to 1300 mg/day, about 1300 to 1400 mg/day, about 1400 to 1500 mg/day, about 1500 to 1600 mg/day, about 1600 to 1700 mg/day, about 1700 to 1800 mg/day, about 1800 to 1900 mg/day, about 1900 to 2000 mg/day, about 2000 to 2500 mg/day, about 2500 to 3000 mg/day, about 3000 to 3500 mg/day, about 3500 to 4000 mg/day, about 4000 to 4500 mg/day or about 4500 to 5000 mg/day. [0270]
  • The label and/or the instructions can also indicate that the pharmaceutical composition can be used alone, or in combination, with a second agent to treat a condition of choice e.g., immune system diseases, autoimmune diseases, immunoproliferative diseases, graft-related disorders, graft versus host disease (GVHD) (e.g., such as may result from bone marrow transplantation, or in the induction of tolerance), immune disorders associated with graft transplantation rejection, immune disorders associated with chronic rejection, immune disorders associated with tissue or cell allo- or xenografts (e.g., kidneys, skin, islets, muscles, hepatocytes, neurons, solid organs and the like), psoriasis, T cell lymphoma, T cell acute lymphoblastic leukemia, testicular angiocentric T cell lymphoma, benign lymphocytic angiitis, as lupus (e.g., lupus erythematosus, lupus nephritis), Hashimoto's thyroiditis, primary myxedema, Graves' disease, pernicious anemia, autoimmune atrophic gastritis, Addison's disease, diabetes (e.g. insulin dependent diabetes mellitus, type I diabetes mellitus, type II diabetes mellitus), good pasture's syndrome, myasthenia gravis, pemphigus, Crohn's disease, sympathetic ophthalmia, autoimmune uveitis, multiple sclerosis, autoimmune hemolytic anemia, idiopathic thrombocytopenia, primary biliary cirrhosis, chronic action hepatitis, ulcerative colitis, Sjogren's syndrome, rheumatic diseases (e.g., rheumatoid arthritis), polymyositis, scleroderma, mixed connective tissue disease, and the like. [0271]
  • In a specific embodiment of the invention, the kit comprises a pharmaceutical composition comprising a pharmaceutically acceptable carrier and an effective amount of a first agent, wherein the first agent is a molecule that blocks B7 interaction with CTLA4 and/or CD28 such as soluble CTLA4 molecules, CD28 molecules, B7 (B7-1 or B7-2) molecules, anti-CTLA4 monoclonal antibodies, anti-CD28 monoclonal antibodies or anti-B7 (B7-1 or B7-2) monoclonal antibodies. In preferred embodiments, the soluble CTLA4 molecules have the amino acid sequence of the extracellular domain of CTLA4 as shown in either FIG. 24 or [0272] 19 (CTLA4Ig or L104EA29Y, respectively).
  • Methods [0273]
  • The invention provides methods for regulating functional CTLA4- and CD28-positive cell interactions with B7-positive cells. The methods comprise contacting the B7-positive cells with a soluble CTLA4 molecule of the invention so as to regulate functional CTLA4- and CD28-positive cell interactions with B7-positive cells, e.g., by interfering with reaction of an endogenous CTLA4 and/or CD28 molecule with a B7 molecule. Suitable amounts of soluble CTLA4 for use in the methods of the invention are described supra. [0274]
  • The present invention also provides methods for inhibiting T-cell function but not T-cell depletion in a human by contacting B7-positive cells in the human with a soluble CTLA4. Examples of soluble CTLA4 include CTLA4Ig and soluble CTLA4 mutant molecule e.g. L104EA29YIg. The present invention further provides methods for treating immune system diseases and auto-immune diseases such as rheumatic diseases. The methods comprise administering a therapeutic composition, comprising soluble CTLA4 molecules of the invention, to a subject in an amount effective to relieve at least one of the symptoms associated with immune system diseases. Additionally, the invention may provide long-term therapy for immune system diseases by blocking the T-cell/B7-positive cell interactions, thereby blocking T-cell activation/stimulation by co-stimulatory signals such as B7 binding to CD28, leading to induction of T-cell anergy or tolerance. Immune system diseases include, but are not limited to, autoimmune diseases, immunoproliferative diseases, and graft-related disorders. Examples of graft-related diseases include graft versus host disease (GVHD) (e.g., such as may result from bone marrow transplantation, or in the induction of tolerance), immune disorders associated with graft transplantation rejection, chronic rejection, and tissue or cell allo- or xenografts, including allo- or xenografts solid organs (e.g., kidneys), skin, islets, muscles, hepatocytes, neurons. Examples of immunoproliferative diseases include, but are not limited to, psoriasis; T cell lymphoma; T cell acute lymphoblastic leukemia; testicular angiocentric T cell lymphoma; benign lymphocytic angiitis; and autoimmune diseases such as lupus (e.g., lupus erythematosus, lupus nephritis), Hashimoto's thyroiditis, primary myxedema, Graves' disease, pernicious anemia, autoimmune atrophic gastritis, Addison's disease, diabetes (e.g. insulin dependent diabetes mellitus, type I diabetes mellitus, type II diabetes mellitus), good pasture's syndrome, myasthenia gravis, pemphigus, Crohn's disease, sympathetic ophthalmia, autoimmune uveitis, multiple sclerosis, autoimmune hemolytic anemia, idiopathic thrombocytopenia, primary biliary cirrhosis, chronic action hepatitis, ulcerative colitis, Sjogren's syndrome, rheumatic diseases (e.g., rheumatoid arthritis), polymyositis, scleroderma, and mixed connective tissue disease. [0275]
  • The soluble CTLA4 molecules of the invention exhibit inhibitory properties in vivo. Under conditions where T-cell/B7-positive cell interactions, for example T cell/B cell interactions, are occurring as a result of contact between T cells and B7-positive cells, binding of introduced CTLA4 molecules to react to B7-positive cells, for example B cells, may interfere, i.e., inhibit, the T cell/B7-positive cell interactions resulting in regulation of immune responses. [0276]
  • The invention provides methods for regulating immune responses. Immune responses downregulated (reduced) by the soluble CTLA4 molecules of the invention may be by way of inhibiting or blocking an immune response already in progress or may involve preventing the induction of an immune response. The soluble CTLA4 molecules of the invention may inhibit the functions of activated T cells, such as T lymphocyte proliferation, cytokine secretion and/or cytokine production, by suppressing T cell responses or by inducing specific tolerance in T cells, or both. Further, the soluble CTLA4 molecules of this invention, interfering with the CTLA4/CD28/B7 pathway may inhibit T-cell proliferation and/or cytokine secretion, and thus result in reduced tissue destruction and induction of T-cell unresponsiveness or anergy. [0277]
  • A preferred embodiment of the invention comprises use of the soluble CTLA4 mutant molecule L104EA29YIg to regulate functional CTLA4- and CD28-positive cell interactions with B7-positive cells, to treat immune system diseases such as rheumatic diseases and/or to downregulate immune responses. The L104EA29YIg of the invention is a soluble CTLA4 mutant molecule comprising at least the two amino acid changes, the leucine (L) to glutamic acid (E) at position +104 and the alanine (A) to tyrosine (Y) change at position +29 (FIG. 19). The L104EA29YIg molecule may encompass further mutations beyond the two specified herein. [0278]
  • A preferred embodiment of the invention comprises use of a molecule to block the interaction of B7 with CTLA4 and/or CD28 in conjunction with a DMARD to regulate an immune response in order to treat an immune system disease such as a rheumatic disease. Suitable amounts of the molecule used to block the B7 interaction with CTLA4 and/or CD28 are described supra. The molecule used to block the B7/CTLA4 interaction may be a soluble CTLA4 such as CTLA4Ig, CTLA4Ig/CD28Ig or L104EA29YIg, a soluble CD28 such as CD28Ig, a soluble B7 (B7-1 or B7-2) such as B7Ig, anti-CTLA4 monoclonal antibodies, anti-CD28 monoclonal antibodies or anti-B7 monoclonal antibodies. The DMARD may be a dihydrofolic acid reductase inhibitor such as methotrexate, cyclophosphamide, cyclosporine, cyclosporin A, chloroquine, hydroxychloroquine, sulfasalazine, sulphasalazopyrine, leflunomide, gold salts, D-penicillamine, azathioprine, anakinra, infliximab, etanercept, TNFα blockers or a biological agent that targets an inflammatory cytokine. [0279]
  • A preferred embodiment includes methods for treating a rheumatic disease, such as rheumatoid arthritis, by administering an effective amount of soluble CTLA4 molecules alone, or in conjunction with an effective amount of methotrexate or a molecule that blocks TNF interactions, to a subject. Administration of an effective amount of the therapeutic composition(s), thereby relieving the subject of at least one of the symptoms associated with the disease, including reducing: joint swelling, joint tenderness, inflammation, morning stiffness, and pain, and structural damage subsequently decreasing the physical disability. The methods of the invention also may be used to reduce at least one symptom associated with rheumatoid arthritis, including reducing erythrocyte sedimentation rates, serum levels of C-reactive protein, soluble ICAM-1, soluble E-selectin and/or soluble IL-2r. [0280]
  • The amount of symptom relief provided by the present invention can be measured using any of the accepted criteria established to measure and document symptom relief in a clinical setting. Acceptable criteria for measuring symptom relief may include scores based on the criteria established by the American College of Rheumatology (e.g., ACR 20), the four measures of symptom relief (in: “CDER Guideline for the Clinical Evaluation of Anti-Inflammatory and Antirheumatic Drugs—FDA 1988), and the Health Assessment Questionnaire (HAQ) (Fries, J. F., et al., 1982 [0281] J of Rheumatology 9:789-793). For a general description of these criteria, see “Guidance for Industry: Clinical Development Programs for Drugs, Devices, and Biological products for the Treatment of Rheumatoid Arthritis (RA)”, February 1999.
  • The present invention provides improving ACR response rates using the methods of the invention. The embodiments of the invention include improving ACR response rates of [0282] ACR 20, 50, and/or 70, using the methods of the invention.
  • The subjects treated by the present invention include mammalian subjects, including, human, monkey, ape, dog, cat, cow, horse, goat, pig, rabbit, mouse and rat. [0283]
  • The present invention provides various methods, local or systemic, for administering the therapeutic compositions of the invention such as soluble CTLA4 molecule alone or in conjunction with a DMARD, such as methotrexate, a molecule that blocks TNF interactions and/or other therapeutic drug. The methods include intravenous, intramuscular, intraperitoneal, oral, inhalation and subcutaneous methods, as well as implantable pump, continuous infusion, gene therapy, liposomes, suppositories, topical contact, vesicles, capsules, biodegradable polymers, hydrogels, controlled release patch and injection methods. The therapeutic agent, compounded with a carrier, is commonly lyophilized for storage and is reconstituted with water or a buffered solution with a neutral pH (about pH 7-8, e.g., pH 7.5) prior to administration. [0284]
  • As is standard practice in the art, the compositions of the invention may be administered to the subject in any pharmaceutically acceptable form. [0285]
  • In accordance with the practice of the invention, the methods comprise administering to a subject the soluble CTLA4 molecules of the invention to regulate CD28- and/or CTLA4-positive cell interactions with B7-positive cells. The B7-positive cells are contacted with an effective amount of the soluble CTLA4 molecules of the invention, or fragments or derivatives thereof, so as to form soluble CTLA4/B7 complexes. Suitable amounts of soluble CTLA4 are described supra. The complexes interfere with interaction between endogenous CTLA4 and CD28 molecules with B7 family molecules. [0286]
  • The soluble CTLA4 molecules may be administered to a subject in an amount and for a time (e.g. length of time and/or multiple times) sufficient to block endogenous B7 molecules from binding their respective ligands, in the subject. Blockage of endogenous B7/ligand binding thereby inhibiting interactions between B7-positive cells with CD28-and/or CTLA4-positive cells. In an embodiment, soluble CTLA4 may be administered to a subject daily, weekly, monthly and/or yearly, in single or multiple times per day/week/month/year, depending on need. For example, in one embodiment, the molecule may initially be administered once every two weeks for a month, and then once every month thereafter. [0287]
  • Dosage of a therapeutic agent is dependant upon many factors including, but not limited to, the type of tissue affected, the type of autoimmune disease being treated, the severity of the disease, a subject's health and response to the treatment with the agents. Accordingly, dosages of the agents can vary depending on each subject and the mode of administration. The soluble CTLA4 molecules may be administered in an amount from about 0.1 to 100 mg/kg weight of the patient/day. Suitable amounts of soluble CTLA4 are described supra. Methotrexate may be administered to a subject in an amount from about 0.1 to 100 mg/week. Suitable amounts of soluble methotrexate are described supra. A molecule that blocks TNF interactions e.g., etanercept, may be administered to a subject in an amount from about 0.1 to 100 mg/week. Suitable amounts of TNF blockers are described supra. [0288]
  • The invention also encompasses the use of the compositions of the invention together with other pharmaceutical agents to treat immune system diseases. For example, rheumatic diseases may be treated with molecules of the invention in conjunction with, but not limited to, immunosuppressants such as corticosteroids, cyclosporin (Mathiesen 1989 [0289] Cancer Lett. 44(2):151-156), prednisone, azathioprine, (R. Handschumacher, in: “Drugs Used for Immunosuppression” pages 1264-1276), TNFα blockers or antagonists (New England Journal of Medicine, vol. 340: 253-259, 1999; The Lancet vol. 354: 1932-39, 1999, Annals of Internal Medicine, vol. 130: 478-486), or any other biological agent targeting any inflammatory cytokine, nonsteroidal antiinflammatory drugs/Cox-2 inhibitors, hydroxychloroquine, sulphasalazopryine, gold salts, etanercept, infliximab, rapamycin, mycophenolate mofetil, azathioprine, tacrolismus, basiliximab, cytoxan, interferon beta-1a, interferon beta-1b, glatiramer acetate, mitoxantrone hydrochloride, anakinra and/or other biologics.
  • The soluble CTLA4 molecules (preferably, L104EA29YIg) can also be used in combination with one or more of the following agents to regulate an immune response: soluble gp39 (also known as CD40 ligand (CD40L), CD154, T-BAM, TRAP), soluble CD29, soluble CD40, soluble CD80 (e.g. ATCC 68627), soluble CD86, soluble CD28 (e.g. ATCC accession number 68628), soluble CD56, soluble Thy-1, soluble CD3, soluble TCR, soluble VLA-4, soluble VCAM-1, soluble LECAM-1, soluble ELAM-1, soluble CD44, antibodies reactive with gp39 (e.g. ATCC HB-10916, ATCC HB-12055 and ATCC HB-12056), antibodies reactive with CD40 (e.g. ATCC HB-9110), antibodies reactive with B7 (e.g. ATCC HB-253, ATCC CRL-2223, ATCC CRL-2226, ATCC HB-301, ATCC HB-11341, etc), antibodies reactive with CD28 (e.g. ATCC HB-11944 or mAb 9.3 as described by Martin et al (J. Clin. Immun. 4(1):18-22, 1980), antibodies reactive with LFA-1 (e.g. ATCC HB-9579 and ATCC TIB-213), antibodies reactive with LFA-2, antibodies reactive with IL-2, antibodies reactive with IL-12, antibodies reactive with IFN-gamma, antibodies reactive with CD2, antibodies reactive with CD48, antibodies reactive with any ICAM (e.g., ICAM-1 (ATCC CRL-2252), ICAM-2 and ICAM-3), antibodies reactive with CTLA4 (e.g. ATCC HB-304), antibodies reactive with Thy-1, antibodies reactive with CD56, antibodies reactive with CD3, antibodies reactive with CD29, antibodies reactive with TCR, antibodies reactive with VLA-4, antibodies reactive with VCAM-1, antibodies reactive with LECAM-1, antibodies reactive with ELAM-1, antibodies reactive with CD44. In certain embodiments, monoclonal antibodies are preferred. In other embodiments, antibody fragments are preferred. As persons skilled in the art will readily understand, the combination can include: the soluble CTLA4 molecules of the invention and one other immunosuppressive agent; the soluble CTLA4 molecules with two other immunosuppressive agents; the soluble CTLA4 molecules with three other immunosuppressive agents; and the like. The determination of the optimal combination and dosages can be determined and optimized using methods well known in the art. [0290]
  • Some specific combinations include the following: L104EA29YIg and CD80 monoclonal antibodies (mAbs); L104EA29YIg and CD86 mAbs; L104EA29YIg, CD80 mAbs, and CD86 mAbs; L104EA29YIg and gp39 mAbs; L104EA29YIg and CD40 mAbs; L104EA29YIg and CD28 mAbs; L104EA29YIg, CD80 and CD86 mAbs, and gp39 mAbs; L104EA29YIg, CD80 and CD86 mAbs and CD40 mAbs; and L104EA29YIg, anti-LFA1 mAb, and anti-gp39 mAb. A specific example of a gp39 mAb is MR1. Other combinations will be readily appreciated and understood by persons skilled in the art. [0291]
  • The soluble CTLA4 molecules of the invention, for example L104EA29YIg, may be administered as the sole active ingredient or together with other drugs in immunomodulating regimens or other anti-inflammatory agents such as DMARDs e.g. for the treatment or prevention of allo- or xenograft acute or chronic rejection or inflammatory or autoimmune disorders, or to induce tolerance. For example, it may be used in combination with a calcineurin inhibitor, e.g. cyclosporin A or FK506; an immunosuppressive macrolide, e.g. rapamycine or a derivative thereof (e.g. 40-O-(2-hydroxy)ethyl-rapamycin); a lymphocyte homing agent, e.g. FTY720 or an analog thereof; corticosteroids; cyclophosphamide; azathioprene; a dihydrofolic acid reductase inhibitor such as methotrexate; leflunomide or an analog thereof; mizoribine; mycophenolic acid; mycophenolate mofetil; 15-deoxyspergualine or an analog thereof, immunosuppressive monoclonal antibodies, e.g., monoclonal antibodies to leukocyte receptors, e.g., MHC, CD2, CD3, CD4, CD11a/CD18, CD7, CD25, [0292] CD 27, B7, CD40, CD45, CD58, CD137, ICOS, CD150 (SLAM), OX40, 4-1BB or their ligands; or other immunomodulatory compounds, e.g. CTLA4/CD28-Ig, or other adhesion molecule inhibitors, e.g. mAbs or low molecular weight inhibitors including LFA-1 antagonists, Selectin antagonists and VLA-4 antagonists. The compound is particularly useful in combination with a compound that interferes with CD40 and its ligand, e.g. antibodies to CD40 and antibodies to CD40-L.
  • Where the soluble CTLA4 mutant molecules of the invention are administered in conjunction with other immunosuppressive/immunomodulatory or anti-inflammatory therapy, e.g. as hereinabove specified, dosages of the co-administered immunosuppressant, immunomodulatory or anti-inflammatory compound will of course vary depending on the type of co-drug employed, e.g. whether it is a steroid or a cyclosporin, on the specific drug employed, on the condition being treated and so forth. [0293]
  • In accordance with the foregoing the present invention provides in a yet further aspect methods as defined above comprising co-administration, e.g. concomitantly or in sequence, of a therapeutically effective amount of soluble CTLA4 molecules of the invention, e.g. CTLA4Ig and/or L104EA29YIg, in free form or in pharmaceutically acceptable salt form, and a second drug substance, said second drug substance being an immunosuppressant, immunomodulatory or anti-inflammatory drug, e.g. as indicated above. [0294]
  • Further provided are therapeutic combinations, e.g. a kit, comprising a soluble CTLA4 molecule, in free form or in pharmaceutically acceptable salt form, to be used concomitantly or in sequence with at least one pharmaceutical composition comprising an immunosuppressant, immunomodulatory or anti-inflammatory drug e.g., a DMARD, NSAID, glucocorticoid or corticosteroid. The kit may comprise instructions for its administration. The kits of the invention can be used in any method of the present invention. [0295]
  • The invention also provides methods for producing the soluble CTLA4 mutant molecules of the invention. Expression of soluble CTLA4 mutant molecules can be in prokaryotic cells or eukaryotic cells. [0296]
  • Prokaryotes most frequently are represented by various strains of bacteria. The bacteria may be a gram positive or a gram negative. Typically, gram-negative bacteria such as [0297] E. coli are preferred. Other microbial strains may also be used. Sequences encoding soluble CTLA4 mutant molecules can be inserted into a vector designed for expressing foreign sequences in prokaryotic cells such as E. coli. These vectors can include commonly used prokaryotic control sequences which are defined herein to include promoters for transcription initiation, optionally with an operator, along with ribosome binding site sequences, including such commonly used promoters as the beta-lactamase (penicillinase) and lactose (lac) promoter systems (Chang, et al., (1977) Nature 198:1056), the tryptophan (trp) promoter system (Goeddel, et al., (1980) Nucleic Acids Res. 8:4057) and the lambda derived PL promoter and N-gene ribosome binding site (Shimatake, et al., (1981) Nature 292:128).
  • Such expression vectors will also include origins of replication and selectable markers, such as a beta-lactamase or neomycin phosphotransferase gene conferring resistance to antibiotics, so that the vectors can replicate in bacteria and cells carrying the plasmids can be selected for when grown in the presence of antibiotics, such as ampicillin or kanamycin. [0298]
  • The expression plasmid can be introduced into prokaryotic cells via a variety of standard methods, including but not limited to CaCl[0299] 2-shock (Cohen, (1972) Proc. Natl. Acad. Sci. USA 69:2110, and Sambrook et al. (eds.), “Molecular Cloning: A Laboratory Manual”, 2nd Edition, Cold Spring Harbor Press, (1989)) and electroporation.
  • In accordance with the practice of the invention, eukaryotic cells are also suitable host cells. Examples of eukaryotic cells include any animal cell, whether primary or immortalized, yeast (e.g., [0300] Saccharomyces cerevisiae, Schizosaccharomyces pombe, and Pichia pastoris), and plant cells. Myeloma, COS and CHO cells are examples of animal cells that may be used as hosts. Particular CHO cells include, but are not limited to, DG44 (Chasin, et la., 1986 Som. Cell. Molec. Genet. 12:555-556; Kolkekar 1997 Biochemistry 36:10901-10909), CHO-K1 (ATCC No. CCL-61), CHO-K1 Tet-On cell line (Clontech), CHO designated ECACC 85050302 (CAMR, Salisbury, Wiltshire, UK), CHO clone 13 (GEIMG, Genova, IT), CHO clone B (GEIMG, Genova, IT), CHO-K1/SF designated ECACC 93061607 (CAMR, Salisbury, Wiltshire, UK), and RR-CHOK1 designated ECACC 92052129 (CAMR, Salisbury, Wiltshire, UK). Exemplary plant cells include tobacco (whole plants, cell culture, or callus), corn, soybean, and rice cells. Corn, soybean, and rice seeds are also acceptable.
  • Nucleic acid sequences encoding the CTLA4 mutant molecules can also be inserted into a vector designed for expressing foreign sequences in an eukaryotic host. The regulatory elements of the vector can vary according to the particular eukaryotic host. [0301]
  • Commonly used eukaryotic control sequences for use in expression vectors include promoters and control sequences compatible with mammalian cells such as, for example, CMV promoter (CDM8 vector) and avian sarcoma virus (ASV) (πrLN vector). Other commonly used promoters include the early and late promoters from Simian Virus 40 (SV40) (Fiers, et al., (1973) [0302] Nature 273:113), or other viral promoters such as those derived from polyoma, Adenovirus 2, and bovine papilloma virus. An inducible promoter, such as hMTII (Karin, et al., (1982) Nature 299:797-802) may also be used.
  • Vectors for expressing CTLA4 mutant molecules in eukaryotes may also carry sequences called enhancer regions. These are important in optimizing gene expression and are found either upstream or downstream of the promoter region. [0303]
  • Examples of expression vectors for eukaryotic host cells include, but are not limited to, vectors for mammalian host cells (e.g., BPV-1, pHyg, pRSV, pSV2, pTK2 (Maniatis); pIRES (Clontech); pRc/CMV2, pRc/RSV, pSFV1 (Life Technologies); pVPakc Vectors, pCMV vectors, pSG5 vectors (Stratagene)), retroviral vectors (e.g., pFB vectors (Stratagene)), pcDNA-3 (Invitrogen) or modified forms thereof, adenoviral vectors; Adeno-associated virus vectors, baculovirus vectors, yeast vectors (e.g., pESC vectors (Stratagene)). [0304]
  • Nucleic acid sequences encoding CTLA4 mutant molecules can integrate into the genome of the eukaryotic host cell and replicate as the host genome replicates. Alternatively, the vector carrying CTLA4 mutant molecules can contain origins of replication allowing for extrachromosomal replication. [0305]
  • For expressing the nucleic acid sequences in [0306] Saccharomyces cerevisiae, the origin of replication from the endogenous yeast plasmid, the 2μ circle can be used. (Broach, (1983) Meth. Enz. 101:307). Alternatively, sequences from the yeast genome capable of promoting autonomous replication can be used (see, for example, Stinchcomb et al., (1979) Nature 282:39); Tschemper et al., (1980) Gene 10:157; and Clarke et al., (1983) Meth. Enz. 101:300).
  • Transcriptional control sequences for yeast vectors include promoters for the synthesis of glycolytic enzymes (Hess et al., (1968) [0307] J. Adv. Enzyme Reg. 7:149; Holland et al., (1978) Biochemistry 17:4900). Additional promoters known in the art include the CMV promoter provided in the CDM8 vector (Toyama and Okayama, (1990) FEBS 268:217-221); the promoter for 3-phosphoglycerate kinase (Hitzeman et al., (1980) J. Biol. Chem. 255:2073), and those for other glycolytic enzymes.
  • Other promoters are inducible because they can be regulated by environmental stimuli or by the growth medium of the cells. These inducible promoters include those from the genes for heat shock proteins, [0308] alcohol dehydrogenase 2, isocytochrome C, acid phosphatase, enzymes associated with nitrogen catabolism, and enzymes responsible for maltose and galactose utilization.
  • Regulatory sequences may also be placed at the 3′ end of the coding sequences. These sequences may act to stabilize messenger RNA. Such terminators are found in the 3′ untranslated region following the coding sequences in several yeast-derived and mammalian genes. [0309]
  • Exemplary vectors for plants and plant cells include, but are not limited to, Agrobacterium T[0310] i plasmids, cauliflower mosaic virus (CaMV), and tomato golden mosaic virus (TGMV).
  • General aspects of mammalian cell host system transformations have been described by Axel (U.S. Pat. No. 4,399,216 issued Aug. 16, 1983). Mammalian cells can be transformed by methods including but not limited to, transfection in the presence of calcium phosphate, microinjection, electroporation, or via transduction with viral vectors. [0311]
  • Methods for introducing foreign DNA sequences into eukaryote genomes, including plant and yeast genomes include; (1) mechanical methods, such as microinjection of DNA into single cells or protoplasts, vortexing cells with glass beads in the presence of DNA, or shooting DNA-coated tungsten or gold spheres into cells or protoplasts; (2) introducing DNA by making cell membranes permeable to macromolecules through polyethylene glycol treatment or subjection to high voltage electrical pulses (electroporation); or (3) the use of liposomes (containing cDNA) which fuse to cell membranes. [0312]
  • Once the CTLA4 mutant molecules of the inventions are expressed, they can be harvested by methods well known in the art such as cell lysis (e.g. sonication, lysozyme and/or detergents) and protein recovery performed using standard protein purification means, e.g., affinity chromatography or ion-exchange chromatography, to yield substantially pure product (R. Scopes in: “[0313] Protein Purification, Principles and Practice”, Third Edition, Springer-Verlag (1994); Sambrook et al. (eds.), “Molecular Cloning: A Laboratory Manual”, 2nd Edition, Cold Spring Harbor Press, (1989)). Expression of CTLA4 mutant molecules can be detected by methods known in the art. For example, the mutant molecules can be detected by Coomassie staining SDS-PAGE gels and immunoblotting using antibodies that bind CTLA4.
  • The following examples are presented to illustrate the present invention and to assist one of ordinary skill in making and using the same. The examples are not intended in any way to otherwise limit the scope of the invention. [0314]
  • EXAMPLE 1
  • The following provides a description of the methods used to generate the nucleotide sequences encoding the CTLA4 molecules of the invention. [0315]
  • A CTLA4Ig encoding plasmid was first constructed, and shown to express CTLA4Ig molecules as described in U.S. Pat. Nos. 5,434,131, 5,885,579 and 5,851,795. Then single-site mutant molecules (e.g., L104EIg) were generated from the CTLA4Ig encoding sequence, expressed and tested for binding kinetics for various B7 molecules. The L104EIg nucleotide sequence (as included in the sequence shown in FIG. 18) was used as a template to generate the double-site CTLA4 mutant sequences (as included in the sequences shown in FIGS. [0316] 19-22) which were expressed as proteins and tested for binding kinetics. The double-site CTLA4 mutant sequences include: L104EA29YIg, L104EA29LIg, L104EA29TIg, and L104EA29WIg. Triple-site mutants were also generated.
  • CTLA4Ig Construction [0317]
  • A genetic construct encoding CTLA4Ig comprising the extracellular domain of CTLA4 and an IgCgamma1 domain was constructed as described in U.S. Pat. Nos. 5,434,131, 5,844,095 and 5,851,795, the contents of which are incorporated by reference herein. The extracellular domain of the CTLA4 gene was cloned by PCR using synthetic oligonucleotides corresponding to the published sequence (Dariavach et al., [0318] Eur. Journ. Immunol. 18:1901-1905 (1988)).
  • Because a signal peptide for CTLA4 was not identified in the CTLA4 gene, the N-terminus of the predicted sequence of CTLA4 was fused to the signal peptide of oncostatin M (Malik et al., [0319] Mol. and Cell. Biol. 9:2847 (1989)) in two steps using overlapping oligonucleotides. For the first step, the oligonucleotide, CTCAGTCTGGTCCTTGCACTCCTGTTTCCAAGCATGGCGAGCATGGCAATGCA CGTGGCCCAGCC (SEQ ID NO: 1) (which encoded the C terminal 15 amino acids from the oncostatin M signal peptide fused to the N terminal 7 amino acids of CTLA4) was used as forward primer, and TTTGGGCTCCTGATCAGAATCTGGGCACGGTTG (SEQ ID NO: 2) (encoding amino acid residues 119-125 of the amino acid sequence encoding CTLA4 receptor and containing a Bcl I restriction enzyme site) as reverse primer. The template for this step was cDNA synthesized from 1 micro g of total RNA from H38 cells (an HTLV II infected T-cell leukemic cell line provided by Drs. Salahudin and Gallo, NCI, Bethesda, Md.). A portion of the PCR product from the first step was reamplified, using an overlapping forward primer, encoding the N terminal portion of the oncostatin M signal peptide and containing a Hind III restriction endonuclease site, CTAGCCACTGAAGCTTCACCAATGGGTGTACTGCTCACACAGAGGACGCTGCTCAGTCTGGTCCTTGCACTC (SEQ ID NO: 3) and the same reverse primer. The product of the PCR reaction was digested with Hind III and Bcl I and ligated together with a Bcl 1/Xba I cleaved cDNA fragment encoding the amino acid sequences corresponding to the hinge, CH2 and CH3 regions of IgC(gamma)1 into the Hind III/Xba I cleaved expression vector, CDM8 or Hind III/Xba I cleaved expression vector piLN (also known as 7πLN).
  • DNA encoding the amino acid sequence corresponding to CTLA4Ig has been deposited with the ATCC under the Budapest Treaty on May 31, 1991, and has been accorded ATCC accession number 68629. [0320]
  • CTLA4Ig Codon Based Mutagenesis: [0321]
  • A mutagenesis and screening strategy was developed to identify mutant CTLA4Ig molecules that had slower rates of dissociation (“off” rates) from CD80 and/or CD86 molecules i.e. improved binding ability. In this embodiment, mutations were carried out in and/or about the residues in the CDR-1, CDR-2 (also known as the C′ strand) and/or CDR-3 regions of the extracellular domain of CTLA4 (as described in U.S. Pat. Nos. 6,090,914, 5,773,253 and 5,844,095; in copending U.S. Patent Application Serial No. 60/214,065; and by Peach, R. J., et al [0322] J Exp Med 1994 180:2049-2058. A CDR-like region encompasses the each CDR region and extends, by several amino acids, upstream and/or downstream of the CDR motif). These sites were chosen based on studies of chimeric CD28/CTLA4 fusion proteins (Peach et al., J. Exp. Med., 1994, 180:2049-2058), and on a model predicting which amino acid residue side chains would be solvent exposed, and a lack of amino acid residue identity or homology at certain positions between CD28 and CTLA4. Also, any residue which is spatially in close proximity (5 to 20 Angstrom Units) to the identified residues is considered part of the present invention.
  • To synthesize and screen soluble CTLA4 mutant molecules with altered affinities for a B7 molecule (e.g. CD80, CD86), a two-step strategy was adopted. The experiments entailed first generating a library of mutations at a specific codon of an extracellular portion of CTLA4 and then screening these by BIAcore analysis to identify mutants with altered reactivity to B7. The Biacore assay system (Pharmacia, Piscataway, N.J.) uses a surface plasmon resonance detector system that essentially involves covalent binding of either CD80Ig or CD86Ig to a dextran-coated sensor chip which is located in a detector. The test molecule can then be injected into the chamber containing the sensor chip and the amount of complementary protein that binds can be assessed based on the change in molecular mass which is physically associated with the dextran-coated side of the sensor chip; the change in molecular mass can be measured by the detector system. [0323]
  • Specifically, single-site mutant nucleotide sequences were generated using non-mutated (e.g., wild-type) DNA encoding CTLA4Ig (U.S. Pat. Nos. 5,434,131, 5,844,095; 5,851,795; and 5,885,796; ATCC Accession No. 68629) as a template. Mutagenic oligonucleotide PCR primers were designed for random mutagenesis of a specific codon by allowing any base at [0324] positions 1 and 2 of the codon, but only guanine or thymine at position 3 (XXG/T or also noted as NNG/T). In this manner, a specific codon encoding an amino acid could be randomly mutated to code for each of the 20 amino acids. In that regard, XXG/T mutagenesis yields 32 potential codons encoding each of the 20 amino acids. PCR products encoding mutations in close proximity to the CDR3-like loop of CTLA4Ig (MYPPPY), were digested with SacI/XbaI and subcloned into similarly cut CTLA4Ig (as included in FIG. 24) πLN expression vector. This method was used to generate the single-site CTLA4 mutant molecule L104EIg (as included in FIG. 18).
  • For mutagenesis in proximity to the CDR-1-like loop of CTLA4Ig, a silent NheI restriction site was first introduced 5′ to this loop, by PCR primer-directed mutagenesis. PCR products were digested with NheI/XbaI and subcloned into similarly cut CTLA4Ig or L104EIg expression vectors. This method was used to generate the double-site CTLA4 mutant molecule L104EA29YIg (as included in FIG. 19). In particular, the nucleic acid molecule encoding the single-site CTLA4 mutant molecule, L104EIg, was used as a template to generate the double-site CTLA4 mutant molecule, L104EA29YIg. [0325]
  • The double-site mutant nucleotide sequences encoding CTLA4 mutant molecules, such as L104EA29YIg (deposited on Jun. 19, 2000 with the American Type Culture Collection (ATCC), 10801 University Blvd., Manassas, Va. 20110-2209 and accorded ATCC accession number PTA-2104), were generated by repeating the mutagenesis procedure described above using L104EIg as a template. This method was used to generate numerous double-site mutants nucleotide sequences such as those encoding CTLA4 molecules L104EA29YIg (as included in the sequence shown in FIG. 19), L104EA29LIg (as included in the sequence shown in FIG. 20), L104EA29TIg (as included in the sequence shown in FIG. 21), and L104EA29WIg (as included in the sequence shown in FIG. 22). Triple-site mutants, such as those encoding L104EA29YS25KIg, L104EA29YS25NIg and L104EA29YS25RIg, were also generated [0326]
  • The soluble CTLA4 molecules were expressed from the nucleotide sequences and used in the phase II clinical studies described in Example 3, infra. [0327]
  • As those skilled-in-the-art will appreciate, replication of nucleic acid sequences, especially by PCR amplification, easily introduces base changes into DNA strands. However, nucleotide changes do not necessarily translate into amino acid changes as some codons redundantly encode the same amino acid. Any changes of nucleotide from the original or wildtype sequence, silent (i.e. causing no change in the translated amino acid) or otherwise, while not explicitly described herein, are encompassed within the scope of the invention. [0328]
  • EXAMPLE 2
  • The following example provides a description of the screening methods used to identify the single- and double-site mutant CTLA polypeptides, expressed from the constructs described in Example 1, that exhibited a higher binding avidity for B7 molecules, compared to non-mutated CTLA4Ig molecules. [0329]
  • Current in vitro and in vivo studies indicate that CTLA4Ig by itself is unable to completely block the priming of antigen specific activated T cells. In vitro studies with CTLA4Ig and either monoclonal antibody specific for CD80 or CD86 measuring inhibition of T cell proliferation indicate that anti-CD80 monoclonal antibody did not augment CTLA4Ig inhibition. However, anti-CD86 monoclonal antibody did augment the inhibition, indicating that CTLA4Ig was not as effective at blocking CD86 interactions. These data support earlier findings by Linsley et al. ([0330] Immunity, (1994), 1:793-801) showing inhibition of CD80-mediated cellular responses required approximately 100 fold lower CTLA4Ig concentrations than for CD86-mediated responses. Based on these findings, it was surmised that soluble CTLA4 mutant molecules having a higher avidity for CD86 than wild type CTLA4 should be better able to block the priming of antigen specific activated cells than CTLA4Ig.
  • To this end, the soluble CTLA4 mutant molecules described in Example 1 above were screened using a novel screening procedure to identify several mutations in the extracellular domain of CTLA4 that improve binding avidity for CD80 and CD86. This screening strategy provided an effective method to directly identify mutants with apparently slower “off” rates without the need for protein purification or quantitation since “off” rate determination is concentration independent (O'Shannessy et al., (1993) [0331] Anal. Biochem., 212:457-468).
  • COS cells were transfected with individual miniprep purified plasmid DNA and propagated for several days. Three day conditioned culture media was applied to BIAcore biosensor chips (Pharmacia Biotech AB, Uppsala, Sweden) coated with soluble CD80Ig or CD86Ig. The specific binding and dissociation of mutant proteins was measured by surface plasmon resonance (O'Shannessy, D. J., et al., 1997 [0332] Anal. Biochem. 212:457-468). All experiments were run on BIAcore™ or BIAcore™ 2000 biosensors at 25° C. Ligands were immobilized on research grade NCM5 sensor chips (Pharmacia) using standard N-ethyl-N′-(dimethylaminopropyl) carbodiimidN-hydroxysuccinimide coupling (Johnsson, B., et al. (1991) Anal. Biochem. 198: 268-277; Khilko, S. N., et al.(1993) J. Biol. Chem 268:5425-15434).
  • Screening Method [0333]
  • COS cells grown in 24 well tissue culture plates were transiently transfected with mutant CTLA4Ig. Culture media containing secreted soluble mutant CTLA4Ig was collected 3 days later. [0334]
  • Conditioned COS cell culture media was allowed to flow over BIAcore biosensor chips derivitized with CD86Ig or CD80Ig (as described in Greene et al., 1996 [0335] J. Biol. Chem. 271:26762-26771), and mutant molecules were identified with off-rates slower than that observed for wild type CTLA4Ig. The DNAs corresponding to selected media samples were sequenced and more DNA prepared to perform larger scale COS cell transient transfection, from which CTLA4Ig mutant protein was prepared following protein A purification of culture media.
  • BIAcore analysis conditions and equilibrium binding data analysis were performed as described in J. Greene et al. 1996 [0336] J. Biol. Chem. 271:26762-26771 and in U.S. patent application Ser. Nos. 09/579,927, and 60/214,065 which are herein incorporated by reference.
  • BIAcore Data Analysis [0337]
  • Senosorgram baselines were normalized to zero response units (RU) prior to analysis. Samples were run over mock-derivatized flow cells to determine background RU values due to bulk refractive index differences between solutions. Equilibrium dissociation constants (K[0338] d) were calculated from plots of Req versus C, where Req is the steady-state response minus the response on a mock-derivatized chip, and C is the molar concentration of analyte. Binding curves were analyzed using commercial nonlinear curve-fitting software (Prism, GraphPAD Software).
  • Experimental data were first fit to a model for a single ligand binding to a single receptor (1-site model, i.e., a simple langmuir system, A+B[0339]
    Figure US20040022787A1-20040205-P00900
    AB), and equilibrium association constants (Kd=[A]·[B]\[AB]) were calculated from the equation R=Rmax·C/(Kd+C). Subsequently, data were fit to the simplest two-site model of ligand binding (i.e., to a receptor having two non-interacting independent binding sites as described by the equation R=Rmax1·C\(Kd1+C)+Rmax2·C\(Kd2+C).
  • The goodness-of-fits of these two models were analyzed visually by comparison with experimental data and statistically by an F test of the sums-of-squares. The simpler one-site model was chosen as the best fit, unless the two-site model fit significantly better (p<0.1). [0340]
  • Association and disassociation analyses were performed using BIA evaluation 2.1 Software (Pharmacia). Association rate constants k[0341] on were calculated in two ways, assuming both homogenous single-site interactions and parallel two-site interactions. For single-site interactions, kon values were calculated according to the equation Rt=Req(1−exp−ks(t−t 0), where Rt is a response at a given time, t; Req is the steady-state response; to is the time at the start of the injection; and ks=dR/dt=kon·Ckoff, where C is a concentration of analyte, calculated in terms of monomeric binding sites. For two-site interactions kon values were calculated according to the equation Rt=Req1(1−exp−ks1(t−t 0)+Req2(1−expks2(t−t 0). For each model, the values of kon were determined from the calculated slope (to about 70% maximal association) of plots of ks versus C.
  • Dissociation data were analyzed according to one site (AB=A+B) or two site (AiBj=Ai+Bj) models, and rate constants (k[0342] off) were calculated from best fit curves. The binding site model was used except when the residuals were greater than machine background (2-10RU, according to machine), in which case the two-binding site model was employed. Half-times of receptor occupancy were calculated using the relationship t1/2=0.693/koff.
  • Flow Cytometry: [0343]
  • Murine mAb L307.4 (anti-CD80) was purchased from Becton Dickinson (San Jose, Calif.) and IT2.2 (anti-B7-0 [also known as CD86]), from Pharmingen (San Diego, Calif.). For immunostaining, CD80-positive and/or CD86-positive CHO cells were removed from their culture vessels by incubation in phosphate-buffered saline (PBS) containing 10 mM EDTA. CHO cells (1-10×[0344] 10 5) were first incubated with mAbs or immunoglobulin fusion proteins in DMEM containing 10% fetal bovine serum (FBS), then washed and incubated with fluorescein isothiocyanate-conjugated goat anti-mouse or anti-human immunoglobulin second step reagents (Tago, Burlingame, Calif.). Cells were given a final wash and analyzed on a FACScan (Becton Dickinson).
  • SDS-PAGE and Size Exclusion Chromatography [0345]
  • SDS-PAGE was performed on Tris/glycine 4-20% acrylamide gels (Novex, San Diego, Calif.). Analytical gels were stained with Coomassie Blue, and images of wet gels were obtained by digital scanning. CTLA4Ig (25 μg) and L104EA29YIg (25 μg) were analyzed by size exclusion chromatography using a TSK-GEL G300 SW[0346] XL column (7.8×300 mm, Tosohaas, Montgomeryville, Pa.) equilibrated in phosphate buffered saline containing 0.02% NAN3 at a flow rate of 1.0 ml/min.
  • CTLA4X[0347] C120S and L104EA29YXX120S.
  • Single chain CTLA4X[0348] C120S was prepared as previously described (Linsley et al., (1995) J. Biol. Chem., 270:15417-15424). Briefly, an oncostatin M CTLA4 (OMCTLA4) expression plasmid was used as a template, the forward primer, GAGGTGATAAAGCTTCACCAATGGGTGTACTGCTCACACAG (SEQ ID NO: 4) was chosen to match sequences in the vector; and the reverse primer, GTGGTGTATTGGTCTAGATCAATCAGAATCTGGGCACGGTTC (SEQ ID NO: 5) corresponded to the last seven amino acids (i.e. amino acids 118-124) in the extracellular domain of CTLA4, and contained a restriction enzyme site, and a stop codon (TGA). The reverse primer specified a C120S (cysteine to serine at position 120) mutation. In particular, the nucleotide sequence GCA (nucleotides 34-36) of the reverse primer shown above is replaced with one of the following nucleotide sequences: AGA, GGA, TGA, CGA, ACT, or GCT. As persons skilled in the art will understand, the nucleotide sequence GCA is a reversed complementary sequence of the codon TGC for cysteine. Similarly, the nucleotide sequences AGA, GGA, TGA, CGA, ACT, or GCT are the reversed complementary sequences of the codons for serine. Polymerase chain reaction products were digested with HindIII/XbaI and directionally subcloned into the expression vector 7rLN (Bristol-Myers Squibb Company, Princeton, N.J.). L104EA29YXC120S was prepared in an identical manner. Each construct was verified by DNA sequencing.
  • Identification and Biochemical Characterization of High Avidity Mutants [0349]
  • Twenty four amino acids were chosen for mutagenesis and the resulting ˜2300 mutant proteins assayed for CD86Ig binding by surface plasmon resonance (SPR; as described, supra). The predominant effects of mutagenesis at each site are summarized in Table II, infra. Random mutagenesis of some amino acids in the CDR-1 region (S25-R33) apparently did not alter ligand binding. Mutagenesis of E31 and R33 and residues M97-Y102 apparently resulted in reduced ligand binding. Mutagenesis of residues, S25, A29, and T30, K93, L96, Y103, L104, and G105, resulted in proteins with slow “on” and/or slow “off” rates. These results confirm previous findings that residues in the CDR-1 (S25-R33) region, and residues in or near M97-Y102 influence ligand binding (Peach et al., (1994) J. Exp. Med., 180:2049-2058). [0350]
  • Mutagenesis of sites S25, T30, K93, L96, Y103, and G105 resulted in the identification of some mutant proteins that had slower “off” rates from CD86Ig. However, in these instances, the slow “off” rate was compromised by a slow “on” rate that resulted in mutant proteins with an overall avidity for CD86Ig that was apparently similar to that seen with wild type CTLA4Ig. In addition, mutagenesis of K93 resulted in significant aggregation that may have been responsible for the kinetic changes observed. [0351]
  • Random mutagenesis of L104 followed by COS cell transfection and screening by SPR of culture media samples over immobilized CD86Ig yielded six media samples containing mutant proteins with approximately 2-fold slower “off” rates than wild type CTLA4Ig. When the corresponding cDNA of these mutants were sequenced, each was found to encode a leucine to glutamic acid mutation (L104E). Apparently, substitution of [0352] leucine 104 to aspartic acid (L104D) did not affect CD86Ig binding.
  • Mutagenesis was then repeated at each site listed in Table II, this time using L104E as the PCR template instead of wild type CTLA4Ig, as described above. SPR analysis, again using immobilized CD86Ig, identified six culture media samples from mutagenesis of [0353] alanine 29 with proteins having approximately 4-fold slower “off” rates than wild type CTLA4Ig. The two slowest were tyrosine substitutions (L104EA29Y), two were leucine (L104EA29L), one was tryptophan (L104EA29W), and one was threonine (L104EA29T). Apparently, no slow “off” rate mutants were identified when alanine 29 was randomly mutated, alone, in wild type CTLA4Ig.
  • The relative molecular mass and state of aggregation of purified L104E and L104EA29YIg was assessed by SDS-PAGE and size exclusion chromatography. L104EA29YIg (˜1 μg; lane 3) and L104EIg (˜1 μg; lane 2) apparently had the same electrophoretic mobility as CTLA4Ig (˜1 μg; lane 1) under reducing (˜50 kDa; +BME; plus 2-mercaptoethanol) and non-reducing (˜100 kDa; −βME) conditions (FIG. 25A). Size exclusion chromatography demonstrated that L104EA29YIg (FIG. 25C) apparently had the same mobility as dimeric CTLA4Ig (FIG. 25B). The major peaks represent protein dimer while the faster eluting minor peak in FIG. 25B represents higher molecular weight aggregates. Approximately 5.0% of CTLA4Ig was present as higher molecular weight aggregates but there was no evidence of aggregation of L104EA29YIg or L104EIg. Therefore, the stronger binding to CD86Ig seen with L104EIg and LI04EA29YIg could not be attributed to aggregation induced by mutagenesis. [0354]
  • Equilibrium and Kinetic Binding Analysis [0355]
  • Equilibrium and kinetic binding analysis was performed on protein A purified CTLA4Ig, L104EIg, and L104EA29YIg using surface plasmon resonance (SPR). The results are shown in Table I, infra. Observed equilibrium dissociation constants (K[0356] d; Table I) were calculated from binding curves generated over a range of concentrations (5.0-200 nM). L104EA29YIg binds more strongly to CD86Ig than does L104EIg or CTLA4Ig. The lower Kd of L104EA29YIg (3.21 nM) than L104EIg (6.06 nM) or CTLA4Ig (13.9 nM) indicates higher binding avidity of L104EA29YIg to CD86Ig. The lower Kd of L104EA29YIg (3.66 nM) than L104EIg (4.47 nM) or CTLA4Ig (6.51 nM) indicates higher binding avidity of L104EA29YIg to CD80Ig.
  • Kinetic binding analysis revealed that the comparative “on” rates for CTLA4Ig, L104EIg, and L104EA29YIg binding to CD80 were similar, as were the “on” rates for CD86Ig (Table I). However, “off” rates for these molecules were not equivalent (Table I). Compared to CTLA4Ig, L104EA29YIg had approximately 2-fold slower “off” rate from CD80Ig, and approximately 4-fold slower “off” rate from CD86Ig. L104E had “off” rates intermediate between L104EA29YIg and CTLA4Ig. Since the introduction of these mutations did not significantly affect “on” rates, the increase in avidity for CD80Ig and CD86Ig observed with L104EA29YIg was likely primarily due to a decrease in “off” rates. [0357]
  • To determine whether the increase in avidity of L104EA29YIg for CD86Ig and CD80Ig was due to the mutations affecting the way each monomer associated as a dimer, or whether there were avidity enhancing structural changes introduced into each monomer, single chain constructs of CTLA4 and L104EA29Y extracellular domains were prepared following mutagenesis of [0358] cysteine 120 to serine as described supra, and by Linsley et al., (1995) J. Biol. Chem., 270:15417-15424 (84). The purified proteins CTLA4XC120S and L104EA29YXC120S were shown to be monomeric by gel permeation chromatography (Linsley et al., (1995), supra), before their ligand binding properties were analyzed by SPR. Results showed that binding affinity of both monomeric proteins for CD86Ig was approximately 35-80-fold less than that seen for their respective dimers (Table I). This supports previously published data establishing that dimerization of CTLA4 was required for high avidity ligand binding (Greene et al., (1996) J. Biol. Chem., 271:26762-26771).
  • L104EA29YX[0359] C120S bound with approximately 2-fold higher affinity than CTLA4XC120S to both CD80Ig and CD86Ig. The increased affinity was due to approximately 3-fold slower rate of dissociation from both ligands. Therefore, stronger ligand binding by L104EA29Y was most likely due to avidity enhancing structural changes that had been introduced into each monomeric chain rather than alterations in which the molecule dimerized.
  • Location and Structural Analysis of Avidity Enhancing Mutations [0360]
  • The solution structure of the extracellular IgV-like domain of CTLA4 has recently been determined by NMR spectroscopy (Metzler et al., (1997) Nature Struct. Biol., 4:527-531). This allowed accurate location of [0361] leucine 104 and alanine 29 in the three dimensional fold (FIG. 26 left and right depictions). Leucine 104 is situated near the highly conserved MYPPPY amino acid sequence. Alanine 29 is situated near the C-terminal end of the CDR-1 (S25-R33) region, which is spatially adjacent to the MYPPPY region. While there is significant interaction between residues at the base of these two regions, there is apparently no direct interaction between L104 and A29 although they both comprise part of a contiguous hydrophobic core in the protein. The structural consequences of the two avidity enhancing mutants were assessed by modeling. The A29Y mutation can be easily accommodated in the cleft between the CDR-1 (S25-R33) region and the MYPPPY region, and may serve to stabilize the conformation of the MYPPPY region. In wild type CTLA4, L104 forms extensive hydrophobic interactions with L96 and V94 near the MYPPPY region. It is highly unlikely that the glutamic acid mutation adopts a conformation similar to that of L104 for two reasons. First, there is insufficient space to accommodate the longer glutamic acid side chain in the structure without significant perturbation to the CDR-1 (S25-R33 region). Second, the energetic costs of burying the negative charge of the glutamic acid side chain in the hydrophobic region would be large. Instead, modeling studies predict that the glutamic acid side chain flips out on to the surface where its charge can be stabilized by solvation. Such a conformational change can easily be accommodated by G105, with minimal distortion to other residues in the regions.
  • Binding of High Avidity Mutants to CHO Cells Expressing CD80 or CD86 [0362]
  • FACS analysis (FIG. 27) of CTLA4Ig and mutant molecules binding to stably transfected CD80+ and CD86+CHO cells was performed as described herein. CD80-positive and CD86-positive CHO cells were incubated with increasing concentrations of CTLA4Ig, L104EA29YIg, or L104EIg, and then washed. Bound immunoglobulin fusion protein was detected using fluorescein isothiocyanate-conjugated goat anti-human immunoglobulin. [0363]
  • As shown in FIG. 27, CD80-positive or CD86-positive CHO cells (1.5×10[0364] 5) were incubated with the indicated concentrations of CTLA4Ig (closed squares), L104EA29YIg (circles), or L104EIg (triangles) for 2 hr. at 23° C., washed, and incubated with fluorescein isothiocyanate-conjugated goat anti-human immunoglobulin antibody. Binding on a total of 5,000 viable cells was analyzed (single determination) on a FACScan, and mean fluorescence intensity (MFI) was determined from data histograms using PC-LYSYS. Data were corrected for background fluorescence measured on cells incubated with second step reagent only (MFI=7). Control L6 mAb (80 μg/ml) gave MFI<30. These results are representative of four independent experiments.
  • Binding of L104EA29YIg, L104EIg, and CTLA4Ig to human CD80-transfected CHO cells is approximately equivalent (FIG. 27A). L104EA29YIg and L104EIg bind more strongly to CHO cells stably transfected with human CD86 than does CTLA4Ig (FIG. 27B). [0365]
  • Functional Assays: [0366]
  • Human CD4-positive T cells were isolated by immunomagnetic negative selection (Linsley et al., (1992) J. Exp. Med. 176:1595-1604). Isolated CD4-positive T cells were stimulated with phorbal myristate acetate (PMA) plus CD80-positive or CD86-positive CHO cells in the presence of titrating concentrations of inhibitor. CD4-positive T cells (8-10×10[0367] 4/well) were cultured in the presence of 1 nM PMA with or without irradiated CHO cell stimulators. Proliferative responses were measured by the addition of 1 μCi/well of [3H]thymidine during the final 7 hours of a 72 hour culture. Inhibition of PMA plus CD80-positive CHO, or CD86-positive CHO, stimulated T cells by L104EA29YIg and CTLA4Ig was performed. The results are shown in FIG. 28. L104EA29YIg inhibits proliferation of CD80-positive PMA treated CHO cells more than CTLA4Ig (FIG. 28A). L104EA29YIg is also more effective than CTLA4Ig at inhibiting proliferation of CD86-positive PMA treated CHO cells (FIG. 28B). Therefore, L104EA29YIg is a more potent inhibitor of both CD80- and CD86-mediated costimulation of T cells.
  • FIG. 29 shows inhibition by L104EA29YIg and CTLA4Ig of allostimulated human T cells prepared above, and further allostimulated with a human B lymphoblastoid cell line (LCL) called PM that expressed CD80 and CD86 (T cells at 3.0×10[0368] 4/well and PM at 8.0×103/well). Primary allostimulation occurred for 6 days, then the cells were pulsed with 3H-thymidine for 7 hours, before incorporation of radiolabel was determined.
  • Secondary allostimulation was performed as follows. Seven day primary allostimulated T cells were harvested over lymphocyte separation medium (LSM) (ICN, Aurora, Ohio) and rested for 24 hours. T cells were then restimulated (secondary), in the presence of titrating amounts of CTLA4Ig or L104EA29YIg, by adding PM in the same ratio as above. Stimulation occurred for 3 days, then the cells were pulsed with radiolabel and harvested as above. The effect of L104EA29YIg on primary allostimulated T cells is shown in FIG. 29A. The effect of L104EA29YIg on secondary allostimulated T cells is shown in FIG. 29B. L104EA29YIg inhibits both primary and secondary T cell proliferative responses better than CTLA4Ig. [0369]
  • To measure cytokine production (FIG. 30), duplicate secondary allostimulation plates were set up. After 3 days, culture media was assayed using ELISA kits (Biosource, Camarillo, Calif.) using conditions recommended by the manufacturer. L104EA29YIg was found to be more potent than CTLA4Ig at blocking T cell IL-2, IL-4, and γ-IFN (gamma-IFN) cytokine production following a secondary allogeneic stimulus (FIGS. [0370] 30A-C).
  • The effects of L104EA29YIg and CTLA4Ig on monkey mixed lymphocyte response (MLR) are shown in FIG. 31. Peripheral blood mononuclear cells (PBMC'S; 3.5×10[0371] 4 cells/well from each monkey) from 2 monkeys were purified over lymphocyte separation medium (LSM) and mixed with 2 μg/ml phytohemaglutinin (PHA). The cells were stimulated 3 days then pulsed with radiolabel 16 hours before harvesting. L104EA29YIg inhibited monkey T cell proliferation better than CTLA4Ig.
    TABLE I
    Equilibrium and apparent kinetic constants are given in the following
    table (values are means ± standard deviation from three different
    experiments):
    Immobilized kon (× 105) koff (× 10−3) Kd
    Protein Analyte M−1 S−1 S−1 nM
    CD80Ig CTLA4Ig 3.44 ± 0.29 2.21 ± 0.18 6.51 ± 1.08
    CD80Ig L104EIg 3.02 ± 0.05 1.35 ± 0.08 4.47 ± 0.36
    CD80Ig L104EA29YIg 2.96 ± 0.20 1.08 ± 0.05 3.66 ± 0.41
    CD80Ig CTLA4XC120S 12.0 ± 1.0  230 ± 10  195 ± 25 
    CD80Ig L104EA29-  8.3 ± 0.26 71 ± 5  85.0 ± 2.5 
    YXC120S
    CD86Ig CTLA4Ig 5.95 ± 0.57 8.16 ± 0.52 13.9 ± 2.27
    CD86Ig L104EIg 7.03 ± 0.22 4.26 ± 0.11 6.06 ± 0.05
    CD86Ig L104EA29YIg 6.42 ± 0.40 2.06 ± 0.03 3.21 ± 0.23
    CD86Ig CTLA4XC120S 16.5 ± 0.5  840 ± 55  511 ± 17 
    CD86Ig L104EA29- 11.4 ± 1.6  300 ± 10  267 ± 29 
    YXC120S
  • [0372]
    TABLE II
    The effect on CD86Ig binding by mutagenesis of CTLA4Ig at the sites
    listed was determined by SPR, described supra. The predominant effect
    is indicated with a “+” sign.
    Effects of Mutagenesis
    No Apparent Slow “on”rate/slow Reduced ligand
    Mutagenesis Site Effect “off rate binding
    S25 +
    P26 +
    G27 +
    K28 +
    A29 +
    T30 +
    E31 +
    R33 +
    K93 +
    L96 +
    M97 +
    Y98 +
    P99 +
    P100 +
    P101 +
    Y102 +
    Y103 +
    L104 +
    G105 +
    I106 +
    G107 +
    Q111 +
    Y113 +
    I115 +
  • EXAMPLE 3
  • The following provides a description of phase II clinical studies of human patients administered soluble CTLA4 mutant molecule L104EA29YIg (also known as LEA29Y or LEA) or CTLA4Ig, to relieve at least one symptom associated with rheumatoid arthritis, including reducing: joint swelling, joint tenderness, inflammation, morning stiffness, and pain. The CTLA4Ig molecule used herein begins with methionine at position +1 (or alternatively with alanine at position −1) and ends with lysine at position +357 as shown in FIG. 24. DNA encoding an embodiment of the CTLA4Ig molecule has been deposited as ATCC 68629. The L104EA29YIg molecule used herein begins with methionine at position +1 (or alternatively with alanine at position −1) and ends with lysine at position +357 as shown in FIG. 19. DNA encoding an embodiment of the L104EA29YIg molecule has been deposited as ATCC PTA 2104. [0373]
  • Additionally, the following provides a description of human patients administered L104EA29YIg or CTLA4Ig to relieve at least one biological surrogate marker associated with rheumatoid arthritis, including reducing erythrocyte sedimentation rates, and serum levels of C-reactive protein and/or IL2 receptor. [0374]
  • Patient Cohorts [0375]
  • A total of 214 patients, including 54 males and 160 females, participated in the study (FIGS. 1A, 1B). The patients at baseline had a mean disease duration of 3.4 (±2.0) years and had failed at least one Disease Modifying Antirheumatic Drug (DMARD). Stable Nonsteroidal Anti-inflammatory Drugs (NSAIDS) or steroids (≦10 mg/day) were permitted and concomitant DMARDS were prohibited. The patients were randomized into groups of 25 to 32 patients per treatment group. Thirty-two patients received a placebo, 92 received L104EA29YIg, and 90 received CTLA4Ig. The patients who followed protocol guidelines and did not discontinue before day 57 received a total of 4 intravenous infusions, one infusion each on [0376] days 1, 15, 29, and 57. All patients were evaluated on days 1, 15, 29, 43, 57, 71, and 85. The doses administered included 0.5, 2.0, or 10.0 mg/kg of L104EA29YIg (denoted as LEA5, LEA2 and LEA10, respectively in FIGS. 1A-1E) or of CTLA4Ig (denoted as CTLA5, CTLA2 and CTLA10, respectively in FIGS. 1A-1E).
  • All subjects were monitored for peri-infusional adverse events and global safety by answering a questionnaire listing potential adverse events. The patients were questioned about potential adverse events that may have occurred within twenty-four hours post-infusion. In addition, the patients were encouraged to spontaneously report any adverse events that they experienced. The physicians routinely monitored laboratory samples from the patients for abnormalities in blood chemistry and hematology e.g. assessed the levels of inflammatory response mediators such as cytokines (TNF, IL-6), tryptase and complement. The primary endpoint was the proportion of subjects meeting the [0377] ACR 20 criteria on day 85.
  • Storage of Test Material [0378]
  • The CTLA4Ig and L104EA29YIg were supplied in single-use glass vials containing 200 mg/vial of CTLA4Ig or 100 mg/vial of L104EA29YIg, respectively. Prior to infusion, the CTLA4Ig and L104EA29YIg were diluted to a final concentration of 25 mg/ml with sterile water for injection (SWFI). [0379]
  • Administration Protocol [0380]
  • All infusions were administered intravenously over 1 hour (FIGS. 1 through 17). All subjects received at least one infusion of study medication. [0381]
  • Group 1: 32 patients, CTLA4Ig or LI04EA29YIg matching placebo. [0382]
  • Group 2: 26 patients; dosage 0.5 mg/kg of CTLA4Ig. [0383]
  • Group 3: 32 patients; dosage 2.0 mg/kg of CTLA4Ig. [0384]
  • Group 4: 32 patients; dosage 10.0 mg/kg of CTLA4Ig. [0385]
  • Group 5: 32 patients; dosage 0.5 mg/kg of L104EA29YIg. [0386]
  • Group 6: 29 patients; dosage 2.0 mg/kg of L104EA29YIg. [0387]
  • Group 7: 31 patients; dosage 10.0 mg/kg of L104EA29YIg. [0388]
  • Clinical Monitoring [0389]
  • Patients were evaluated for baseline symptoms of disease activity prior to receiving any infusions. These baseline evaluations included: joint swelling, joint tenderness, inflammation, morning stiffness, disease activity evaluated by patient and physician as well as disability evaluated by Health Questionnaire Assessment (HAQ) (reported as a physical function score in FIG. 1C), and pain (FIGS. 1A to [0390] 1D). Additionally, the baseline evaluations included erythrocyte sedimentation rates (ESR), and serum levels of C-reactive protein (CRP) and soluble IL-2 receptor (IL-2r) (FIGS. 1C and 1D).
  • The clinical response studies were based on the criteria established by the American College of Rheumatology (ACR). A subject satisfied the ACR20 criterion if there was a 20 percent improvement in tender and swollen joint counts and 20 percent improvement in three of the five remaining symptoms measured, such as patient and physician global disease changes, pain, disability, and an acute phase reactant (Felson, D. T., et al., 1993 [0391] Arthritis and Rheumatism 36:729-740; Felson, D. T., et al., 1995 Arthritis and Rheumatism 38:1-9). Similarly, a subject satisfied the ACR50 or ACR70 criterion if there was a 50 or 70 percent improvement, respectively, in tender and swollen joint counts and 50 or 70 percent improvement, respectively, in three of the five remaining symptoms measured, such as patient and physician global disease changes, pain, physical disability, and an acute phase reactant such as CRP or ESR.
  • Biomarkers [0392]
  • Potential biomarkers of disease activity (rheumatoid factor, CRP, ESR, soluble IL-2R, soluble ICAM-1, soluble E-selectin, and MMP-3) were also assessed. Validated enzyme immunoassay (EIA) methods were used to determine the serum concentration of IL-2sRα, sICAM-1, sE-selectin and MMP-3. TNFα and IL-6 were assessed at infusion pre and 2 hours post, if necessary. [0393]
  • IL-2sRα, sICAM-1, and sE-selectin were measured using commercially available calorimetric EIA kits from R&D Systems, Inc. (Minneapolis, Minn.). The lower and upper limits of quantitation were 312-20,000 pg/mL, 40-907 ng/mL and 10-206 ng/mL, respectively. The inter-assay coefficient of variation ranged from 4.48-8.4%, 3.8-5.0% and 5.5-9.0% respectively. According to the kit manufacturer, normal serum values range from 676-2,132 pg/mL, respectively. [0394]
  • MMP-3 was measured using a commercially available calorimetric EIA kit from Amersham Pharmacia Biotech (Piscataway, N.J.). The lower and upper limits of quantitation were 30-7,680 ng/mL. The inter-assay coefficient of variation ranged from 6.3-10.6%. According to the kit manufacturer, normal serum values range from 28-99 ng/mL. [0395]
  • IL-6 and TNFα were measured using commercially available chemiluminescent EIA kits from R&D Systems, Inc. (Minneapolis, Minn.). The lower and upper limits of quantitation were 0.3-3,000 pg/mL and 0.7-7,000 pg/mL, respectively. The inter-assay coefficient of variation ranged from 3.1-5.7% and 6.4-20.7%, respectively. According to the kit manufacturer, normal serum values range from <0.3-12 pg/mL and <0.7-7.5 pg/mL. [0396]
  • Antibody Testing [0397]
  • Serum samples were obtained for assessment of drug-specific antibodies prior to dosing on [0398] day 1, and approximately on days 15, 29, 57, 85 and 169. Due to high, preexisting titers directed to the immunoglobulin (Ig) portion of the molecule, specific antibody formation against CTLA4Ig and LEA29Y without Ig constant regions was also assessed.
  • Ninety-six well Immulon II ELISA plates (Dynex, Chantilly, Va.) were coated with CTLA4Ig, CTLA4Ig without the Ig constant regions, LEA29Y, or LEA29Y without the Ig constant regions at 2, 4, 2, or 1 μg/ml in phosphate buffered saline (PBS), respectively, and incubated overnight at 2-8° C. The plates were washed with PBS containing 0.05[0399] % Tween 20 and blocked for 1 hour at 37° C. with PBS containing 1% bovine serum albumin (BSA). The plates were then washed and serial dilutions of the test sera or quality control (QC) sera were added to the appropriate wells and incubated for 2 hours at 37° C. Sera was diluted threefold in PBS with 0.25% BSA and 0.05% Tween 20 starting at a 1:10 dilution. Plates were washed and an alkaline-phosphatase-conjugated goat anti-human kappa and lambda (Southern Biotechnology Associates, Inc., Birmingham, Ala.) antibody cocktail was added. Following a 1-hour incubation at 37° C., the plates were washed and 1 mg/ml para-nitrophenyl phosphate in diethanolamine buffer was added to each well. After 30 minutes at 25° C., the reactions were stopped with 3N NaOH and the absorbance (dual wavelength: 405 nm and 550 nm) was recorded. Results were expressed as endpoint titer (EPT), defined as the reciprocal of the highest dilution that resulted in an absorbance reading fivefold greater than or equal to the mean plate-background absorbance. Plate background was determined as the absorbance measurement recorded in the absence of serum. Values were considered positive for seroconversion if they were at least two serial dilutions (ninefold) or greater relative to predose EPT values. Serum QC samples positive for either CTLA4Ig- or LEA29Y-specific antibodies were generated from immunized monkeys. An aliquot of the appropriate QC sample was assayed during each analytical run. Analytical runs were accepted only when the QC samples were within the assay acceptance criteria.
  • Results [0400]
  • CTLA4Ig and L104EA29YIg were generally well-tolerated at all dose-levels. Peri-infusional adverse events were similar across all dose groups, with the exception of headaches. Headache response of patients on [0401] day 85 increased dose-dependently 23%, 44%, and 53% in CTLA4Ig-treated patients, and 34%, 45%, and 61% in L104EA29YIg-treated patients, at 0.5, 2.0, and 10.0 mg/kg respectively. In contrast, 31% of the patients administered placebos experienced headaches.
  • The percent of patients that discontinued from the clinical study due to arthritis flares and other adverse events is summarized in FIG. 2. A much higher percentage of patients on placebo discontinued treatment due to arthritis flare. The CTLA4Ig treated patients discontinued treatment less with increasing doses. Very few patients treated with L104EA29YIg discontinued treatment. These results indicate a good inverse dose-dependent response for CTLA4Ig, and a stronger therapeutic response with L104EA29YIg therapy. [0402]
  • The ACR-20, −50, and −70 responses of patients treated with CTLA4Ig, L104EA29YIg, or placebo at [0403] day 85 are summarized in FIG. 3A. Similarly, FIGS. 3B and C describe the ACR-20 responses with 95% confidence limits. The responses appear to be dose-dependent with a clear significant response at 10 mg/kg per body weight of the patient.
  • The percent of patients having reduced swollen and tender joint counts compared to the patients having no response to treatment with CTLA4Ig, L104EA29YIg, or placebo, is shown in FIGS. 4A and B. The therapeutic responses appear to be dose-dependent. A larger percentage of patients show improvement of 20, 50, 70, and even 100% in the 2 and 10 mg/kg groups for both products. [0404]
  • The percent of patients having reduced pain, disease activity evaluated by patient and physician mean score units with CTLA4Ig, L104EA29YIg, or placebo, is shown in FIGS. 5A, B, C, and D. The therapeutic responses, as monitored by the Likert scale, appear to be dose-dependent in favor of the active treatment groups as compared to placebo on [0405] day 85. The Likert scale is a validated verbal rating scale using adjectives to rank the symptoms (The American College of Rheumatology Preliminary Core Set of Disease Activity Measures for Rheumatoid Arthritis Clinical Trials: Arthritis and Rheumatism, June 1993, 36(6):729-740).
  • The patient and physician assessments of disease activity change from the baseline by at least 2 units, resulting from treatment with CTLA4Ig, L104EA29YIg, or placebo, are shown in FIGS. 6A and B. The responses appear to be dose-dependent with more marked improvement for the higher doses of active drugs. [0406]
  • The percent reduction in C-reactive protein (CRP) levels in patients treated with CTLA4Ig, L104EA29YIg, or placebo, is shown in FIGS. 7A and B. The responses appear to be dose-dependent with a clear decrease for the 2 and 10 mg/kg active treatment groups. In addition, FIG. 7B showed that the difference is quite significant compared to placebo with 95% confidence intervals. FIG. 7C shows the changes in serum level changes from baseline at [0407] day 85.
  • The amount of serum soluble IL-2 receptor in patients treated with CTLA4Ig, L104EA29YIg, or placebo, is shown in FIG. 8. The reduction in soluble IL-2 receptor levels appears to be dose-dependent. [0408]
  • The amount of serum soluble ICAM-1 and soluble E-selectin in patients treated with CTLA4Ig, L104EA29YIg, or placebo, is shown in FIG. 33. The reduction in soluble ICAM-1 and soluble E-selectin levels appears to be dose-dependent. [0409]
  • The median and mean tender joint counts in patients treated with CTLA4Ig or placebo over time are shown in FIGS. 9A and B. The change from baseline (e.g., reduction in tender joints) appears to be more important in the 2 and 10 mg/kg treated groups, than in the placebo or 0.5 mg/kg groups. [0410]
  • The median and mean swollen joint counts in patients treated with CTLA4Ig or placebo over time are shown in FIGS. 10A and B. The change from baseline (e.g., reduction in swollen joints) appears to be more important in the 2 and 10 mg/kg treated groups than placebo or 0.5 mg/kg groups. [0411]
  • The mean pain assessment scores over time in patients treated with CTLA4Ig or placebo are shown in FIG. 11. The change from baseline (e.g., reduction in pain) appears to be more important in the 2 and 10 mg/kg treated groups than placebo or 0.5 mg/kg groups. [0412]
  • The mean disease activity assessment scores assessed by patient or physician in patients treated with CTLA4Ig or placebo over time are shown in FIGS. 12A and B. The change from baseline (e.g., reduction in disease activity) appears to be more important in the 2 and 10 mg/kg treated groups than placebo or 0.5 mg/kg groups. [0413]
  • The median and mean tender joint counts in patients treated with L104EA29YIg (denoted as LEA in the figures) or placebo over time are shown in FIGS. 13A and B. The change from baseline (e.g., reduction in tender joints) appears to be dose-dependent. [0414]
  • The median and mean swollen joint counts in patients treated with L104EA29YIg (denoted as LEA in the figures) or placebo over time are shown in FIGS. 14A and B. The change from baseline (e.g., reduction in swollen joints) appears to be more important in the 2 and 10 mg/kg treated groups than placebo or 0.5 mg/kg groups. [0415]
  • The mean pain assessment scores in patients treated with L104EA29YIg (denoted as LEA in the figures) or placebo over time are shown in FIG. 15. The change from baseline (e.g., reduction in pain) appears to be dose-dependent. [0416]
  • The mean disease activity assessment scores evaluated by patient or physician in patients treated with L104EA29YIg (denoted as LEA in the figures) or placebo over time are shown in FIGS. 16A and B. The change from baseline (e.g., reduction in disease activity) appears to be dose-dependent. [0417]
  • The percent improvement of physical disability assessed by HAQ at [0418] day 85 for patients treated with CTLA4Ig, L104EA29YIg, or placebo are shown in FIG. 17 (Health Assessment Questionnaire (HAQ); Fries, J. F., et al., 1982 J. of Rheumatology 9:789-793). There is a clear dose dependent improvement with this parameter.
  • The changes from baseline for soluble IL-2r and C-reactive protein levels were dose-dependent in both treatment groups. After treatment, soluble IL-2r levels were −2%, −10%, and −22% for CTLA4Ig and −4%, −18%, and −32% for L104EA29YIg at 0.5, 2.0, and 10.0 mg/kg respectively, compared to +3% for the placebo. C-reactive protein levels were +12%, −15%, and −32% for CTLA4Ig and +47%, −33%, and −47% for L104EA29YIg at 0.5, 2.0, and 10.0 mg/kg respectively, compared to +20% for the placebo (FIG. 7A). [0419]
  • No clinically remarkable findings with respect to routine hematology testing, chemistry laboratory testing with the exception of slight suppressions in IgA and IgG levels at the higher doses of both drugs, physical findings, or vital signs assessments were observed. Notably, neither medication induced drug-specific antibodies. [0420]
  • EXAMPLE 4
  • The following examples describe phase II clinical studies of human patients that will be administered L104EA29YIg, to reduce or prevent structural damage, including bone or joint erosion using validated radiographic scales. This improvement in reducing or preventing structural damage is parallel to the clinical improvement measured by the clinical parameters. [0421]
  • The status of the bone structure is monitored in some of the human patients prior to treatment with CTLA4Ig or L104EA29YIg. These patients are administered from 0.5 to 20 mg/kg of CTLA4Ig or L104EA29YIg chronically every two to twelve weeks (alone or in combination with other agents) to maintain their therapeutic improvement over time. Radiographs of patients' hands and feet are taken at predefined intervals: 6 months, and then yearly, as recommended by the FDA guidelines. These patients are monitored in long-term extension after 6 and 12 months to determine if treatment with CTLA4Ig or L104EA29YIg reduces the progression of bone deterioration, and then yearly. The patients are monitored by radiographic methods, including X-ray and/or magnetic resonance imaging (MRI), according to standard practice in the art (Larsen, A. K. and M. Eek 1977 [0422] Acta. Radiol. Diag. 18:481-491; Sharp, J. T., et al., 1985 Arthritis and Rheumatism 28:1326-1335). The results of the radiographic data are evaluated for prevention of structural damage, including slowing the progression of bone erosion and cartilage damage, with joint space narrowing and/or prevention of new erosions.
  • EXAMPLE 5
  • A Study to Evaluate the Safety and Clinical Efficacy of Two Different Doses of CTLA4Ig Administered Intravenously to Subjects with Active Rheumatoid Arthritis [0423]
  • While Receiving Methotrexate Rheumatoid Arthritis (RA) treatment is rapidly changing with an increased willingness to use more aggressive therapies to achieve larger increases in efficacy and higher success rates. The ultimate goal is to improve the subject condition in a more intensive way, by raising the rate of major and complete clinical response, to treatment and maintaining this benefit with acceptable safety. [0424]
  • Methotrexate remains the cornerstone of the RA treatment. It was the first agent that demonstrated early onset of action, superior efficacy and tolerability compared to the classical DMARDs (e.g. gold, hydroxychloroquine, sulfasalazine) used to treat RA. Clinical benefit may be seen as early as 3 weeks after initiating treatment, and the maximal improvement is generally achieved by 6 months. However, methotrexate has a number of limitations. For example, despite its increased tolerability, the window between efficacy and liver toxicity is quite narrow. Subjects treated with methotrexate require careful monitoring and unacceptable toxicity is often the reason for discontinuation of treatment. [0425]
  • Methotrexate also does not appear to efficiently control disease progression or joint deterioration. For some subjects, practitioners feel compelled to add a second DMARD with the hope of increasing efficacy despite the risk of increased toxicity. Alternatively, co-treatment with methotrexate and a costimulator blocker (e.g. CD80 and CD86 blockers such as CTLA4Ig) that target the auto-immune mechanism that lies upstream of the cytokine inflammatory cascade, may also increase efficacy. [0426]
  • As noted in Example 3, above, significant clinical responses and reductions in surrogate markers of disease activity were observed for CTLA4Ig at doses of 2 and 10 mg/kg with a good tolerability profile. It has also been confirmed that the composition CTLA4Ig, used in Example 3 above, did not induce any side effects. As a result, it was decided to continue the clinical development of CTLA4Ig for rheumatoid arthritis in Phase IIB. [0427]
  • The following provides a description of a Phase IIB clinical study of human patients administered soluble CTLA4 molecule with methotrexate, and the results of the study after six months. [0428]
  • This example describes a twelve month study in which primary efficacy was assessed after all subjects completed six months of treatment or discontinue therapy. Efficacy, safety, and disease progression were also assessed throughout the duration of the study. [0429]
  • The study utilized a randomized, double blind, placebo controlled, parallel dosing design. The study was designed to evaluate the safety, clinical activity, immunogenicity and pharmacokinetics of two doses of CTLA4Ig: 2 or 10 mg/kg. A total of approximately 330 subjects with active RA and receiving methotrexate were randomized to 1 of 3 dosing arms: CTLA4Ig at 2 mg/kg (N=110), 10 mg/kg (N=110) and placebo control group (N=110) given monthly infusions for 12 months. All groups continued on weekly methotrexate treatment (10-30 mg weekly) (FIGS. [0430] 57-62).
  • CTLA4Ig or a placebo were also administered on [0431] Day 15. Each dose of study medication was infused intravenously over approximately 30 minutes. The primary efficacy endpoint was the ACR 20 response rate after 6 months.
  • For the first 6 months, subjects were not allowed to alter their doses of corticosteroids, glucocorticoids or NSAIDs. Increases in methotrexate were also not permitted during the first six months. Decreases in methotrexate were permitted only if it was felt to be causing toxicity. Subjects were treated with methotrexate for at least 6 months, and at a stable dose for 28 days prior to first treatment of CTLA4Ig or placebo. DMARDs other than methotrexate were not permitted. Low-dose stable corticosteroids use (at 10 mg daily or less) and/or use of stable non-steroidal anti-inflammatory drugs (NSAIDs), including acetyl salicylic acid (ASA), was allowed. Analgesics that did not contain ASA or NSAIDs were permitted in subjects experiencing pain not adequately controlled by the baseline and study medications, except for 12 hours before a joint evaluation. Decreases in NSAIDs were permitted but only if due to adverse events such as gastrointestinal toxicity. [0432]
  • Test Product, Dose and Mode of Administration, Duration of Treatment [0433]
  • CTLA4Ig at 2 mg/kg or 10 mg/kg was infused every two weeks for the first month, and monthly thereafter for 12 months. [0434]
  • All subjects received weekly doses of methotrexate (10-30 mg) for at least six months prior to randomization and maintained at the entry dose for the first 6 months of the trial. Doses could only be decreased for toxicity during the first six months. [0435]
  • Criteria for Evaluation [0436]
  • The primary endpoint of the first stage of the study was the proportion of subjects meeting the American College of Rheumatology criteria for 20% improvement (ACR 20) on Day 180 (month six). The [0437] ACR 20 definition of improvement is a 20% improvement from baseline in the number of tender and swollen joint counts, and a 20% improvement from baseline in 3 of the following 5 core set measurements: subject global assessment of pain, subject global assessment of disease activity, physician global assessment of disease activity, subject assessment of physical function and acute phase reactant value (C-reactive protein (CRP)). The evaluation for 50% improvement (ACR 50) and 70% improvement (ACR 70) follow similarly. Subjects who discontinued the study due to lack of efficacy (i.e. worsening RA) were considered as ACR non-responders from that time on. For all subjects who dropped out for other reasons, their ACR response at the time of discontinuation was carried forward.
  • Statistical Methods [0438]
  • Two doses of CTLA4Ig (2 mg/kg and 10 mg/kg) were compared with the placebo control group. All subjects were maintained at the same stable entry doses of methotrexate. The primary analysis was the comparison of [0439] CTLA4Ig 10 mg/kg with placebo. Sample sizes were based on a 5% level (2-tailed) of significance. Based on published studies, the placebo plus methotrexate control ACR 20 response rate at 6 months is about 25%. A sample of 107 subjects (adjusted for a possible 15% dropout) per treatment arm was determined to yield a 94% power to detect a difference of 25% at the 5% level (two-tailed). Similarly, the sample was determined to yield a power of 95% and 90% to detect differences of 20% and 14% in ACR 50 and ACR 70, respectively. If the comparison between CTLA4Ig 10 mg/kg and placebo was significant with regards to ACR 20, then the comparison between CTLA4Ig 2 mg/kg and placebo was carried out. This second testing should have a power of 88%. This sequentially rejective procedure based on Chi-square tests was also used to test for differences in ACR 50 and ACR 70 responses.
  • All efficacy analyses were based on a data set containing all available assessments from all subjects who received at least one dose of study medication. [0440]
  • Percent changes from baseline were also reported for the individual components of the ACR. For subjects who discontinued, their last observation was carried forward. [0441]
  • Results [0442]
  • Demography and Baseline Characteristics: [0443]
    TABLE III
    Subject Disposition and Demographics
    Methotrexate + Methotrexate +
    CTLA4Ig CTLA4Ig Methotrexate +
    10 mg/kg 2 mg/kg Placebo
    Enrolled/  115  105  119
    Randomized
    Completed   99 (86.1%)   82 (78.1%)   78 (65.5%)
    Discontinued   16 (13.9%)   23 (21.9%)   41 (34.5%)
    Adverse Events   2 (1.7%)   7 (6.7%)   7 (5.9%)
    Lack of Efficacy   12 (10.4%)   13 (12.4%)   29 (24.4%)
    Other   2 (1.7%)   3 (2.9%)   5 (4.2%)
    Age (yrs)-Mean 55.8 (17-83) 54.4 (23-80) 54.7 (23-80)
    (Range)
    Weight (kg)-Mean 77.8 (40.1- 78.7 (48.4- 79.9 (44-140)
    (Range) 144) 186.8)
    Sex 75% females 63% females 66% females
    Race 87% white 87% white 87% white
    Duration of Disease
    (yrs)
    Mean ± SD 9.7 ± 9.8 9.7 ± 8.1 8.9 ± 8.3
  • Demographic and baseline clinical characteristics were similar among the treatment groups. Sixty three to 75 percent of subjects were female, 87% were Caucasian. The mean duration of the disease at entry was 9.7±9.8, 9.7±8.1, and 8.9±8.3 years respectively in the 10, 2 mg/kg and the control group. The mean weight in kg was very similar between 77.8 and 79.9 kg with a range of 40.1 to 186.8 kg (Table III). [0444]
  • After 6 months, more subjects had discontinued from the control group (35.5%) than from the active treatment groups; 13.9% and 21.9% for the 10 and 2 mg/kg treated groups, respectively. The main reason was lack of efficacy: with 24.3% discontinuing in the control group, as opposed to 12.4% and 10.4% discontinuing in the 2 and 10 mg/kg groups, respectively. The discontinuation rate due to adverse events was lower in the 10 mg/kg group with 1.7%, while it was 6.7% and 5.9% in the 2 mg/kg and the control groups, respectively. [0445]
  • During the first 3-4 months, the discontinuations appeared at a faster rate in the control group compared to the active-treatment groups. After [0446] Day 120, the discontinuations for all treatment groups stabilized for the duration of the primary treatment period (six months).
    TABLE IV
    Baseline Clinical Characteristics
    Methotrexate + Methotrexate +
    CTLA4Ig CTLA4Ig Methotrexate +
    10 mg/kg 2 mg/kg Placebo
    (n = 115) (n = 105) (n = 119)
    Tender Joints 30.8 ± 12.2 28.2 ± 12.0 29.2 ± 13.0
    (mean ± SD)
    Swollen Joints 21.3 ± 8.4  20.2 ± 8.9  21.8 ± 8.8 
    (mean ± SD)
    Pain (VAS 100 mm) 62.1 ± 21.4 64.5 ± 22.3 65.2 ± 22.1
    (mean ± SD)
    Physical Function 1.0 ± 0.5 1.0 ± 0.5 1.0 ± 0.6
    (MHAQ score of 0 to
    3)(mean ± SD)
    Subject global 60.1 ± 20.7 59.4 ± 23.7 62.8 ± 21.6
    assessment
    (VAS 100 mm)
    (mean ± SD)
    Physician global 62.1 ± 14.8 61.0 ± 16.7 63.3 ± 15.5
    assessment
    (VAS 100 mm)
    (mean ± SD)
    CRP (mg/dL) 2.9 ± 2.8 3.2 ± 2.6 3.2 ± 3.2
    Morning Stiffness 97.9 ± 63.1 104.1 ± 63.9  106.0 ± 64.2 
    (in min.)
  • The mean number of tender and swollen joints at baseline was comparable among the three treatment groups. The mean number of tender joints and swollen joints in the 10 mg group was 30.8±12.2 and 21.3±8.4, respectively. The mean number of tender joints and swollen joints in the 2 mg group was 28.2±12.0, and 20.2±8.9, respectively. The mean number of tender joints and swollen joints in the control group was 29.2±13.0, and 21.8±8.8, respectively. These assessments and all other clinical assessments were similar among all treatment groups (Table IV). [0447]
  • ACR Responses and Core Components: [0448]
    TABLE V
    ACR Response at 6 months
    Methotrexate + Methotrexate +
    CTLA4Ig CTLA4Ig Methotrexate +
    10 mg/kg 2 mg/kg Placebo
    (n = 115) (n = 105) (n = 119)
    ACR 20 60.0% 41.9% 35.3%
    Difference from 24.7  6.6
    control group
    95% CI 11.9, 37.5 −6.2, 19.4
    p-value <0.001  0.31
    ACR 50 36.5% 22.9% 11.8%
    Difference from 24.8 11.1
    control group
    95% CI 13.8, 35.7  1.2, 20.9
    p-value <0.001  0.027
    ACR 70 16.5% 10.5% 1.7%
    Difference from 14.8  8.8
    control group
    95% CI  7.5, 22.2  2.7, 14.9
    p-value <0.001  0.005
  • The improvements in [0449] ACR 20, 50, and 70 response rates in the 10 mg/kg treatment group, at six months relative to the methotrexate control group, were statistically significant (FIGS. 34-38, 40). The improvements in ACR 50, and ACR 70 for the 2 mg/kg group were also statistically significant. The difference in ACR 20 response between the 2 mg/kg group and the control group was 6.6%. This difference was not statistically significant, p=0.31 (Table V, FIG. 49).
  • FIGS. [0450] 34-37 presents the ACR response rates from Day 1 to Day 180. FIGS. 38 and 40 presents the ACR20, −50 and −70 response rates on day 180 for the various treatment groups. The ACR 50 and ACR 70 response rates suggest the possibility that maximal efficacy may not have been achieved at 10 mg/kg.
  • FIG. 39 shows the proportion of new tender and swollen joints at [0451] day 180 of the study after therapy with methotrexate alone or in combination with CTLA4Ig (administered at 2 or 10 mg/kg body weight of subject).
  • FIG. 46 shows the mean percent improvement in physical function from baseline as measured by HAQ. [0452]
    TABLE VI
    Individual ACR Components at Day 180
    (Mean Percent Improvement)
    Methotrexate + Methotrexate +
    CTLA4Ig CTLA4Ig Methotrexate +
    10 mg/kg 2 mg/kg Placebo
    Core Components (n = 115) (n = 105) (n = 119)
    Tender Joints 59.9% 43.3% 32.1%
    Swollen Joints 54.9% 45.1% 33.4%
    Pain 46.4% 22.7%  8.4%
    Physical Function 41.5% 17.3% 14.1%
    (mHAQ)
    Subject global 40.8%  9.6% 17.6%
    assessment
    Physician global 52.0% 38.6% 25.6%
    assessment
    CRP 31.5% 16.2% −23.6% 
  • The 2 and 10 mg/kg dose groups demonstrated some degree of efficacy among all clinical components of the ACR response criteria (Table VI; FIGS. [0453] 41-45, 47-48); the subject's global assessment in the 2 mg/kg dose group being the only exception. The reduction of tender and swollen joints appears dose-dependent. The number of tender joints was decreased by 59.9%, 43.3% and 32.1% in the 10 mg/kg, 2 mg/kg and control groups, respectively. A similar pattern was observed for the swollen joint counts with a decrease of 54.9%, 45.1% and 33.4% in the 10 mg/kg, 2 mg/kg and control groups, respectively. The greatest differences relative to the control group were observed with the pain assessment which decreased 46.4% and 22.7% relative to baseline for 10 mg and 2 mg/kg CTLA4Ig, respectively, compared to 8.4% in the control group. The mean CRP decreased 31.5% and 16.2% relative to baseline in the 10 and 2 mg/kg groups compared to an increase of 23.6% in the control group.
  • Health-Related Quality of Life [0454]
  • The impact of CTLA4Ig on health-related quality of life (HRQOL) was measured by the Medical Outcomes Study Short Form-36 (SF-36). The SF-36 was administered to all subjects at baseline, 90 and 180 days. The SF-36 consists of 36 items which covers eight domains (physical function, role-physical, bodily pain, general health, vitality, social function, role emotional, and mental health). These individual domains are used to derive the physical and mental component summary scores which range from 0 to 100, with higher scores indicating better quality of life. Absolute differences of 5 or more in the SF-36 scores were considered clinically meaningful. [0455]
  • Compared to subjects treated with placebo, subjects in the [0456] CTLA4Ig 10 mg/kg group also experienced statistically significantly greater improvement in all 8 domains of the SF-36 (FIGS. 50-51). For subjects treated with CTLA4Ig 2 mg/kg, the improvements were also greater than those treated with placebo, but the differences were not statistically significant (FIGS. 50-51).
  • Baseline SF-36 scores were comparable between the three treatment groups. Improvements in quality of life show a clear dose-response trend after 6 months of treatment. Subjects in the [0457] CTLA4Ig 10 mg/kg treatment group demonstrated clinically and statistically significant improvements from baseline in all 8 domains of the SF-36. The greatest effects were shown in the role-physical, bodily pain, and role-emotional domains. This positive finding was consistent with the efficacy results. For subjects treated with CTLA4Ig 2 mg/kg, improvements from baseline were also statistically significant for all domains except mental health.
  • Pharmacokinetics: [0458]
    PHARMACOKINETIC PARAMETER VALUES
    CMAX TMAX AUC(TAU) T-HALF CLT VSS
    (μG/ML) (H) (μG · H/ML) (Days) (ML/H/KG) (L/KG)
     2 mg/kg
    MEAN 57.96 0.50* 10176.14 13.50 0.23 0.07
    SD 16.93 (0.00, 4.00) 3069.84 5.91 0.13 0.04
    N 15 15 15 15 15 15
    10 mg/kg
    MEAN 292.09 0.50* 50102.56 13.11 0.22 0.07
    SD 67.78 (0.00, 4.00) 15345.95 5.32 0.09 0.03
    N 14 14 14 14 14 14
  • The pharmacokinetics of CTLA4Ig were derived from serum concentration versus time data between [0459] dosing days 60 and 90. Samples were collected prior to dosing on day 60, at 0.5, and 4 h after dosing, on days 67, 74, 81, and prior to dosing on Day 90. The preliminary data indicate that both Cmax and AUC values increase in a proportion comparable to the dose increment. For nominal doses increasing in a 1:5 proportion, both the Cmax and AUC values increased in the proportion of 1:5.04 and 1:4.92, respectively. T-HALF, CLT, and Vss values appeared to be comparable and dose independent.
  • Mean Vss values were 0.07 L/kg for both dose levels, which was approximately 1.6-fold the plasma volume. [0460]
  • Pharmacodynamics: [0461]
    TABLE VII
    Mean Baseline Values for Pharmacodynamic Biomarkers
    Methotrexate + Methotrexate +
    CTLA4Ig CTLA4Ig Methotrexate +
    10 mg/kg 2 mg/kg Placebo
    Biomarker (n = 115) (n = 105) (n = 119)
    CRP (mg/dL) 2.9 3.2 3.2
    RF (IU/L) 207 274 179
    IL-2r (pg/ml) 1388 1407 1398
    IL-6 (pg/ml) 26.7 31.7 21.4
    TNFα (pg/ml) 11.8 6.0 11.9
  • Serum levels of pharmacodynamic biomarkers were analyzed at various times during the study. Baseline values are shown in Table VII. The values on [0462] Day 180 relative to baseline are shown in the FIGS. 52-56.
  • CRP levels decreased from baseline in both CTLA4Ig-treated groups more than in the control group, with greater reduction observed in the 10 mg/kg dosing group (see FIGS. 47, 48 and [0463] 52).
  • Rheumatoid factor levels decreased from baseline in both CTLA4Ig-treated groups more than in the control group, with greater reduction observed in the 10 mg/kg dosing group (see FIG. 53). [0464]
  • Soluble IL-2r levels decreased from baseline in both CTLA4Ig-treated groups more than in the control group, with greater reduction observed in the 10 mg/kg dosing group (see FIG. 54). [0465]
  • Serum IL-6 levels decreased from in both CTLA4Ig-treated groups more than in the control group (see FIG. 55). [0466]
  • The effects of CTLA4Ig on serum TNFα levels were inconclusive. The 2 mg/kg group increased and the 10 mg/kg group decreased relative to the control group (see FIG. 56). [0467]
  • Safety: [0468]
  • CTLA4Ig was well tolerated at all doses. There were no deaths, malignancies or opportunistic infections in any subjects receiving CTLA4Ig. Serious adverse events (SAEs) and non-serious adverse events (NSAEs) were similar or less frequent in the active-treatment groups compared to the control group. [0469]
  • Fewer subjects in the 10 mg/kg group discontinued due to adverse events relative to the control group (1.7% vs 5.9%, respectively). The discontinuations due to adverse events in the 2 mg/kg were similar to the control group (6.7% vs 5.9%, respectively). The SAEs followed a pattern similar to the discontinuations due to adverse events. [0470]
  • No serious adverse events in the 10 mg/kg dose group were considered related to the study drug. [0471]
  • Immunogenicity: [0472]
  • No anti-drug antibody responses were detected through [0473] Day 180 at both dose levels of CTLA4Ig.
  • CTLA4Ig significantly reduced the signs and symptoms of rheumatoid arthritis in subjects receiving methotrexate as assessed by ACR response criteria. The effects of CTLA4Ig appear to increase in proportion to dose level. The improvement from baseline in all ACR core components is higher in the 10 mg/kg group than the 2 mg/kg group. CTLA4Ig at 10 mg/kg doses demonstrated clinically and statistically significant improvements in all 8 domains of the SF-36. All pharmacodynamic biomarkers assayed appeared to decrease in proportion to CTLA4Ig dose level except for TNFα. CTLA4Ig was safe and well tolerated in subjects with rheumatoid arthritis receiving methotrexate. The adverse event profile for both CTLA4Ig doses was similar to the control group. [0474]
  • EXAMPLE 6
  • A Study of a Co-Stimulation Blocker, CTLA4Ig, Given Monthly in Combination with Etanercept to Patients with Active Rheumatoid Arthritis. [0475]
  • The following example provides a description of the administration of CTLA4Ig, in combination with etanercept, to treat patients with active Rheumatoid Arthritis. [0476]
  • Etanercept, along with infliximab, comprises a new generation of Rheumatoid Arthritis drugs which targets Tumor Necrosis Factor (TNF. Etanercept is a dimeric fusion protein having an extracellular portion of the TNF receptor linked to the Fc portion of human immunoglobulin (IgG1). This fusion protein binds to TNF, blocks its interactions with cell surface TNF receptors and render TNF molecules biologically inactive. [0477]
  • This example describes a twelve month study in which efficacy was assessed after all subjects completed six months of treatment or discontinued therapy. Efficacy, safety and disease progression were also assessed throughout the duration of the study. [0478]
  • The study utilized a randomized, double-blind, placebo controlled, parallel dosing design. A total of approximately 141 subjects with active RA and receiving etanercept (25 mg twice weekly) were randomized to 1 of 2 dosing groups: 1) a group receiving CTLA4Ig at 2 mg/kg (n=94) plus etanercept or 2) a placebo group receiving etanercept only (n=47). [0479]
  • Test Product, Dose and Mode of Administration, Duration of Treatment [0480]
  • All subjects received etanercept (25 mg twice weekly) for at least 3 months prior to treatment. [0481]
  • Infusions of CTLA4Ig were given on [0482] Days 1, 15, 30, and monthly thereafter, for 6 months (primary treatment phase). Each dose of study medication was infused intravenously for approximately 30 minutes.
  • The primary treatment phase of the study took place during the first six months of treatment. During this period, subjects were required to remain on stable doses of etanercept (25 mg twice weekly). DMARDs other than etanercept were not permitted. Low-dose stable corticosteroid (at 10 mg daily or less) and/or stable non-steroidal anti-inflammatory drug (NSAID), including acetyl salicylic acid (ASA), use was allowed. Analgesics (that do not contain ASA or NSAIDs) were permitted in subjects experiencing pain that was not adequately controlled by the baseline and study medications, except for 12 hours before a joint evaluation. [0483]
  • Criteria for Evaluation [0484]
  • The primary endpoint of this study was to collect data regarding the proportion of subjects meeting modified American College of Rheumatology (ACR) criteria for 20% improvement (ACR 20) after six months. The modified [0485] ACR 20 criteria were used to accommodate the low CRP levels in this study's subject population. The modified ACR criteria were defined as 1) a greater than 20% improvement in tender and swollen joint count and 2) a greater than 20% improvement in 2 of the remaining 4 core data set measures (global pain, physician, subject, functional assessment). CRP, which is normally a part of the standard ACR core data sets, was not included in the modified ACR criteria due to the low levels of CRP in subjects using TNF blockers, such as etanercept. The standard ACR criteria, and two alternative criteria (SF-36 Physical Health and SF-36 Mental Health) were also evaluated as secondary endpoints.
  • Statistical Methods [0486]
  • Treatment of a group of patients with [0487] CTLA4Ig 2 mg/kg in combination with etanercept was compared with a control group treated with placebo plus etanercept. Based on previous studies with etanercept in similar patient populations, it was assumed that the modified ACR 20 response rate (modified criteria for evaluation) at 6 months would be 35% in the control group. This is the rate of response expected among subjects who did not respond adequately to etanercept therapy. Using a 2:1 randomization, a sample of 141 (adjusted for a possible 10% dropout) subjects (47 control/94 CTLA4Ig) yields a 90% power to detect a difference of 30% at the 5% level of significance (2-tailed, based on a chi-square test with no adjustment for continuity correction).
  • Similarly, the sample was determined to yield a power of 91% and 83% to detect differences of 30 and 25% in [0488] ACR 50 and 70, respectively. However, due to slow enrollment, only 122 subjects were randomized and 121 treated and analyzed (one subject was randomized but never received treatment).
  • Demography and Baseline Characteristics [0489]
    TABLE 1
    Subject Disposition at Day 180
    CTLA4Ig + Placebo +
    etanercept etanercept TOTAL
    Randomized* 85 36 121
    Completed 68 (80%) 22 (61%)  90 (74%)
    Discontinued 17 (20%) 14 (39%)  31 (26%)
    Adverse Events  6 (7.0%)  1 (2.7%)  7 (6%)
    Lack of Efficacy  6 (7.0%) 12 (33%)  18 (15%)
    Other  5 (5.8%)  1 (2.7%)  6 (5%)
  • After six months, the proportion of total discontinuations were higher (39%) in the placebo plus etanercept treatment group compared to the CTLA4Ig plus etanercept group (20%). The difference was driven by a higher rate of discontinuation due to lack of efficacy in the placebo plus etanercept group (Table 1). [0490]
  • Demographic characteristics were similar between treatment groups. The majority of subjects were female and Caucasian. The mean duration of the disease was 13 years and the mean age was 52 years (Table 2). [0491]
    TABLE 2
    Mean Baseline Demographic and Clinical Characteristics
    CTLA4Ig + Placebo +
    etanercept etanercept TOTAL
    N = 85 N = 36 N = 121
    Mean Age: yrs (Range) 50 (24-74) 55 (28-72)  52 (24-74)
    Mean Weight: kg (Range) 81 (45-154) 79 (46-126)  81 (45-154)
    Gender: female: n (%) 66 (78%) 26 (72%)  92 (76%)
    Race: Caucasian-n (%) 80 (94%) 36 (100%) 116 (96%)
    Mean Duration of Disease: 13.0 ± 10.1 12.8 ± 8.6 13.0 ± 9.7
    yrs ± sd
    Tender Joints (out of 68)- 28.7 ± 14.0 29.5 ± 13.7 28.9 ± 13.8
    mean ± sd
    Swollen Joints-(out of 66)- 19.6 ± 9.4 20.3 ± 11.0 19.8 ± 9.9
    mean ± sd
  • Baseline clinical characteristics were similar between treatment groups including a mean of 29 tender joints and 20 swollen joints. With the exception of CRP values, which were lower, the baseline characteristics were typical of subjects with active rheumatoid arthritis and enrolled in clinical studies (Table 2). [0492]
  • ACR Responses and Core Components [0493]
  • The improvements in the [0494] ACR 20 and ACR 70 responses in the CTLA4Ig+etanercept group were statistically significant compared to the CTLA4Ig+placebo group (Table 3 and FIG. 63).
    TABLE 3
    Modified ACR Response at Day 180-number of subjects (%)*
    ACR 20 ACR 50 ACR 70
    CTLA4Ig + etanercept*** 48.2% 25.9% 10.6%
    Diff. from Placebo + etanercept 20.5%  6.4% 10.6%
    95% CI (1.2, 39.7) (−10.2, 23.1) (0.4, 20.8)
    p-Value  0.037**  0.448  0.042**
  • By two months of treatment, numerically higher responses on all components of the ACR criteria were observed for the CTLA4Ig plus etanercept group. Three of the seven ACR components are shown in FIGS. [0495] 64A-C.
  • The mean improvements in the individual components of the ACR criteria on [0496] Day 180 were consistently greater in the CTLA4Ig plus etanercept treatment group compared to the placebo plus etanercept group (Table 4).
    TABLE 4
    Mean Percent (SE) Improvement in Individual ACR
    Components at Day 180
    CTLA4Ig + Placebo +
    etanercept etanercept
    ACR Component N = 85 N = 36
    Tender Joints 42% (5.5) 24% (8.3)
    Swollen Joints 37% (5.0) 21% (8.1)
    Pain 34% (4.3) −1% (10.8)
    Physical Function (MHAQ) 31% (5.2) −5% (13.8)
    Subject Global Assessment 27% (5.4)  3% (9.5)
    Physician Global Assessment 43% (4.3) 27% (5.8)
  • Quality of Life [0497]
  • Compared to baseline, subjects in the CTLA4Ig plus etanercept group demonstrated statistically significant improvements at [0498] Day 180 in all 8 subscales of the SF-36-compared to only one (physical function) in subjects in the placebo plus etanercept group. The absolute changes in HRQOL subscales were considered clinically meaningful.
  • Compared to the placebo plus etanercept group, subjects in the CTLA4Ig plus etanercept group experienced statistically significantly greater improvement in 4 subscales of the SF-36: role-physical, bodily pain, vitality, and social function (FIG. 65). Improvements in the other 4 subscales were also greater than the placebo plus etanercept group, although they were not statistically significant. [0499]
  • Safety [0500]
  • No deaths or opportunistic infections occurred during the first six months of this study. Among the most frequently reported adverse events, headache, upper respiratory infection, musculo/skeletal pain, nausea/vomiting, hypertension, and diarrhea occurred at a higher rate in the CTLA4Ig plus etanercept group compared to the placebo plus etanercept group. Sinus abnormalities and rash were slightly higher in the CTLA4Ig plus etanercept group, as well. [0501]
  • More subjects in the CTLA4Ig plus etanercept group experienced serious adverse events (SAE) (7.1%) than the etanercept plus placebo group (2.8%). However, no SAEs were considered related to the study drug. [0502]
  • Two subjects receiving CTLA4Ig and etanercept had a dermatological malignancy. One subject had a basal cell carcinoma that was excised after the [0503] Day 150 visit. The other subject had a squamous cell carcinoma which was a pre-existing lesion that the subject decided to have removed after the Day 120 visit. Another subject experienced angioedema that was considered by the investigator to be a drug reaction to azithromycin.
  • All adverse events (AEs) leading to discontinuation were of either of mild or of moderate intensity. One discontinuation in the CTLA4Ig plus etanercept group, due to a tremor, was considered a serious adverse event. [0504]
  • Immunogenicity [0505]
  • No subjects receiving CTLA4Ig seroconverted for CTLA4Ig or CTLA4-T specific antibodies. No significant change in GMTs for CTLA4Ig or CTLA4-T specific antibodies was observed. [0506]
  • Comparison between CTLA4Ig/Etanercept and CTLA4Ig/Mlthotrexate ACR Responses [0507]
    TABLE 5
    CTLA4Ig + etanercept vs. CTLA4Ig + methotrexate
    ACR responses (% improvement):
    CTLA4Ig + Etanercepta CTLA4Ig + Methotrexateb
    (IM101-101) (IM101-100)
    2 mg/kg 0 mg/kg c 10 mg/kg 2 mg/kg 0 mg/kgc
    N = 85 N = 36 N = 115 N = 105 N = 119
    ACR 20 48.2%d 27.8% 60.0%d 41.9% 35.3%
    ACR
    50 29.3% 19.4% 36.5%d 22.9%d 11.8%
    ACR
    70 10.6%d   0% 16.5%d 10.5%d 1.7%
  • The efficacy of CTLA4Ig plus etanercept at 2 mg/kg was similar to that observed in subjects receiving the same dose of CTLA4Ig plus methotrexate therapy (Example 5). However, the criteria for evaluation in the methotrexate (Example 5) trial was the standard ACR, that includes CRP among the core components, while in the etanercept trial (Example 6) the criteria for evaluation was the modified ACR, that excludes CRP. [0508]
  • CONCLUSION
  • Preliminary assessment of the study at six months found that CTLA4Ig (2 mg/kg) in combination with etanercept reduced the signs and symptoms of rheumatoid arthritis, as compared with etanercept alone. The increases in the modified ACR20 and [0509] ACR 70 assays were statistically significant. Efficacy of CTLA4Ig plus etanercept therapy was observed within one month of the start of treatment. CTLA4Ig was generally safe and well tolerated when administered in combination with etanercept with the safety profile similar to etanercept therapy alone. CTLA4Ig was not immunogenic during the six month trial period. Additionally, the efficacy of CTLA4Ig therapy in combination with etanercept (Example 6) was similar to the same dose of CTLA4Ig with methotrexate (Example 5).
  • EXAMPLE 7
  • One-Year Results of a Phase IIB, Multicenter, Randomized, Double-Blind, Placebo-Controlled Study to Evaluate the Safety and Clinical Efficacy of Two Different Doses of BMS-188667 Administered Intravenously to Subjects with Active Rheumatoid Arthritis While Receiving Methotrexate. [0510]
  • The following example provides the one-year results from a Phase IIB, multi-center, randomized, double-blind, placebo controlled clinical study to evaluate the safety and clinical efficacy of administering two different doses of CTLA4Ig in combination with methotrexate to treat patients with active Rheumatoid Arthritis (RA). The study presented in this example is a continuance of the six-month study presented in Example 5. [0511]
  • Based on preliminary efficacy results from Example 3, supra, and the standard practice of adding other therapies to MTX in the treatment of RA, this study was designed to test the hypothesis that CTLA4Ig (BMS-188667) combined with MTX may have greater clinical efficacy when compared with MTX plus placebo in RA subjects who still have active disease despite MTX treatment. [0512]
  • The results presented in this clinical study report are based on data from an analysis performed after all subjects completed 6 months of treatment and again after all subjects completed 12 months of treatment. [0513]
  • Throughout this Example, the 10 mg/kg CTLA4Ig plus MTX group may be referred to as the 10 mg/kg group, the 2 mg/kg plus MTX group is referred to as the 2 mg/kg group, and the CTLA4Ig (BMS-188667) placebo plus MTX group is referred to as the placebo group. [0514]
  • Study Methodology [0515]
  • This study compared the clinical efficacy of two different doses (10 and 2 mg/kg) of CTLA4Ig (BMS-188667) combined with methotrexate (MTX) or with MTX plus placebo in subjects with active RA as assessed by ACR at 6 month and 12 month intervals. This study enrolled adult subjects with active RA who had had an inadequate response to MTX. [0516]
  • Results after one-year of monitoring subjects with active rheumatoid arthritis who were intravenously administere: 1) CTLA4Ig at a dosage of 2 mg/kg body weight with methotrexate, 2) CTLA4Ig at a dosage of 10 mg/kg body weight with methotrexate, or 3) a placebo with methotrexate (hereinafter known as placebo), are presented herein. [0517]
  • Subjects with active RA, despite treatment with MTX and who met the inclusion/exclusion criteria for this study were randomized 1:1:1 to receive one of the following treatments on a background of MTX therapy: CTLA4Ig (BMS-188667) 10 mg/kg, CTLA4Ig (BMS-188667) 2 mg/kg, or placebo. Subjects must have been treated with MTX (10 mg to 30 mg weekly) for at least 6 months, at a stable dose for 28 days prior to [0518] Day 1.
  • Treatment Groups: Subjects were randomized 1:1:1 to one of three treatment groups: [0519]
  • 1) Group 1: CTLA4Ig (BMS-188667) 10 mg/kg by intravenous infusion [0520]
  • 2) Group 2: CTLA4Ig (BMS-188667) 2 mg/kg by intravenous infusion [0521]
  • 3) Group 3: CTLA4Ig (BMS-188667) placebo by intravenous infusion [0522]
  • Infusion doses were based upon the subject's body weight from the pre-treatment visit immediately prior to the [0523] Day 1 visit (for a subject on MTX monotherapy, the weight was obtained at the screening visit; for a subject on MTX combination therapy [in combination with other DMARDs], the weight was obtained from the washout visit, Day −2). The infusion doses were not modified during Day 1 to Day 360.
  • Infusions were to occur at approximately the same time of day throughout the study. All doses of study medication were administered in a fixed volume of 75 mL at a constant rate over approximately 30 minutes. The intravenous bag and line were flushed with 25 mL of [0524] dextrose 5% in water (D5W) solution at the end of each infusion. All intravenous infusions were administered with the subject in the seated position. Subjects were observed for Adverse Events (Aes) and changes in vital signs (blood pressure, heart-rate, body temperature) from the start of each infusion (pre-dose, 15, 30, 45, 60, 75, 90, 120 minutes) and for a minimum of 2 hours after the start of the infusion. The observation period could be extended, if clinically indicated.
  • During the primary phase ([0525] Day 1 to Day 180) of the study, concomitant administration of selected medications was allowed. The permitted medications included:
  • MTX: Continued use of current dose (no increases, and decreases only for toxicity) [0526]
  • Systemic (non-topical) corticosteroids: Provided that the dose was stable and the total dose was less than or equal to the equivalent of [0527] prednisone 10 mg/day. Intra-articular injections were to be avoided; however, if necessary, up to two intra-articular injections were permitted. NOTE: A joint that received an intra-articular injection was counted as “active” in ALL subsequent assessments/evaluations.
  • NSAIDs, including ASA: Provided the dose was stable [0528]
  • Acetaminophen, combination products including acetaminophen and narcotic analgesics (i.e., acetaminophen with codeine phosphate, acetaminophen with propoxyphene napsylate, acetaminophen with oxycodone hydrochloride, acetaminophen with oxycodone bitartrate, etc.), or tramadol: For subjects experiencing pain not adequately controlled by baseline or study medication (except for 12 hours before a joint evaluation) [0529]
  • Table 1 is a schedule of study procedures and evaluations. [0530]
    TABLE 1
    Schedule of Study Procedures and Evaluations
    Treatment Period
    Pretreatment (Day) Treatment Daye,f,j,h
    Screen
    Visit Day (−28 to −2) (−2) 1 15 30 60 90 120 150 180 210 240 270 300 330 360
    Screening assessments
    Informed consent X
    Complete History and Physical X Xi
    CXR Xa
    ECG Xa X
    Stabilize/Withdraw prohibited X
    medications (if necessary)b
    Enroll Subject X Xm
    Randomize Subjectk X
    Dosingh X X X X X X X X X X X X X
    Interim Assessmentsf
    Duration of morning stiffness X X X X X X X X X X X X
    Interim History and Physical X X X X X X X X X X
    Tender joint count X X X X X X X X X X X X X
    Swollen joint count X X X X X X X X X X X X X
    Subject's assessment of pain X X X X X X X X X X X X
    Subject's global assess of disease X X X X X X X X X X X X
    activity
    Physician's global assess of X X X X X X X X X X X X
    disease activity
    Subject's assess of physical X X X X X X X X X X X X
    function
    Short form-36 health X X X X X
    questionnaire (SF-36)
    Subjects response to therapy X X X
    Safety Assessments
    Adverse event monitoring X X X X X X X X X X X X X Xo
    Weightg X X X
    Mammogram (females only)l X X
    Vital signs X X X X X X X X X X X X X X X
    Labs
    CBC X X X X X X X X X X X X X X X
    Chemistry panel X X X X X X X X X X X X X X X
    Urinalysis X X
    Urine/serum pregnancy testd X X X X X X X X X X X X X X X
    Hepatitis B surface antigen X
    Hepatitis C antibody X
    Pharmacodynamics (PD)
    Rheumatoid factor X X X X
    CRP X X X X X X X X X X X X X
    IL-2R X X X X X X X X X X
    Exploratory cytokines (ICAM-1 X X X X
    e-Selectin, IL-6 and TNFα)
    Pharmacokinetics X X X X
    Immunoglobulin determinations
    Quantitative immunoglobulins X X X
    (IgG, IgA, IgM)
    Immunogenicity
    Anti-BMS-188667Ab testing X X X X X X
    Radiographic assessmentsn
    X-rays (hands/wrists and feet) X X X
    # criteria, the DMARDs must have been washed out for at least 28 days prior to Day 1.
    # potential. Serum pregnancy test was to be processed locally.
    # identical to assessments performed on Day 360. The assessments for this visit replaced what might have
    # been scheduled on the day of discontinuation. Changes in current DMARD, steroid, or NSAIDs therapy
    # were not permitted until after these assessments were performed. Subjects were to be contacted 30 days
    # after discontinuation to capture safety data (adverse events).
    # study were be based on this weight.
    # Central Randomization System for randomization.
    # (documentation must be on file) prior to signing consent. Subjects who discontinued from the study after
    # Day 1 required a follow-up mammogram on the one year anniversary of the mammogram that was
    # performed during the screening period.
    # first nine months of treatment.
    # days after the last dose of study medication.
  • Efficacy Assessments [0531]
  • Clinical Measurements and Responses [0532]
  • Clinical response was assessed using the American College of Rheumatology (ACR) Core Data Set and Response Definitions. For this assessment, data were collected on seven components: 1) tender joint count (standardized 68 joint count); 2) swollen joint count (standardized 66 joint count); 3) subject global assessment of pain; 4) subject global assessment of disease activity; 5) physician global assessment of disease activity; 6) subject assessment of physical function (MHAQ); and 7) an acute phase reactant value CRP. [0533]
  • The [0534] ACR 20, ACR 50, and ACR 70 definition of response corresponds to a 20%, 50%, or 70% improvement, respectively, over baseline in tender and swollen joints (components 1 and 2) and a 20%, 50%, and 70% improvement, respectively, in three of the five remaining core data set measures (components 3 to 7). A Major Clinical Response is defined as maintenance of an ACR 70 response over a continuous 6-month period. See Table 1 for the days that data for each component was collected.
  • The primary efficacy analysis tested for differences in [0535] ACR 20 response between the two CTLA4Ig (BMS-188667) treatment groups and the placebo group at 6 months (Day 180). A sequential testing procedure was employed. First, a Chi-square test was used to compare the data for the 10 mg/kg CTLA4Ig group with the data for the placebo group at the 0.05 level of significance. If this was significant, the data for the 2 mg/kg CTLA4Ig group was compared with the placebo group at the 0.05 level. This testing procedure preserved the overall alpha level at 5%. Similar analyses were carried out for the ACR 50 and ACR 70 responses at 6 months. Differences in ACR 20, ACR 50 and ACR 70 responses between each CTLA4Ig (BMS-188667) treatment group and the placebo group were summarized using point estimates and 95% confidence intervals. Subjects who discontinued the study due to lack of efficacy (i.e., worsening RA) were considered ACR non-responders at all subsequent time points. For all subjects who discontinued for other reasons, their last ACR response was carried forward.
  • [0536] ACR 20, ACR 50, and ACR 70 response rates on Day 360 were compared between each CTLA4Ig (BMS-188667) treatment group and placebo at the Dunnett-adjusted 0.027 (two-tailed) level of significance.
  • The proportion of responders achieving an [0537] ACR 20 response at each time point was also plotted over time, and the Cochran Mantel-Haenszel test (W. G. Cochran, 1954, Some Methods of Strengthening the Common Chi-Square Test, Biometrics 10:417-451; N. Mantel and W. Haenszel, 1959, Biostatistical Aspects of the Analysis of Data from Retrospective Studies of Disease, J Nat Cancer Inst, 22:719-748) was used to compare the frequency of subjects achieving an ACR 20 response in each CTLA4Ig (BMS-188667) group versus the placebo group.
  • [0538] ACR 20, ACR 50, and ACR 70 responses on Days 15, 30, 60, 90, 120, 150, 180, 240, 300, and 360 were also presented for the two CTLA4Ig (BMS-188667) groups and the placebo group. The differences in ACR responses between the CTLA4Ig (BMS-188667) groups and placebo group were summarized using 95% confidence intervals. The ACR data plotted over time were used to assess onset-of-action and to determine time to maximal response.
  • A Major Clinical Response was defined as the maintenance of an [0539] ACR 70 response over a continuous 6-month period. At the 12-month analysis, the proportion of subjects who achieved a Major Clinical Response among the three groups was summarized.
  • In order to assess the integrity of the planned analyses, all subjects who received study medication and discontinued the study for any reason were considered ACR non-responders at all scheduled study visits subsequent to discontinuation. [0540]
  • The cumulative index, ACR-N, was evaluated at each follow-up assessment, and the AUC was assessed for up to 6 months and up to 12 months. The trapezoidal rule was used to compute the AUC. The ACR-N AUC was compared between the two CTLA4Ig (BMS-188667) treatment groups and the placebo group using an analysis of variance (ANOVA) for 6- and 12-month data. This allowed for the assessment of subject response throughout the study. These analyses were carried out on the LOCF data sets. [0541]
  • The distributional assumptions regarding the normality of the ACR-N AUC data were checked using the Shapiro-Wilks test on standardized residuals from the ANOVA model at the 10% level of significance. [0542]
  • Surrogate biomarkers were also used to assess the efficacy of the CTLA4Ig+MTX or placebo+MTX treatment regimens. Potential biomarkers for immunomodulation or inflammation in RA include CRP, soluble IL-2r, RF, soluble ICAM-1, E-selectin, serum IL-6, and TNFα. These parameters were summarized by treatment group, using frequencies and mean change from baseline to [0543] Day 180 and Day 360.
  • An Adverse Event (AE) was defined as any new or worsening illness, sign symptom or clinically significant laboratory test abnormality noted by the Investigator during the course of the study, regardless of causality. A serious adverse event (SAE) was defined as an AE that met any of the following criteria: was fatal; was life-threatening; resulted in or prolonged hospitalization; resulted in persistent or significant disability or incapacity, was cancer, was a congenital anomaly/birth defect, resulted in an overdose, resulted in the development of drug dependency or drug abuse, or was an important medical event. [0544]
  • Vital sign measurements were obtained at screening and at each study visit during and following study drug administration. Vital sign measurements (seated blood pressure, heart rate, and body temperature) were summarized by treatment group using means. [0545]
  • The two CTLA4Ig (BMS-188667) treatment groups (10 and 2 mg/kg) were compared with the placebo group. The primary analysis was the comparison of 6-month ACR response rate for 10 mg/kg and placebo groups, to be followed by the comparison of 2 mg/kg with placebo only if the former was significant. Sample sizes were based on a 5% level (two-tailed) of significance. The [0546] ACR 20 response rate for a placebo group at 6 months was estimated to be about 25% (Weinblatt M, Kremer J M, Bankhurst A D et. al. A trial of etanercept, a recombinant TNF:Tc fusion protein in patients with RA receiving methotrexate. NEJM 1999; 340: 253-259). A sample of 107 subjects per treatment group (adjusted for a possible 15% discontinuation rate) was determined to yield a 94% power to detect a difference of 25% at the 5% level (two-tailed). Table 2 summarizes the power needed to detect the specified treatment differences in ACR 20, ACR 50, and ACR 70 responses at 6 months.
    TABLE 2
    Response Rates and Power with 107a Subjects per Group
    Response Control Rate (%) Treatment Difference Power (%)
    ACR 20 25  25 94
    ACR 50 5 20 95
    ACR 70 1 14 90
  • If the primary comparisons of the 10 mg/kg CTLA4Ig with placebo were significant, then for the comparison of the 2 mg/kg CTLA4Ig with placebo groups, the power of the test would be at least 0.88, 0.90, and 0.81 for the [0547] comparison involving ACR 20, ACR 50, and ACR 70 responses at 6 months, respectively (Koch D D, Gansky S A. Statistical considerations for multiplicity in confirmatory protocols. Drug Info Journal 1996; 30: 523-534).
  • Statistical Analyses [0548]
  • Study Population [0549]
  • Disposition of Subjects [0550]
  • Of 524 subjects who were enrolled in this study, 339 subjects were randomized: 115 to the 10 mg/kg group; 105 to the 2 mg/kg group; and 119 to the placebo group (FIG. 68). The most frequent reason for not being randomized was failure to meet inclusion and/or exclusion criteria. [0551]
  • Primary Phase (Days 1-180) [0552]
  • A total of 256 subjects (75.5% of those randomized) completed the primary phase of the study; 83 subjects discontinued during this period (Table 3). Overall, discontinuation was more than 2-fold higher with placebo compared with 10 mg/kg CTLA4Ig group. Discontinuation due to lack of efficacy and discontinuation due to an AE were also more than 2-fold higher with placebo than with 10 mg/kg CTLA4Ig group. [0553]
    TABLE 3
    Reasons for Discontinuation: Primary Phase (Days 1-180)
    CTLA4Ig
    (BMS 188667)
    10 mg/kg 2 mg/kg Placebo Total
    No. Treated, n 115 105 119 339
    No. Discontinued, n (%)  17 (14.8)  25 (23.8)  41 (34.5)  83 (24.5)
    Adverse Event  3 (2.6)  7 (6.7)  9 (7.6)  19 (5.6)
    Lack of Efficacy  12 (10.4)  16 (15.2)  28 (23.5)  56 (16.5)
    Withdrawal of Consent  2 (1.7)  2 (1.9)  4 (3.4)  8 (2.4)
    Completed 180 Days of  98 (85.2)  80 (76.2)  78 (65.5) 256 (75.5)
    Therapy, n (%)
  • Cumulative Discontinuations (Days 1-360) [0554]
  • A total of 235 subjects (69.3% of those randomized) completed both phases of the study; 104 subjects discontinued by Day 360 (Table 4). The same general pattern in disontinuations noted in the primary phase (2-fold higher incidence with placebo compared with 10 mg/kg CTLA4Ig group) was also observed overall (Days 1-360). This included the overall discontinuation rate, discontinuations due to a lack of efficacy and discontinuations due to an AE. [0555]
    TABLE 4
    Reasons for Discontinuation: Both Phases (Days 1-360)
    CTLA4Ig
    (BMS-188667)
    10 mg/kg 2 mg/kg Placebo Total
    No. Treated, n 115 105 119 339
    No. Discontinued, n (%)  25 (21.7)  31 (29.5)  48 (40.3) 104 (30.7)
    Adverse Event  5 (43)b  9 (8.6)  11 (9.2)  25 (7.4)
    Death  0  1 (1.0)  0  1 (0.3)
    Lost to Follow-up  1 (0.9)  2 (1.9)  0  3 (0.9)
    Othera  1 (0.9)  0  1 (0.8)  2 (0.6)
    Lack of Efficacy  13 (11.3)  17 (16.2)  30 (25.2)  60 (17.7)
    Withdrawal of Consent  5 (4.3)  2 (1.9)  6 (5.0)  13 (3.8)
    Completed 360 Days of  90 (78.3)  74 (70.5)  71 (59.7) 235 (69.3)
    Therapy, n (%)
  • A Kaplan-Meier plot of the cumulative proportion of subjects who discontinued for any reason during the first 12 months is presented in FIG. 69; the cumulative proportion of subjects who discontinued due to lack of efficacy in presented in FIG. 70. Note that in both graphs after approximately 30 days of therapy, discontinuation rates with placebo were consistently higher compared with both CTLA4Ig (BMS-188667) groups. Additionally, after approximately 150 days of therapy, discontinuation rates for 2 mg/kg CTLA4Ig group were higher than those for 10 mg/kg. [0556]
  • Demography and Subject Characteristics [0557]
  • Overall, baseline demographic characteristics and baseline clinical RA characteristics were generally comparable across the three treatment groups and were typical of relatively advanced RA encountered in clinical practice (Table 5 and Table 6). The majority of subjects were white females approximately 55 years old with a mean duration of RA of approximately 9 to 10 years, a relatively large number of active joints (approximately 29 tender and 21 swollen joints) and visual analogue scores (VAS) approximately 59-65 mm (100 mm scale). [0558]
    TABLE 5
    Baseline Demographic Characteristics
    CTLA4Ig (BMS-188667)
    10 mg/kg 2 mg/kg Placebo
    No. Randomized 115 105 119
    Age (yrs)
    Mean ± SD (Range) 55.8 ± 12.5 (17, 83) 54.4 ± 11.3 (23, 80) 54.7 ± 12.0 (23, 80)
    Weight (kg)
    Mean ± SD (Range) 77.8 ± 18.6 (40.1, 144.0) 78.7 ± 21.4 (48.4, 186.8) 79.9 ± 17.6 (44.0, 140.0)
    Gender
    Males, n (%)  29 (25)  39 (37)  40 (34)
    Females, n (%)  86 (75)  66 (63)  79 (66)
    Race
    White, n (%) 100 (87)  91 (87) 104 (87)
    Black, n (%)  6 (5)  0  3 (3)
    Other, n (%)  9 (8)  14 (13)  12 (10)
    Duration of RA (yrs) n = 114a n = 105 n = 117a
    Mean ± SD (Range)  9.7 ± 9.8 (0, 38) 9.7 ± 8.1 (0, 36)  8.9 ± 8.3 (0, 41)
  • Although not a component of the ACR criteria, duration of morning stiffness was also assessed and was nearly 2-hours in each of the three groups. Positive results for RF at baseline were also assessed, and the CTLA4Ig (BMS-188667) treatment groups had higher percentages of subjects who tested positive for RF (86% for both the 10 mg/kg and 2 mg/kg CTLA4Ig groups compared to 76% for the placebo group). [0559]
    TABLE 6
    Baseline Clinical Rheumatoid Arthritis Characteristics
    CTLA4Ig (BMS-188667)
    10 mg/kg 2 mg/kg Placebo
    Characteristic (n = 115) (n = 105) (n = 119)
    Tender Joints, n 115 105 119
    Mean ± SD 30.8 ± 12.2  28.2 ± 12.0  29.2 ± 13.0
    Range 11.0, 66.0  3.0, 62.0  4.0, 68.0
    Swollen Joints, n 115 105 119
    Mean ± SD 21.3 ± 8.4  20.2 ± 8.9  21.8 ± 8.8
    Range  9.0, 54.0  4.0, 48.0  8.0, 64.0
    Pain (VAS 100 mm), n 113 104 119
    Mean ± SD 62.1 ± 21.4  64.3 ± 22.3  65.2 ± 22.1
    Range  0.0, 99.0  8.0, 100.0  3.0, 95.0
    Physical Function (MHAQ 115 105 119
    0-3), n
    Mean ± SD  1.0 ± 0.5  1.0 ± 0.5  1.0 ± 0.6
    Range  0.0, 2.5  0.0, 2.5  0.0, 2.3
    Subject Global Assess (VAS 113 105 119
    100 mm), n
    Mean ± SD 60.1 ± 20.7  59.4 ± 23.7  62.8 ± 21.6
    Range 10.0, 100.0  8.0, 99.0  4.0, 94.0
    MD Global Assess (VAS 100 113 105 119
    mm), n
    Mean ± SD 62.1 ± 14.8  61.0 ± 16.7  63.3 ± 15.5
    Range 20.0, 98.0  8.0, 95.0 18.0, 93.0
    CRP(mg/dL),n 112  99 115
    Mean ± SD  2.9 ± 2.8  3.2 ± 2.5  3.2 ± 3.2
    Range  0.2, 19.9  0.2, 10.8  0.2, 20.9
    Morning Stiffness (in 115 103 119
    minutes), n
    Mean ± SD 97.9 ± 63.1 104.1 ± 63.9 106.0 ± 64.2
    Range  0.0, 180.0  0.0, 180.0  0.0, 180.0
    Rheumatoid Factor (IU/mL),  99  90  90
    n
    % Positive 86% 86% 76%
  • Baseline demographics and RA characteristics of the overall population of subjects who had at least one dose of study drug and discontinued due to lack of efficacy were generally comparable to the entire study population, however, a greater proportion of subjects in this subpopulation had been diagnosed with RA for >10 years (45%) compared to the overall study population (34%). [0560]
  • Medical History Findings and Prior Medications [0561]
  • Medical history findings for subjects in this study were consistent with relatively advanced RA and were generally similar among treatment groups. The most frequently occurring findings (in >40% of the subjects) were musculoskeletal findings (not including RA symptoms; 59.3%), gastrointestinal findings (45.1%), and genitourinary findings (42.2%). Other important medical history findings included cardiovascular disease in approximately 39% of subjects in all treatment groups and endocrine/metabolic findings in approximately 29% of all subjects. [0562]
  • Overall use of MTX, systemic (non-topical) corticosteroids, DMARDs and biologic RA medications prior to entering the study was generally comparable across the three treatment groups (Table 7). All subjects were to have received prior treatment with rheumatic medications, including MTX, to be eligible for the study. Prior treatment with MTX was not recorded for 4 subjects. Systemic (non-topical) corticosteroid use prior to randomization was comparable among the three treatment groups, with slightly more subjects in the 2 mg/kg CTLA4Ig and placebo groups taking systemic (non-topical) corticosteroids (˜67-68%) compared to subjects in the 10 mg/kg CTLA4Ig group (60.0%). Use of other DMARDs and biologic RA medications prior to entering the study varied from 0 to 12% across treatment groups with no overall predominance in any treatment group. Mean dosing of MTX and of systemic (non-topical) corticosteroids on [0563] Day 1 were comparable among all three treatment groups (˜15-16 mg/wk, ˜6-7 mgs/day, respectively.
    TABLE 7
    Summary of Rheumatic Medications Prior to Enrollment
    CTLA4Ig
    (BMS-188667)
    10 mg/kg 2 mg/kg Placebo
    Prior Rheumatic Medication, n (%)a (n = 115) (n = 105) (n = 119)
    No. Subjects on Prior Medications 114 (99.1) 103 (98.1) 118 (99.2)
    Methotrexateb 114 (99.1) 103 (98.1) 118 (99.2)
    Systemic (non-topical) corticosteroids  69 (60.0)  71 (67.6)  80 (67.2)
    Other DMARDs  19 (16.5)  19 (18.1)  25 (21.0)
    Sulfasalazine  9 (7.8)  2 (1.9)  10 (8.4)
    Hydroxychloroquine  8 (7.0)  6 (5.7)  14 (11.8)
    Cyclosporine  2 (1.7)  4 (3.8)  4 (3.4)
    Infliximab  2 (1.7)  2 (1.9)  2 (1.7)
    Etanercept  1 (0.9)  4 (3.8)  1 (0.8)
    Chloroquine  1 (0.9)  0  0
    Leflunomide  0  2 (1.9)  2 (1.7)
  • Study Therapy [0564]
  • Of the three treatment groups, the 10 mg/kg CTLA4Ig group had the longest mean duration of exposure for both study phases and the placebo group had the shortest mean duration of exposure for both study phases (Day 180: 163 days, 156 days, 140 days; Day 360: 286 days, 268 days, and 234 days; 10 mg/kg, 2 mg/kg, and placebo, respectively). [0565]
  • At Day 180 (end of the primary phase), the proportion of subjects receiving infusions was higher in the 10 mg/kg CTLA4Ig group (85%) compared with the 2 mg/kg CTLA4Ig group (79%) and the placebo group (66%) (Table 8). At Day 330 (day of last scheduled infusion in the secondary phase), the proportion of subjects receiving infusions was also higher in the 10 mg/kg CTLA4Ig group (78%) compared with the 2 mg/kg CTLA4Ig group (70%) and the placebo group (59%). [0566]
    TABLE 8
    Subjects Who Received Infusions on Given Study Days
    Number (%) of Subjects
    CTLA4Ig (BMS-188667)
    10 mg/kg 2 mg/kg Placebo
    Day (n = 115) (n = 105) (n = 119)
     1 115 (100) 105 (100) 119 (100)
     15 114 (99) 104 (99) 117 (98)
     30 113 (98) 101 (96) 111 (93)
     60 108 (94)  97 (92) 103 (87)
     90 106 (92)  94 (90)  94 (79)
    120 100 (87)  86 (82)  83 (70)
    150  98 (85)  83 (79)  81 (68)
    180  98 (85)  83 (79)  78 (66)
    210  94 (82)  80 (86)  78 (66)
    240  95 (83)  78 (74)  76 (64)
    270  93 (81)  77 (73)  73 (61)
    300  90 (78)  74 (70)  72 (61)
    330  90 (78)  73 (70)  70 (59)
  • Methotrexate [0567]
  • Subjects were to have been treated with a “stable” dose of MTX (10-30 mg weekly) for at least 6 months, for 28 days prior to [0568] Day 1. With the exception of 4 subjects, all subjects received between 10 and 30 mg of MTX weekly in addition to CTLA4Ig (BMS-188667) during the primary phase (Day 1-180). During the secondary phase (Day 181-360), the dose of MTX could have been adjusted provided it remained between 10 and 30 mg
  • Measurements of Treatment Compliance [0569]
  • During the primary phase, the number of missed infusions of study drug was <2 at any time table (Table 9). During the secondary phase, subjects in the placebo group appeared to have missed slightly fewer infusions than subjects in the CTLA4Ig (BMS-188667) groups. However, more placebo than CTLA4Ig (BMS-188667) subjects discontinued by these later time points (see supra). [0570]
    TABLE 9
    Number of Missed Infusions of Study Drug
    CTLA4Ig (BMS-188667)
    10 mg/kg 2 mg/kg Placebo
    (n = 115) (n = 105) (n = 119)
    Day 1 0 0 0
    Day 15 1 1 0
    Day 30 0 1 1
    Day 60 0 0 0
    Day 90 0 0 0
    Day 120 0 1 2
    Day 150 1 2 0
    Day 180 1 0 1
    Day 210 4 0 0
    Day 240 1 2 1
    Day 270 0 2 0
    Day 300 1 1 0
    Day 330 0 0 0
  • Concomitant Therapy [0571]
  • Systemic (non-topical) corticosteroid use was generally comparable among the three groups during screening/enrollment (58-67%) and during the primary phase of the study (67-71%), Tables 10 and 11, respectively. While corticosteroid use decreased in all three treatment groups by [0572] Day 360, more subjects in the 10 mg/kg CTLA4Ig group took systemic (non-topical) corticosteroids (63.5%) compared to the other two treatment groups (53.3% and 45.4% for the 2 mg/kg CTLA4Ig and placebo groups, respectively). Several subjects (CTLA4Ig: 0-3%, placebo: 0-10%) received DMARDs other than MTX during screening/enrollment.
    TABLE 10
    Summary of Rheumatic Concomitant Medications During
    Screening/Enrollment
    CTLA4Ig
    (BMS-188667)
    10 mg/kg 2 mg/kg Placebo
    Rheumatic Medication, n (%)a (n = 115) (n = 105) (n = 119)
    No. Subjects on Prior Medications 114 (99.1) 103 (98.1) 118 (99.2)
    Methotrexate 114 (99.1) 103 (98.1) 118 (99.2)
    Systemic (non-topical) corticosteroids  67 (58.3)  70 (66.7)  75 (63.0)
    Other DMARDs  5 (4.3)  6 (5.7)  14 (11.8)
    Sulfasalazine  3 (2.6)  1 (1.0)  4 (3.4)
    Hydroxychloroquine  2 (1.7)  3 (2.9)  12 (10.1)
    Cyclosporine  1 (0.9)  1 (1.0)  2 (1.7)
    Etanercept  0  1 (1.0)  0
  • [0573]
    TABLE 11
    Subjects Who Received Clinically Relevant Concomitant
    Medications During Both Study Phases
    CTLA4Ig
    (BMS-188667)
    10 mg/kg 2 mg/kg Placebo
    Medicationa (n = 115) (n = 105) (n = 119)
    Systemic (non-topical) corticosteroids 77 (67.0) 71 (67.6) 85 (71.4)
    (Primary Phase)
    Systemic (non-topical) corticosteroids 73 (63.5) 56 (53.3) 54 (45.4)
    (Secondary Phase)
  • Efficacy Results [0574]
  • The CTLA4Ig (BMS-188667) 10 mg/kg group had superior efficacy compared to the placebo group at [0575] Day 180 and Day 360. For the 2 mg/kg CTLA4Ig group, results for some efficacy parameters were significantly better compared to the placebo group, results for most other efficacy parameters were numerically higher compared to placebo.
  • ACR Responses at [0576] Day 180
  • Analysis of the primary efficacy variable for this study, ACR20 response rate at [0577] Day 180, showed that the 10 mg/kg CTLA4Ig group was significantly (p<0.001) more effective than placebo (Table 12, FIG. 71A and FIG. 71B).
  • The ACR50 and ACR70 responses at [0578] Day 180 for the 10 mg/kg CTLA4Ig group were also significantly higher compared to the placebo group (Table 12, FIG. 71A and FIG. 71B). The ACR50 and the ACR70 responses at Day 180 for the 2 mg/kg CTLA4Ig group were significantly higher compared to the placebo group. The ACR20 response at Day 180 for the 2 mg/kg CTLA4Ig group was slightly higher compared to the placebo group; however, no statistically significant differences were observed.
    TABLE 12
    ACR Responses at Day 180
    CTLA4Ig (BMS-188667)
    10 mg/kg 2 mg/kg Placebo
    (n = 115) (n = 105) (n = 119)
    ACR 20
    n (%)   70 (60.9)   44 (41.9) 42 (35.3)
    CI 25.6 (12.8, 38.4)  6.6 (−6.2, 19.4) N/A
    p-value <0.001a  0.31 N/A
    ACR 50
    n (%)   42 (36.5)   24 (22.9) 14 (11.8)
    CI 24.8 (13.8, 35.7) 11.1 (1.2, 20.9) N/A
    p-value <0.001a  0.027a N/A
    ACR 70
    n (%)   19 (16.5)   11 (10.5)  2 (1.7)
    CI 14.8 (7.5, 22.2)  8.8 (2.7, 14.9) N/A
    p-value <0.001a  0.005a N/A
  • ACR Responses at [0579] Day 360
  • At [0580] Day 360, ACR20, ACR50 and ACR70 responses for the 10 mg/kg CTLA4Ig group were significantly (p<0.001) higher compared to the placebo group (Table 13, FIG. 72A and FIG. 72B). Although the same response rates for the 2 mg/kg CTLA4Ig group were numerically higher compared to the placebo group, these differences were not statistically significant.
    TABLE 13
    ACR Responses at Day 360
    CTLA4Ig (BMS-188667)
    10 mg/kg 2 mg/kg Placebo
    (n = 115) (n = 105) (n = 119)
    ACR 20
    n (%)   72 (62.6)  43 (41.0) 43 (36.1)
    CI 26.5 (13.7, 39.3) 4.8 (−7.9, 17.6) N/A
    p-value <0.001a 0.459 N/A
    ACR 50
    n (%)   48 (41.7)  23 (21.9) 24 (20.2)
    CI 21.6 (9.7, 33.4) 1.7 (−8.9, 12.4) N/A
    p-value <0.001a 0.75 N/A
    ACR 70
    n (%)   24 (20.9)  13 (12.4)  9 (7.6)
    CI 13.3 (4.4, 22.2) 4.8 (−3.0, 12.6) N/A
    p-value  0.003a 0.227 N/A
  • ACR Responses by Visit [0581]
  • For the comparison of the 10 mg/kg CTLA4Ig group to the placebo group, statistically significant improvements were observed for all three response rates ([0582] ACR 20, ACR 50, and ACR 70) by Day 90, and these values remained statistically significant at every time point up to and including Day 360 (p<0.008 for all three ACR response rates) (FIG. 73A, FIG. 73B, and FIG. 73C). In fact, statistically significant improvements in ACR 50 and ACR 70 response for the 10 mg/kg CTLA4Ig group occurred as early as Day 30 (p=0.039 and p=0.04, respectively).
  • For the 2 mg/kg CTLA4Ig group, statistically significant improvements compared to placebo were observed in [0583] ACR 50 and ACR 70 responses at Day 180 (p=0.027 and p=0.005, respectively). At Day 360, improvements in ACR response were slightly greater in the 2 mg/kg CTLA4Ig group compared to the placebo group; however, no statistically significant differences were observed.
  • After adjusting for visit using the Cochran-Mantel Haenszel test, a significant difference in [0584] ACR 20 response was observed for the 10 mg/kg CTLA4Ig group compared to the placebo group at both Day 180 and Day 360. No significant difference was observed between the 2 mg/kg CTLA4Ig and placebo groups at both timepoints. Similar results were obtained for ACR 50 response at both time points. For ACR 70 response at both time points, a significant difference was observed for both CTLA4Ig (BMS-188667) treatment groups compared to the placebo group.
  • Summary of Major Clinical Response [0585]
  • Major Clinical Response was defined as maintenance of an [0586] ACR 70 response over a continuous 6-month period. The percentages of subjects who achieved a Major Clinical Response at Day 360 were significantly higher in both the 10 mg/kg and 2 mg/kg CTLA4Ig groups (7.8% and 5.7%, respectively) when compared to the placebo group (0.8%; p=0.008 and 0.036, respectively) (Table 14).
    TABLE 14
    Summary of Major Clinical Response by Day 360
    CTLA4Ig (BMS-188667)
    10 mg/kg 2 mg/kg Placebo
    (n = 115) (n = 105) (n = 119)
    No. Subjects with a Major   9 (7.8)   6 (5.7) 1 (0.8)
    Response
    Diff (CI) 7.0 (1.8, 12.2) 4.9 (0.3, 9.4) N/A
    p-value 0.008a 0.036a N/A
  • Mean Numeric ACR (ACR-N) and ACR-N Area Under the Curve (ACR-N-AUC) [0587]
  • Overall, mean numeric ACR (ACR-N) for all treatment groups increased over time during the first 6 months of the study (FIG. 74). During the second 6 months, mean ACR-N increased slightly with 10 mg/kg CTLA4Ig, but remained relatively unchanged with 2 mg/kg CTLA4Ig and placebo. At each study visit, the ACR-N was consistently higher for the 10 mg/kg CTLA4Ig group compared to the 2 mg/kg CTLA4Ig and placebo groups. [0588]
  • Compared to the placebo group, the differences in values for ACR-N AUC (area under the curve) for the 10 mg/kg CTLA4Ig group was significantly (p<0.001) higher by [0589] Day 360.
  • Percentage Improvement from Baseline at [0590] Day 180
  • For the 10 mg/kg CTLA4Ig group, improvements in each individual ACR component (tender and swollen joint counts, CRP, pain, subject global assessment, physician global assessment, and physical function) at [0591] Day 180 were statistically significant relative to improvements for the placebo group (Table 15).
  • For the 2 mg/kg CTLA4Ig group, statistically significant improvements compared to the placebo group were observed in physician global assessment and CRP at [0592] Day 180. Furthermore, CRP levels in the placebo group actually worsened at Day 180. Change from baseline in mean duration of morning stiffness was comparable among the three treatment groups at Day 180.
    TABLE 15
    Mean Percentage Improvement from Baseline at Day 180
    (Individual Components of ACR Criteria)
    CTLA4Ig (BMS-188667)
    10 mg/kg 2 mg/kg Placebo
    Component (n = 115) (n = 105) (n = 119)
    Tender Joints n = 114 n = 104 n = 118
    Mean % Change 59.78* 43.15   31.88
    Swollen Joints n = 114 n = 104 n = 118
    Mean % Change 55.28* 45.34*   33.49
    CRP n = 108 n = 98 n = 114
    Mean % Change 31.79* 16.41* −23.43
    Pain n = 109 n = 102 n = 118
    Mean % Change 46.19* 22.09*    8.20
    Subject Global Assessment n = 111 n = 103 n = 118
    Mean % Change 40.76*  9.07   17.48
    MD Global Assessment n = 111 n = 103 n = 116
    Mean % Change 51.91* 38.71*   25.14
    Physical Function n = 107 n = 98 n = 110
    Mean % Change 41.21* 21.63   13.71
    Duration Morning Stiffness n = 98 n = 82 n =80
    Mean ± SD (minutes) 61.9 ± 55.4 60.8 ± 66.1 55.9 ± 66.2
  • Percentage Improvement from Baseline at [0593] Day 360
  • For the 10 mg/kg CTLA4Ig group, improvements in each individual ACR component (tender and swollen joint counts, CRP, pain, subject global assessment, physician global assessment, and physical function) at [0594] Day 360 were statistically significant relative to improvements for the placebo group. Mean percentage improvements from baseline to Day 360 are presented in Table 16 for all clinical parameters of the ACR criteria.
  • For the 2 mg/kg CTLA4Ig group, statistically significant improvements compared to the placebo group were observed in physician global assessment and CRP at [0595] Day 360. Furthermore, CRP levels in the placebo group actually worsened at Day 360. At Day 360, the CTLA4Ig (BMS-188667) treatment groups had greater changes from baseline in duration of morning stiffness compared to the placebo group.
    TABLE 16
    Mean Percentage Improvement from Baseline at Day 360
    (Individual Components of ACR Criteria)
    CTLA4Tg (BMS-188667)
    10 mg/kg 2 mg/kg Placebo
    Component (n = 115) (n = 105) (n = 119)
    Tender Joints n = 115 n = 105 n=119
    Mean % Change 66.39* 43.54*   29.97
    Swollen Joints n = 115 n = 105 n = 119
    Mean % Change 59.74* 46.40   36.17
    CRP n = 112 n = 98 n = 115
    Mean % Change 27.59* 10.31* −31.26
    Pain n = 112 n = 104 n = 119
    Mean % Change 44.93* 26.26   12.55
    Subject Global Assessment n = 113 n = 105 n = 119
    Mean % Change 41.01* 16.08    1.99
    MD Global Assessment n = 113 n = 105 n = 119
    Mean % Change 53.48* 37.87*   24.14
    Physical Function n = 109 n = 100 n = 111
    Mean % Change 42.32* 22.94   10.25
    Duration Morning Stiffness n = 88 n = 71 n = 72
    Mean ± SD 66.2 ± 59.5* 66.6 ± 72.2 49.7 ± 73.9
  • New Active Joints [0596]
  • The proportion of new active joints was determined using the validated 28-joint count (out of 68 total tender joints and out of 66 total swollen joints) proposed by Smollen et al (Smollen J S, Breedveld F C, Eberl G, Jones I et al. Validity and reliability of the twenty-eight joint count for the assessment of RA activity. Arthritis & Rheum 1993; 38: 38-43). The proportion of new active joints (both tender and swollen) at [0597] Day 180 was lowest for subjects receiving 10 mg/kg CTLA4Ig (FIG. 75).
  • At [0598] Day 180, the percentages of subjects reporting no new tender joints and no new swollen joints was highest in the 10 mg/kg CTLA4Ig group (FIG. 76A, FIG. 77A). The percentage of subjects who reported no new tender joints and no new swollen joints was approximately 59% and 52%, respectively, in the 10 mg/kg CTLA4Ig group; 38% and 44%, respectively, in the 2 mg/kg CTLA4Ig group; and 41% and 37%, respectively, in the placebo group.
  • The proportion of new active joints (both tender and swollen) at [0599] Day 360 was lowest for subjects receiving 10 mg/kg CTLA4Ig (FIG. 78). This pattern for the proportion of new active joints mirrored the pattern seen at Day 180.
  • Similarly, at [0600] Day 360, the proportion of subjects reporting no new tender joints and no new swollen joints was highest in the 10 mg/kg CTLA4Ig group (FIG. 76B, and FIG. 77B). The percentage of subjects who reported no new tender and no new swollen joints was approximately 71% and 61%, respectively, in the 10 mg/kg CTLA4Ig group; 41% and 44%, respectively, in the 2 mg/kg CTLA4Ig group; and 42% for both counts in the placebo group.
  • Improvement in Clinical Parameters Among Subjects with an ACR Response [0601]
  • Among [0602] ACR 20, ACR 50, and ACR 70 responders, improvement in the core components of the ACR criteria were slightly greater for the two CTLA4Ig (BMS-188667) treatment groups compared to placebo.
  • The onset of action for subjects who received the 10 mg/kg CTLA4Ig dose occurred after approximately 15 days, with significant increases in [0603] ACR 20 improvement occurring at ≧Day 60 for ACR50 at ≧Day 90 for ACR70 at ≧Day 30 and in each instance, continuing until Day 360 (see FIG. 73A, FIG. 73B and FIG. 73C).
  • Changes from Baseline for the Health Outcomes Short Form Questionnaire (SF-36) [0604]
  • The impact of CTLA4Ig (BMS-188667) on health-related quality of life was assessed using the Health Outcomes Short Form Questionnaire SF-36 (summary scores range from 0 to 100 with higher scores indicating a better quality of life). Analyses were performed on the LOCF (last observation carried forward) data set as well as the as the observed data set. [0605]
  • For the 10 mg/kg CTLA4Ig group, statistically significant improvement from baseline compared to the placebo group was observed in all four mental health and all four physical health domains of the SF-36 at [0606] Day 180, using the LOCF analysis (i.e., 95% CIs did not include 0) (FIGS. 79A, 79B). For the 2 mg/kg CTLA4Ig group, there were numerical improvements in the mental health or physical health domains compared to placebo at Day 180, however, these improvements were not statistically significant.
  • Results of analyses performed on the as-observed data set were similar to those observed for the LOCF data set except that the “role emotional” domain at [0607] Day 180 was not significantly improved (but was numerically improved) for the comparison between the 10 mg/kg CTLA4Ig and placebo groups using the as-observed data set.
  • The physical component and the mental health component summary measures at [0608] Day 180 are shown in Table 17.
    TABLE 17
    Mean Change from Baseline to Day 180 for the SF-36 (Physical
    and Mental Health Components)
    CTLA4Ig (BMS-188667)
    10 mg/kg 2 mg/kg Placebo
    Summary Score (n = 115) (n = 105) (n = 119)
    Mental Health Component n = 115 n = 103 n = 118
    Baseline Mean 44.52 43.06 41.75
    Postbaseline Mean 48.69 45.59 44.04
    Mean Change from Baseline  4.17  2.53  2.30
    95% CI (2.46, 5.88) (0.39, 4.67) (0.42, 4.17)
    Physical Component n = 115 n = 103 n = 118
    Baseline Mean 31.13 30.80 32.33
    Postbaseline Mean 39.30 35.47 35.21
    Mean Change from Baseline  8.16  4.67  2.88
    95% CI (6.33, 9.99) (3.25, 6.09) (1.54, 4.22)
  • Results of the Health Outcomes at [0609] Day 360 were similar to those seen at Day 180. For the 10 mg/kg CTLA4Ig group, statistically significant improvements from baseline compared to the placebo group were observed in all four mental and all four physical domains of the SF-36 at Day 360, using the LOCF analysis (i.e., 95% CIs did not include 0) (FIGS. 80A, and 80B). For the 2 mg/kg CTLA4Ig group, a statistically significant difference in three of four physical domains at Day 360 and one of four mental domains at Day 360 compared to the placebo group was observed.
  • Results of analyses performed on the as-observed data set were similar to those observed for the LOCF data set. [0610]
  • The physical component and mental health component summary measures at [0611] Day 360 is shown in Table 18.
    TABLE 18
    Mean Change from Baseline to Day 360 for the SF-36
    (Summaries of Physical Component and Mental Health
    Component)
    CTLA4Ig (BMS-188667)
    10 mg/kg 2 mg/kg Placebo
    Summary Score (n = 115) (n = 105) (n = 119)
    Mental Health Component n = 115 n = 103 n = 118
    Baseline Mean 44.52 43.06 44.75
    Postbaseline Mean 48.83 45.65 43.22
    Mean Change from Baseline  4.31  2.59  1.47
    95% CI (2.64, 5.98) (0.64, 4.55) (−0.14, 3.08)
    Physical Component n = 115 n = 103 n = 118
    Baseline Mean 31.13 30.80 32.33
    Postbaseline Mean 38.93 36.49 34.93
    Mean Change from Baseline  7.79  5.69  2.60
    95% CI (5.90, 9.68) (4.10, 7.28) (1.09, 4.11)
  • Biomarker and Pharmacodynamic Data [0612]
  • There were significant improvements (decreases) in 5 of the 6 biomarker/pharmacodynamic (PD) parameters with 10 mg/kg CTLA4Ig at Day 180 (soluble IL-2r, rheumatoid factor (RF), ICAM-1, E-selectin and IL-6) and a numerical decrease in TNF-α (Table 19). There were significant improvements (decreases) in 3 of the 6 biomarker/PD parameters with 2 mg/kg CTLA4Ig at Day 180 (soluble IL-2r, RF and IL-6) and a numerical improvement in ICAM-1. There were no significant changes in any of the biomarker/PD parameters with placebo at [0613] Day 180. There appears to be a dose response relationship with the improvements (decreases) in biomarker/PD parameters.
    TABLE 19
    Pharmacodynamic Measures at Day 180
    CTLA4Ig (BMS-188667)
    10 mg/kg 2 mg/kg Placebo
    Parameter (n = 115) (n = 105) (n = 119)
    Soluble IL-2r n = 95 n = 84 n = 76
    (Normal range: 640-2543 pg/mL)
    Baseline Mean (± SD) 1426.19 ± 751.76 1396.82 ± 610.21 1429.13 ± 667.84
    Postbaseline Mean (± SD) 1112.62 ± 699.68 1261.31 ± 473.66 1470.03 ± 637.75
    Mean Change −316.23 −135.51 43.59
    95% CI (−417.73, −214.72) (−241.48, −29.53) (−71.24, 158.43)
    Rheumatoid Factor n = 95 n = 84 n = 74
    (Normal Range: 0-20 IU/mL)
    Baseline Mean (± SD)  289.71 ± 401.95  256.19 ± 307.92  196.11 ± 265.48
    Postbaseline Mean (± SD)  185.43 ± 269.52  227.82 ± 276.27  204.36 ± 320.09
    Mean Change −104.27  −28.12 −0.62
    95% CI (−151.53, −57.01) (−52.13, −4.11) (−31.67, 30.43)
    ICAM-1 n = 95 n = 82 n = 75
    Baseline Mean (± SD)  404.89 ± 137.72  393.47 ± 150.85  387.33 ± 230.93
    Postbaseline Mean (± SD)  364.74 ± 109.47  387.25 ± 142.73  386.17 ± 163.82
    Mean Change  −40.42  −6.22  1.09
    95% CI (−58.06, −22.78) (−27.49, 15.05) (−31.88, 34.05)
    E-selectin n = 89 n = 80 n = 71
    Baseline Mean (± SD)  68.07 ± 32.93  67.32 ± 37.13  68.23 ± 43.09
    Postbaseline Mean (± SD)  61.01 ± 31.53  67.86 ± 40.20  67.37 ± 35.66
    Mean Change  −8.41   0.54 −0.68
    95% CI (−13.24, −3.58) (−5.95, 7.03) (−6.87, 5.51)
    Serum IL-6 n = 86 n = 74 n = 69
    (Normal Range: 0.3-14.8 pg/mL)
    Baseline Mean (± SD)  28.47 ± 38.28  31.75 ± 42.29  21.20 ± 26.51
    Postbaseline Mean (± SD)   9.25 ± 15.85  16.00 ± 22.13  23.98 ± 37.92
    Mean Change  −20.30  −16.10  1.98
    95% CI (−27.55, −13.06) (−24.20, −8.00) (−7.21, 11.17)
    TNFα n = 84 n = 74 n = 69
    (1.2-8.0 pg/mL)
    Baseline Mean (± SD)  11.17 ± 23.72   7.51 ± 13.25  13.12 ± 23.20
    Postbaseline Mean (± SD)   7.57 ± 7.90   6.20 ± 4.48   9.59 ± 11.21
    Mean Change   −3.66  −1.21 −3.54
    95% CI (−8.62, 1.30) (−4.32, 1.90) (−7.82, 0.75)
  • Overall, the pattern in the changes in biomarker/PD data at [0614] Day 360 were similar to that seen at Day 180. There were significant improvements (decreases) in 5 of the 6 biomarker/PD parameters with 10 mg/kg CTLA4Ig at Day 360 (soluble IL-2r, RF, ICAM-1, E-selectin and IL-6) and a numerical, but not statistically significant improvement observed for TNF-α (Table 20). There was a significant improvement (decrease) in IL-6 only with 2 mg/kg CTLA4Ig at Day 360, however, numerical improvements were seen with RF and ICAM-1. There were no significant changes in any of the biomarker/PD parameters with placebo at Day 360. As seen with Day 180 data, it appeared that all of the improvements (decreases) in biomarker/PD parameters occurred in a dose response manner.
  • A comparison of the postbaseline means for the biomarker/PD parameters at [0615] Day 180 to those at Day 360 reveals important trends. For the 10 mg/kg CTLA4Ig group, all biomarkers/PD measures continued to decrease, with the exception of soluble IL-2r which increased slightly. For the 2 mg/kg CTLA4Ig group, mean values for 3 of the biomarkers/PD parameters either decreased slightly (ICAM-1, serum IL-6) or remained relatively constant (E-selectin) and mean values for the other 3 biomarkers/PD measures increased slightly (soluble IL-2r, RF, TNF α). For the placebo group, mean values for all of the biomarkers/PD parameters increased slightly at Day 360, with the exception of TNF α which remained relatively unchanged.
    TABLE 20
    Pharmacodynamic Measures at Day 360
    CTLA4Ig (BMS-188667)
    10 mg/kg 2 mg/kg Placebo
    Measure (n = 115) (n = 105) (n = 119)
    Soluble IL-2r n = 68 n = 56 n = 55
    (Normal range: 640-2543 pg/mL)
    Baseline Mean (± SD) 1372.10 ± 770.11 1373.86 ± 567.75 1459.93 ± 695.07
    Postbaseline Mean (± SD) 1185.51 ± 638.95 1413.84 ± 452.50 1666.59 ± 611.97
    Mean Change −194.31  39.99 206.22
    95% CI (−305.67, −82.96) (−69.87, 149.84) (35.88, 376.56)
    Rheumatoid Factor n = 69 n = 55 n = 58
    (Normal Range: 0-20 IU/mL)
    Baseline Mean (± SD)  261.43 ± 333.58  258.42 ± 318.65  179.12 ± 207.72
    Postbaseline Mean (± SD)  143.13 ± 180.80  236.61 ± 287.36  206.42 ± 256.27
    Mean Change −118.30 −25.64  20.90
    95% CI (−175.19, −61.42) (−58.50, 7.23) (−10.72, 52.51)
    ICAM-1 n = 77 n = 68 n = 64
    Baseline Mean (± SD)  406.44 ± 145.22  393.41 ± 132.97  405.67 ± 245.16
    Postbaseline Mean (± SD)  354.90 ± 111.40  380.42 ± 113.20  405.07 ± 194.15
    Mean Change  −55.15 −12.98  1.47
    95% CI (−74.80, −35.49) (−35.36, 9.39) (−26.41, 29.35)
    E-selectin n = 75 n = 68 n = 62
    Baseline Mean (± SD)  68.84 ± 34.38  66.75 ± 37.10  69.72 ± 44.38
    Postbaseline Mean (± SD)  58.77 ± 26.61  67.58 ± 31.50  71.90 ± 47.43
    Mean Change  −10.89  0.83  2.34
    95% CI (−15.70, −6.08) (−5.62, 7.28) (−4.53, 9.20)
    Serum IL-6 n = 56 n = 47 n = 48
    (Normal Range: 0.3-14.8 pg/mL)
    Baseline Mean (± SD)  27.68 ± 38.56  27.19 ± 32.45  17.27 ± 22.47
    Postbaseline Mean (± SD)   7.64 ± 14.21  13.93 ± 19.00  17.72 ± 29.76
    Mean Change  −20.88 −12.72  −0.19
    95% CI (−31.56, −10.19) (−22.49, −2.94) (−7.55, 7.18)
    TNFa n = 61 n = 48 n = 50
    (1.2-8.0 pg/mL)
    Baseline Mean (± SD)   9.71 ± 22.80   6.27 ± 3.62  10.81 ± 21.24
    Postbaseline Mean (± SD)   6.67 ± 4.80   7.18 ± 8.14   9.36 ± 26.43
    Mean Change  −3.02  1.08  −1.41
    95% CI (−8.70, 2.67) (−1.26, 3.42) (−5.14, 2.33)
  • The data are shown graphically for these biomarker/PD measures, as well as for changes in CRP levels, in FIGS. 81 through 87. [0616]
  • In order to assess the integrity of the planned analyses, all subjects who received study medication and discontinued the study for any reason were considered ACR non-responders at all scheduled study visits subsequent to discontinuation. Results of these analyses (Table 21) were consistent with the efficacy results already presented. The proportion of subjects who received 10 mg/kg CTLA4Ig and achieved an [0617] ACR 20, ACR 50, or ACR 70 response at Day 180 was significantly (p<0.001) higher compared to the proportion of subjects who received placebo. For the 2 mg/kg CTLA4Ig group, a significantly (p≦0.009) higher proportion of subjects achieved either an ACR 50 or ACR 70 response.
    TABLE 21
    ACR Response at Day 180 (Non-Completer Equals
    Non-Responder)
    CTLA4Ig (BMS-188667)
    10 mg/kg 2 mg/kg Placebo
    (n = 115) (n = 105) (n = 119)
    ACR 20, n (%)   67 (58.3)   41 (39.0) 38 (31.9)
    Diff (CI) 26.3 (13.6, 39.1)  7.1 (−5.4, 19.7) N/A
    p-value <0.001a 0.266 N/A
    ACR
    50, n (%)   41 (35.7)   24 (22.9) 12 (10.1)
    Diff (CI) 25.6 (14.8, 36.3) 12.8 (3.1, 22.4) N/A
    p-value <0.001a 0.009a N/A
    ACR
    70, n (%)   19 (16.5)   11 (10.5)  2 (1.7)
    Diff (CI) 14.8 (7.5, 22.2)  8.8 (2.7, 14.9) N/A
    p-value <0.001a 0.005a N/A
  • In addition, all primary efficacy analyses were performed on the WOCF (worst observation carried forward) data set. ACR responses based on the WOCF data set were slightly lower than those reported in Table 13 and were comparable to those presented in Table 21. These findings confirm the consistency of ACR response rates in the CTLA4Ig (BMS-188667) treatment groups. [0618]
  • The dosages of anti-rheumatic concomitant medications were to be collected to assess the need for these medications at 6 and 12 months; however, the available data were inadequate to perform these analyses. Only baseline values for mean dose of methotrexate and systemic (non-topical) corticosteroids are provided. [0619]
  • Efficacy Conclusions [0620]
  • CTLA4Ig (BMS-188667) administered at 10 mg/kg (+MTX) had superior efficacy compared to placebo (+MTX) at [0621] Day 180 and Day 360. For the following efficacy parameters, administration of 10 mg/kg CTLA4Ig was significantly better than placebo:
  • Primary efficacy variable: ACR20 response at Day 180 (p<0.001) [0622]
  • ACR50 and ACR70 responses at Day 180 (p<0.001) [0623]
  • ACR20, ACR50 and ACR70 responses at Day 360 (p≦0.003) [0624]
  • Statistically significant differences in ACR50 and ACR70 responses observed by Day 30 (p=0.039 and p=0.04), statistically significant differences in all 3 response rates ([0625] ACR 20, ACR50 and ACR70) observed by Day 90; these values remained statistically significant at every timepoint up to and including Day 360 (p≦0.008)
  • Proportions of subjects who achieved a Major Clinical Response (maintenance of an [0626] ACR 70 response over a continuous 6-month period) at Day 360 (p=0.008)
  • Mean numeric ACR-AUC by Day 360 (p<0.001) [0627]
  • Mean percentage improvements in each individual ACR component at [0628] Day 180 and Day 360 (p<0.05, 95% CIs did not include 0)
  • Improvements in all four mental and all four physical domains of the Health Outcomes evaluation (SF-36) at both [0629] Day 180 and Day 360 (p<0.05, 95% CIs did not include 0)
  • In addition to the above statistically significant differences, the 10 mg/kg CTLA4Ig group had a lower number of new active joints and a higher number of subjects reporting no new active tender and swollen joints compared with the placebo group at [0630] Day 180 and at Day 360.
  • Significant improvement with 10 mg/kg CTLA4Ig compared with placebo was seen in nearly all measured pharmacodynamic paramenters (soluble IL12r, RF, ICAM-1, E-selectin and IL-6) and numerical improvement in TNF-α up to 1 year. [0631]
  • For the 2 mg/kg CTLA4Ig group, some efficacy parameters were significantly better compared to the placebo group: [0632]
  • ACR50 response at Day 180 (p=0.027) [0633]
  • ACR70 response at Day 180 (p=0.005) [0634]
  • Statistically significant differences in ACR70 observed by Day 60 (p=0.032) and statistically significant differences in [0635] ACR 50 and ACR 70 at Day 180 (p=0.027 and p=0.005)
  • Proportions of subjects who achieved a Major Clinical Response (maintenance of an [0636] ACR 70 response over a continuous 6-month period) at Day 360 (p=0.036)
  • Mean percentage improvements in some of the individual ACR component at [0637] Day 180 and Day 360 (p<0.05, 95% CIs did not include 0)
  • For many other efficacy parameters, 2 mg/kg CTLA4Ig was numerically better than placebo. [0638]
  • Safety Results [0639]
  • Overall, the safety profile of CTLA4Ig (BMS-188667) was similar to placebo. There were no major safety problems. [0640]
  • Clinical Laboratory Evaluation [0641]
  • Overall, no new safety issues emerged from the evaluation of mean changes in laboratory values. Mean values for hemoglobin, WBCs, neutrophils, platelets, ALT, AST, GGT and total protein were within the normal range at baseline and remained within the normal range during the study. In general, results of the laboratory tests did not reveal consistent out-of range values or abnormal trends that could be attributed to study medication. [0642]
  • Vital Signs, Physical Findings, and Observations Related to Safety [0643]
  • On each day of study drug administration, vital signs (body temperature, heart rate, and seated blood pressure) were monitored pre-dose and at 15, 30, 45, 60, 75, 90 and 120 minutes post-infusion. Overall, mean values for all vital sign parameters were within normal range and stable throughout the 360-day study period for all treatment groups. [0644]
  • 1 22 1 65 DNA Homo sapiens 1 ctcagtctgg tccttgcact cctgtttcca agcatggcga gcatggcaat gcacgtggcc 60 cagcc 65 2 33 DNA Homo sapiens 2 tttgggctcc tgatcagaat ctgggcacgg ttg 33 3 72 DNA Homo sapiens 3 ctagccactg aagcttcacc aatgggtgta ctgctcacac agaggacgct gctcagtctg 60 gtccttgcac tc 72 4 41 DNA Artificial Oncostatin M CTLA4 (OMCTLA4) forward primer 4 gaggtgataa agcttcacca atgggtgtac tgctcacaca g 41 5 42 DNA Artificial Oncostatin M CTLA4 (OMCTLA4) reverse primer 5 gtggtgtatt ggtctagatc aatcagaatc tgggcacggt tc 42 6 1152 DNA Artificial L104EIg 6 atg ggt gta ctg ctc aca cag agg acg ctg ctc agt ctg gtc ctt gca 48 Met Gly Val Leu Leu Thr Gln Arg Thr Leu Leu Ser Leu Val Leu Ala -25 -20 -15 ctc ctg ttt cca agc atg gcg agc atg gca atg cac gtg gcc cag cct 96 Leu Leu Phe Pro Ser Met Ala Ser Met Ala Met His Val Ala Gln Pro -10 -5 -1 1 5 gct gtg gta ctg gcc agc agc cga ggc atc gct agc ttt gtg tgt gag 144 Ala Val Val Leu Ala Ser Ser Arg Gly Ile Ala Ser Phe Val Cys Glu 10 15 20 tat gca tct cca ggc aaa gcc act gag gtc cgg gtg aca gtg ctt cgg 192 Tyr Ala Ser Pro Gly Lys Ala Thr Glu Val Arg Val Thr Val Leu Arg 25 30 35 cag gct gac agc cag gtg act gaa gtc tgt gcg gca acc tac atg atg 240 Gln Ala Asp Ser Gln Val Thr Glu Val Cys Ala Ala Thr Tyr Met Met 40 45 50 ggg aat gag ttg acc ttc cta gat gat tcc atc tgc acg ggc acc tcc 288 Gly Asn Glu Leu Thr Phe Leu Asp Asp Ser Ile Cys Thr Gly Thr Ser 55 60 65 70 agt gga aat caa gtg aac ctc act atc caa gga ctg agg gcc atg gac 336 Ser Gly Asn Gln Val Asn Leu Thr Ile Gln Gly Leu Arg Ala Met Asp 75 80 85 acg gga ctc tac atc tgc aag gtg gag ctc atg tac cca ccg cca tac 384 Thr Gly Leu Tyr Ile Cys Lys Val Glu Leu Met Tyr Pro Pro Pro Tyr 90 95 100 tac gag ggc ata ggc aac gga acc cag att tat gta att gat cca gaa 432 Tyr Glu Gly Ile Gly Asn Gly Thr Gln Ile Tyr Val Ile Asp Pro Glu 105 110 115 ccg tgc cca gat tct gat cag gag ccc aaa tct tct gac aaa act cac 480 Pro Cys Pro Asp Ser Asp Gln Glu Pro Lys Ser Ser Asp Lys Thr His 120 125 130 aca tcc cca ccg tcc cca gca cct gaa ctc ctg ggg gga tcg tca gtc 528 Thr Ser Pro Pro Ser Pro Ala Pro Glu Leu Leu Gly Gly Ser Ser Val 135 140 145 150 ttc ctc ttc ccc cca aaa ccc aag gac acc ctc atg atc tcc cgg acc 576 Phe Leu Phe Pro Pro Lys Pro Lys Asp Thr Leu Met Ile Ser Arg Thr 155 160 165 cct gag gtc aca tgc gtg gtg gtg gac gtg agc cac gaa gac cct gag 624 Pro Glu Val Thr Cys Val Val Val Asp Val Ser His Glu Asp Pro Glu 170 175 180 gtc aag ttc aac tgg tac gtg gac ggc gtg gag gtg cat aat gcc aag 672 Val Lys Phe Asn Trp Tyr Val Asp Gly Val Glu Val His Asn Ala Lys 185 190 195 aca aag ccg cgg gag gag cag tac aac agc acg tac cgt gtg gtc agc 720 Thr Lys Pro Arg Glu Glu Gln Tyr Asn Ser Thr Tyr Arg Val Val Ser 200 205 210 gtc ctc acc gtc ctg cac cag gac tgg ctg aat ggc aag gag tac aag 768 Val Leu Thr Val Leu His Gln Asp Trp Leu Asn Gly Lys Glu Tyr Lys 215 220 225 230 tgc aag gtc tcc aac aaa gcc ctc cca gcc ccc atc gag aaa acc atc 816 Cys Lys Val Ser Asn Lys Ala Leu Pro Ala Pro Ile Glu Lys Thr Ile 235 240 245 tcc aaa gcc aaa ggg cag ccc cga gaa cca cag gtg tac acc ctg ccc 864 Ser Lys Ala Lys Gly Gln Pro Arg Glu Pro Gln Val Tyr Thr Leu Pro 250 255 260 cca tcc cgg gat gag ctg acc aag aac cag gtc agc ctg acc tgc ctg 912 Pro Ser Arg Asp Glu Leu Thr Lys Asn Gln Val Ser Leu Thr Cys Leu 265 270 275 gtc aaa ggc ttc tat ccc agc gac atc gcc gtg gag tgg gag agc aat 960 Val Lys Gly Phe Tyr Pro Ser Asp Ile Ala Val Glu Trp Glu Ser Asn 280 285 290 ggg cag ccg gag aac aac tac aag acc acg cct ccc gtg ctg gac tcc 1008 Gly Gln Pro Glu Asn Asn Tyr Lys Thr Thr Pro Pro Val Leu Asp Ser 295 300 305 310 gac ggc tcc ttc ttc ctc tac agc aag ctc acc gtg gac aag agc agg 1056 Asp Gly Ser Phe Phe Leu Tyr Ser Lys Leu Thr Val Asp Lys Ser Arg 315 320 325 tgg cag cag ggg aac gtc ttc tca tgc tcc gtg atg cat gag gct ctg 1104 Trp Gln Gln Gly Asn Val Phe Ser Cys Ser Val Met His Glu Ala Leu 330 335 340 cac aac cac tac acg cag aag agc ctc tcc ctg tct ccg ggt aaa tga 1152 His Asn His Tyr Thr Gln Lys Ser Leu Ser Leu Ser Pro Gly Lys 345 350 355 7 383 PRT Artificial L104EIg 7 Met Gly Val Leu Leu Thr Gln Arg Thr Leu Leu Ser Leu Val Leu Ala -25 -20 -15 Leu Leu Phe Pro Ser Met Ala Ser Met Ala Met His Val Ala Gln Pro -10 -5 -1 1 5 Ala Val Val Leu Ala Ser Ser Arg Gly Ile Ala Ser Phe Val Cys Glu 10 15 20 Tyr Ala Ser Pro Gly Lys Ala Thr Glu Val Arg Val Thr Val Leu Arg 25 30 35 Gln Ala Asp Ser Gln Val Thr Glu Val Cys Ala Ala Thr Tyr Met Met 40 45 50 Gly Asn Glu Leu Thr Phe Leu Asp Asp Ser Ile Cys Thr Gly Thr Ser 55 60 65 70 Ser Gly Asn Gln Val Asn Leu Thr Ile Gln Gly Leu Arg Ala Met Asp 75 80 85 Thr Gly Leu Tyr Ile Cys Lys Val Glu Leu Met Tyr Pro Pro Pro Tyr 90 95 100 Tyr Glu Gly Ile Gly Asn Gly Thr Gln Ile Tyr Val Ile Asp Pro Glu 105 110 115 Pro Cys Pro Asp Ser Asp Gln Glu Pro Lys Ser Ser Asp Lys Thr His 120 125 130 Thr Ser Pro Pro Ser Pro Ala Pro Glu Leu Leu Gly Gly Ser Ser Val 135 140 145 150 Phe Leu Phe Pro Pro Lys Pro Lys Asp Thr Leu Met Ile Ser Arg Thr 155 160 165 Pro Glu Val Thr Cys Val Val Val Asp Val Ser His Glu Asp Pro Glu 170 175 180 Val Lys Phe Asn Trp Tyr Val Asp Gly Val Glu Val His Asn Ala Lys 185 190 195 Thr Lys Pro Arg Glu Glu Gln Tyr Asn Ser Thr Tyr Arg Val Val Ser 200 205 210 Val Leu Thr Val Leu His Gln Asp Trp Leu Asn Gly Lys Glu Tyr Lys 215 220 225 230 Cys Lys Val Ser Asn Lys Ala Leu Pro Ala Pro Ile Glu Lys Thr Ile 235 240 245 Ser Lys Ala Lys Gly Gln Pro Arg Glu Pro Gln Val Tyr Thr Leu Pro 250 255 260 Pro Ser Arg Asp Glu Leu Thr Lys Asn Gln Val Ser Leu Thr Cys Leu 265 270 275 Val Lys Gly Phe Tyr Pro Ser Asp Ile Ala Val Glu Trp Glu Ser Asn 280 285 290 Gly Gln Pro Glu Asn Asn Tyr Lys Thr Thr Pro Pro Val Leu Asp Ser 295 300 305 310 Asp Gly Ser Phe Phe Leu Tyr Ser Lys Leu Thr Val Asp Lys Ser Arg 315 320 325 Trp Gln Gln Gly Asn Val Phe Ser Cys Ser Val Met His Glu Ala Leu 330 335 340 His Asn His Tyr Thr Gln Lys Ser Leu Ser Leu Ser Pro Gly Lys 345 350 355 8 1152 DNA Artificial L104EA29YIg 8 atg ggt gta ctg ctc aca cag agg acg ctg ctc agt ctg gtc ctt gca 48 Met Gly Val Leu Leu Thr Gln Arg Thr Leu Leu Ser Leu Val Leu Ala -25 -20 -15 ctc ctg ttt cca agc atg gcg agc atg gca atg cac gtg gcc cag cct 96 Leu Leu Phe Pro Ser Met Ala Ser Met Ala Met His Val Ala Gln Pro -10 -5 -1 1 5 gct gtg gta ctg gcc agc agc cga ggc atc gct agc ttt gtg tgt gag 144 Ala Val Val Leu Ala Ser Ser Arg Gly Ile Ala Ser Phe Val Cys Glu 10 15 20 tat gca tct cca ggc aaa tat act gag gtc cgg gtg aca gtg ctt cgg 192 Tyr Ala Ser Pro Gly Lys Tyr Thr Glu Val Arg Val Thr Val Leu Arg 25 30 35 cag gct gac agc cag gtg act gaa gtc tgt gcg gca acc tac atg atg 240 Gln Ala Asp Ser Gln Val Thr Glu Val Cys Ala Ala Thr Tyr Met Met 40 45 50 ggg aat gag ttg acc ttc cta gat gat tcc atc tgc acg ggc acc tcc 288 Gly Asn Glu Leu Thr Phe Leu Asp Asp Ser Ile Cys Thr Gly Thr Ser 55 60 65 70 agt gga aat caa gtg aac ctc act atc caa gga ctg agg gcc atg gac 336 Ser Gly Asn Gln Val Asn Leu Thr Ile Gln Gly Leu Arg Ala Met Asp 75 80 85 acg gga ctc tac atc tgc aag gtg gag ctc atg tac cca ccg cca tac 384 Thr Gly Leu Tyr Ile Cys Lys Val Glu Leu Met Tyr Pro Pro Pro Tyr 90 95 100 tac gag ggc ata ggc aac gga acc cag att tat gta att gat cca gaa 432 Tyr Glu Gly Ile Gly Asn Gly Thr Gln Ile Tyr Val Ile Asp Pro Glu 105 110 115 ccg tgc cca gat tct gat cag gag ccc aaa tct tct gac aaa act cac 480 Pro Cys Pro Asp Ser Asp Gln Glu Pro Lys Ser Ser Asp Lys Thr His 120 125 130 aca tcc cca ccg tcc cca gca cct gaa ctc ctg ggg gga tcg tca gtc 528 Thr Ser Pro Pro Ser Pro Ala Pro Glu Leu Leu Gly Gly Ser Ser Val 135 140 145 150 ttc ctc ttc ccc cca aaa ccc aag gac acc ctc atg atc tcc cgg acc 576 Phe Leu Phe Pro Pro Lys Pro Lys Asp Thr Leu Met Ile Ser Arg Thr 155 160 165 cct gag gtc aca tgc gtg gtg gtg gac gtg agc cac gaa gac cct gag 624 Pro Glu Val Thr Cys Val Val Val Asp Val Ser His Glu Asp Pro Glu 170 175 180 gtc aag ttc aac tgg tac gtg gac ggc gtg gag gtg cat aat gcc aag 672 Val Lys Phe Asn Trp Tyr Val Asp Gly Val Glu Val His Asn Ala Lys 185 190 195 aca aag ccg cgg gag gag cag tac aac agc acg tac cgt gtg gtc agc 720 Thr Lys Pro Arg Glu Glu Gln Tyr Asn Ser Thr Tyr Arg Val Val Ser 200 205 210 gtc ctc acc gtc ctg cac cag gac tgg ctg aat ggc aag gag tac aag 768 Val Leu Thr Val Leu His Gln Asp Trp Leu Asn Gly Lys Glu Tyr Lys 215 220 225 230 tgc aag gtc tcc aac aaa gcc ctc cca gcc ccc atc gag aaa acc atc 816 Cys Lys Val Ser Asn Lys Ala Leu Pro Ala Pro Ile Glu Lys Thr Ile 235 240 245 tcc aaa gcc aaa ggg cag ccc cga gaa cca cag gtg tac acc ctg ccc 864 Ser Lys Ala Lys Gly Gln Pro Arg Glu Pro Gln Val Tyr Thr Leu Pro 250 255 260 cca tcc cgg gat gag ctg acc aag aac cag gtc agc ctg acc tgc ctg 912 Pro Ser Arg Asp Glu Leu Thr Lys Asn Gln Val Ser Leu Thr Cys Leu 265 270 275 gtc aaa ggc ttc tat ccc agc gac atc gcc gtg gag tgg gag agc aat 960 Val Lys Gly Phe Tyr Pro Ser Asp Ile Ala Val Glu Trp Glu Ser Asn 280 285 290 ggg cag ccg gag aac aac tac aag acc acg cct ccc gtg ctg gac tcc 1008 Gly Gln Pro Glu Asn Asn Tyr Lys Thr Thr Pro Pro Val Leu Asp Ser 295 300 305 310 gac ggc tcc ttc ttc ctc tac agc aag ctc acc gtg gac aag agc agg 1056 Asp Gly Ser Phe Phe Leu Tyr Ser Lys Leu Thr Val Asp Lys Ser Arg 315 320 325 tgg cag cag ggg aac gtc ttc tca tgc tcc gtg atg cat gag gct ctg 1104 Trp Gln Gln Gly Asn Val Phe Ser Cys Ser Val Met His Glu Ala Leu 330 335 340 cac aac cac tac acg cag aag agc ctc tcc ctg tct ccg ggt aaa tga 1152 His Asn His Tyr Thr Gln Lys Ser Leu Ser Leu Ser Pro Gly Lys 345 350 355 9 383 PRT Artificial L104EA29YIg 9 Met Gly Val Leu Leu Thr Gln Arg Thr Leu Leu Ser Leu Val Leu Ala -25 -20 -15 Leu Leu Phe Pro Ser Met Ala Ser Met Ala Met His Val Ala Gln Pro -10 -5 -1 1 5 Ala Val Val Leu Ala Ser Ser Arg Gly Ile Ala Ser Phe Val Cys Glu 10 15 20 Tyr Ala Ser Pro Gly Lys Tyr Thr Glu Val Arg Val Thr Val Leu Arg 25 30 35 Gln Ala Asp Ser Gln Val Thr Glu Val Cys Ala Ala Thr Tyr Met Met 40 45 50 Gly Asn Glu Leu Thr Phe Leu Asp Asp Ser Ile Cys Thr Gly Thr Ser 55 60 65 70 Ser Gly Asn Gln Val Asn Leu Thr Ile Gln Gly Leu Arg Ala Met Asp 75 80 85 Thr Gly Leu Tyr Ile Cys Lys Val Glu Leu Met Tyr Pro Pro Pro Tyr 90 95 100 Tyr Glu Gly Ile Gly Asn Gly Thr Gln Ile Tyr Val Ile Asp Pro Glu 105 110 115 Pro Cys Pro Asp Ser Asp Gln Glu Pro Lys Ser Ser Asp Lys Thr His 120 125 130 Thr Ser Pro Pro Ser Pro Ala Pro Glu Leu Leu Gly Gly Ser Ser Val 135 140 145 150 Phe Leu Phe Pro Pro Lys Pro Lys Asp Thr Leu Met Ile Ser Arg Thr 155 160 165 Pro Glu Val Thr Cys Val Val Val Asp Val Ser His Glu Asp Pro Glu 170 175 180 Val Lys Phe Asn Trp Tyr Val Asp Gly Val Glu Val His Asn Ala Lys 185 190 195 Thr Lys Pro Arg Glu Glu Gln Tyr Asn Ser Thr Tyr Arg Val Val Ser 200 205 210 Val Leu Thr Val Leu His Gln Asp Trp Leu Asn Gly Lys Glu Tyr Lys 215 220 225 230 Cys Lys Val Ser Asn Lys Ala Leu Pro Ala Pro Ile Glu Lys Thr Ile 235 240 245 Ser Lys Ala Lys Gly Gln Pro Arg Glu Pro Gln Val Tyr Thr Leu Pro 250 255 260 Pro Ser Arg Asp Glu Leu Thr Lys Asn Gln Val Ser Leu Thr Cys Leu 265 270 275 Val Lys Gly Phe Tyr Pro Ser Asp Ile Ala Val Glu Trp Glu Ser Asn 280 285 290 Gly Gln Pro Glu Asn Asn Tyr Lys Thr Thr Pro Pro Val Leu Asp Ser 295 300 305 310 Asp Gly Ser Phe Phe Leu Tyr Ser Lys Leu Thr Val Asp Lys Ser Arg 315 320 325 Trp Gln Gln Gly Asn Val Phe Ser Cys Ser Val Met His Glu Ala Leu 330 335 340 His Asn His Tyr Thr Gln Lys Ser Leu Ser Leu Ser Pro Gly Lys 345 350 355 10 1152 DNA Artificial L104EA29LIg 10 atg ggt gta ctg ctc aca cag agg acg ctg ctc agt ctg gtc ctt gca 48 Met Gly Val Leu Leu Thr Gln Arg Thr Leu Leu Ser Leu Val Leu Ala -25 -20 -15 ctc ctg ttt cca agc atg gcg agc atg gca atg cac gtg gcc cag cct 96 Leu Leu Phe Pro Ser Met Ala Ser Met Ala Met His Val Ala Gln Pro -10 -5 -1 1 5 gct gtg gta ctg gcc agc agc cga ggc atc gct agc ttt gtg tgt gag 144 Ala Val Val Leu Ala Ser Ser Arg Gly Ile Ala Ser Phe Val Cys Glu 10 15 20 tat gca tct cca ggc aaa ttg act gag gtc cgg gtg aca gtg ctt cgg 192 Tyr Ala Ser Pro Gly Lys Leu Thr Glu Val Arg Val Thr Val Leu Arg 25 30 35 cag gct gac agc cag gtg act gaa gtc tgt gcg gca acc tac atg atg 240 Gln Ala Asp Ser Gln Val Thr Glu Val Cys Ala Ala Thr Tyr Met Met 40 45 50 ggg aat gag ttg acc ttc cta gat gat tcc atc tgc acg ggc acc tcc 288 Gly Asn Glu Leu Thr Phe Leu Asp Asp Ser Ile Cys Thr Gly Thr Ser 55 60 65 70 agt gga aat caa gtg aac ctc act atc caa gga ctg agg gcc atg gac 336 Ser Gly Asn Gln Val Asn Leu Thr Ile Gln Gly Leu Arg Ala Met Asp 75 80 85 acg gga ctc tac atc tgc aag gtg gag ctc atg tac cca ccg cca tac 384 Thr Gly Leu Tyr Ile Cys Lys Val Glu Leu Met Tyr Pro Pro Pro Tyr 90 95 100 tac gag ggc ata ggc aac gga acc cag att tat gta att gat cca gaa 432 Tyr Glu Gly Ile Gly Asn Gly Thr Gln Ile Tyr Val Ile Asp Pro Glu 105 110 115 ccg tgc cca gat tct gat cag gag ccc aaa tct tct gac aaa act cac 480 Pro Cys Pro Asp Ser Asp Gln Glu Pro Lys Ser Ser Asp Lys Thr His 120 125 130 aca tcc cca ccg tcc cca gca cct gaa ctc ctg ggg gga tcg tca gtc 528 Thr Ser Pro Pro Ser Pro Ala Pro Glu Leu Leu Gly Gly Ser Ser Val 135 140 145 150 ttc ctc ttc ccc cca aaa ccc aag gac acc ctc atg atc tcc cgg acc 576 Phe Leu Phe Pro Pro Lys Pro Lys Asp Thr Leu Met Ile Ser Arg Thr 155 160 165 cct gag gtc aca tgc gtg gtg gtg gac gtg agc cac gaa gac cct gag 624 Pro Glu Val Thr Cys Val Val Val Asp Val Ser His Glu Asp Pro Glu 170 175 180 gtc aag ttc aac tgg tac gtg gac ggc gtg gag gtg cat aat gcc aag 672 Val Lys Phe Asn Trp Tyr Val Asp Gly Val Glu Val His Asn Ala Lys 185 190 195 aca aag ccg cgg gag gag cag tac aac agc acg tac cgt gtg gtc agc 720 Thr Lys Pro Arg Glu Glu Gln Tyr Asn Ser Thr Tyr Arg Val Val Ser 200 205 210 gtc ctc acc gtc ctg cac cag gac tgg ctg aat ggc aag gag tac aag 768 Val Leu Thr Val Leu His Gln Asp Trp Leu Asn Gly Lys Glu Tyr Lys 215 220 225 230 tgc aag gtc tcc aac aaa gcc ctc cca gcc ccc atc gag aaa acc atc 816 Cys Lys Val Ser Asn Lys Ala Leu Pro Ala Pro Ile Glu Lys Thr Ile 235 240 245 tcc aaa gcc aaa ggg cag ccc cga gaa cca cag gtg tac acc ctg ccc 864 Ser Lys Ala Lys Gly Gln Pro Arg Glu Pro Gln Val Tyr Thr Leu Pro 250 255 260 cca tcc cgg gat gag ctg acc aag aac cag gtc agc ctg acc tgc ctg 912 Pro Ser Arg Asp Glu Leu Thr Lys Asn Gln Val Ser Leu Thr Cys Leu 265 270 275 gtc aaa ggc ttc tat ccc agc gac atc gcc gtg gag tgg gag agc aat 960 Val Lys Gly Phe Tyr Pro Ser Asp Ile Ala Val Glu Trp Glu Ser Asn 280 285 290 ggg cag ccg gag aac aac tac aag acc acg cct ccc gtg ctg gac tcc 1008 Gly Gln Pro Glu Asn Asn Tyr Lys Thr Thr Pro Pro Val Leu Asp Ser 295 300 305 310 gac ggc tcc ttc ttc ctc tac agc aag ctc acc gtg gac aag agc agg 1056 Asp Gly Ser Phe Phe Leu Tyr Ser Lys Leu Thr Val Asp Lys Ser Arg 315 320 325 tgg cag cag ggg aac gtc ttc tca tgc tcc gtg atg cat gag gct ctg 1104 Trp Gln Gln Gly Asn Val Phe Ser Cys Ser Val Met His Glu Ala Leu 330 335 340 cac aac cac tac acg cag aag agc ctc tcc ctg tct ccg ggt aaa tga 1152 His Asn His Tyr Thr Gln Lys Ser Leu Ser Leu Ser Pro Gly Lys 345 350 355 11 383 PRT Artificial L104EA29LIg 11 Met Gly Val Leu Leu Thr Gln Arg Thr Leu Leu Ser Leu Val Leu Ala -25 -20 -15 Leu Leu Phe Pro Ser Met Ala Ser Met Ala Met His Val Ala Gln Pro -10 -5 -1 1 5 Ala Val Val Leu Ala Ser Ser Arg Gly Ile Ala Ser Phe Val Cys Glu 10 15 20 Tyr Ala Ser Pro Gly Lys Leu Thr Glu Val Arg Val Thr Val Leu Arg 25 30 35 Gln Ala Asp Ser Gln Val Thr Glu Val Cys Ala Ala Thr Tyr Met Met 40 45 50 Gly Asn Glu Leu Thr Phe Leu Asp Asp Ser Ile Cys Thr Gly Thr Ser 55 60 65 70 Ser Gly Asn Gln Val Asn Leu Thr Ile Gln Gly Leu Arg Ala Met Asp 75 80 85 Thr Gly Leu Tyr Ile Cys Lys Val Glu Leu Met Tyr Pro Pro Pro Tyr 90 95 100 Tyr Glu Gly Ile Gly Asn Gly Thr Gln Ile Tyr Val Ile Asp Pro Glu 105 110 115 Pro Cys Pro Asp Ser Asp Gln Glu Pro Lys Ser Ser Asp Lys Thr His 120 125 130 Thr Ser Pro Pro Ser Pro Ala Pro Glu Leu Leu Gly Gly Ser Ser Val 135 140 145 150 Phe Leu Phe Pro Pro Lys Pro Lys Asp Thr Leu Met Ile Ser Arg Thr 155 160 165 Pro Glu Val Thr Cys Val Val Val Asp Val Ser His Glu Asp Pro Glu 170 175 180 Val Lys Phe Asn Trp Tyr Val Asp Gly Val Glu Val His Asn Ala Lys 185 190 195 Thr Lys Pro Arg Glu Glu Gln Tyr Asn Ser Thr Tyr Arg Val Val Ser 200 205 210 Val Leu Thr Val Leu His Gln Asp Trp Leu Asn Gly Lys Glu Tyr Lys 215 220 225 230 Cys Lys Val Ser Asn Lys Ala Leu Pro Ala Pro Ile Glu Lys Thr Ile 235 240 245 Ser Lys Ala Lys Gly Gln Pro Arg Glu Pro Gln Val Tyr Thr Leu Pro 250 255 260 Pro Ser Arg Asp Glu Leu Thr Lys Asn Gln Val Ser Leu Thr Cys Leu 265 270 275 Val Lys Gly Phe Tyr Pro Ser Asp Ile Ala Val Glu Trp Glu Ser Asn 280 285 290 Gly Gln Pro Glu Asn Asn Tyr Lys Thr Thr Pro Pro Val Leu Asp Ser 295 300 305 310 Asp Gly Ser Phe Phe Leu Tyr Ser Lys Leu Thr Val Asp Lys Ser Arg 315 320 325 Trp Gln Gln Gly Asn Val Phe Ser Cys Ser Val Met His Glu Ala Leu 330 335 340 His Asn His Tyr Thr Gln Lys Ser Leu Ser Leu Ser Pro Gly Lys 345 350 355 12 1152 DNA Artificial L104EA29TIg 12 atg ggt gta ctg ctc aca cag agg acg ctg ctc agt ctg gtc ctt gca 48 Met Gly Val Leu Leu Thr Gln Arg Thr Leu Leu Ser Leu Val Leu Ala -25 -20 -15 ctc ctg ttt cca agc atg gcg agc atg gca atg cac gtg gcc cag cct 96 Leu Leu Phe Pro Ser Met Ala Ser Met Ala Met His Val Ala Gln Pro -10 -5 -1 1 5 gct gtg gta ctg gcc agc agc cga ggc atc gct agc ttt gtg tgt gag 144 Ala Val Val Leu Ala Ser Ser Arg Gly Ile Ala Ser Phe Val Cys Glu 10 15 20 tat gca tct cca ggc aaa act act gag gtc cgg gtg aca gtg ctt cgg 192 Tyr Ala Ser Pro Gly Lys Thr Thr Glu Val Arg Val Thr Val Leu Arg 25 30 35 cag gct gac agc cag gtg act gaa gtc tgt gcg gca acc tac atg atg 240 Gln Ala Asp Ser Gln Val Thr Glu Val Cys Ala Ala Thr Tyr Met Met 40 45 50 ggg aat gag ttg acc ttc cta gat gat tcc atc tgc acg ggc acc tcc 288 Gly Asn Glu Leu Thr Phe Leu Asp Asp Ser Ile Cys Thr Gly Thr Ser 55 60 65 70 agt gga aat caa gtg aac ctc act atc caa gga ctg agg gcc atg gac 336 Ser Gly Asn Gln Val Asn Leu Thr Ile Gln Gly Leu Arg Ala Met Asp 75 80 85 acg gga ctc tac atc tgc aag gtg gag ctc atg tac cca ccg cca tac 384 Thr Gly Leu Tyr Ile Cys Lys Val Glu Leu Met Tyr Pro Pro Pro Tyr 90 95 100 tac gag ggc ata ggc aac gga acc cag att tat gta att gat cca gaa 432 Tyr Glu Gly Ile Gly Asn Gly Thr Gln Ile Tyr Val Ile Asp Pro Glu 105 110 115 ccg tgc cca gat tct gat cag gag ccc aaa tct tct gac aaa act cac 480 Pro Cys Pro Asp Ser Asp Gln Glu Pro Lys Ser Ser Asp Lys Thr His 120 125 130 aca tcc cca ccg tcc cca gca cct gaa ctc ctg ggg gga tcg tca gtc 528 Thr Ser Pro Pro Ser Pro Ala Pro Glu Leu Leu Gly Gly Ser Ser Val 135 140 145 150 ttc ctc ttc ccc cca aaa ccc aag gac acc ctc atg atc tcc cgg acc 576 Phe Leu Phe Pro Pro Lys Pro Lys Asp Thr Leu Met Ile Ser Arg Thr 155 160 165 cct gag gtc aca tgc gtg gtg gtg gac gtg agc cac gaa gac cct gag 624 Pro Glu Val Thr Cys Val Val Val Asp Val Ser His Glu Asp Pro Glu 170 175 180 gtc aag ttc aac tgg tac gtg gac ggc gtg gag gtg cat aat gcc aag 672 Val Lys Phe Asn Trp Tyr Val Asp Gly Val Glu Val His Asn Ala Lys 185 190 195 aca aag ccg cgg gag gag cag tac aac agc acg tac cgt gtg gtc agc 720 Thr Lys Pro Arg Glu Glu Gln Tyr Asn Ser Thr Tyr Arg Val Val Ser 200 205 210 gtc ctc acc gtc ctg cac cag gac tgg ctg aat ggc aag gag tac aag 768 Val Leu Thr Val Leu His Gln Asp Trp Leu Asn Gly Lys Glu Tyr Lys 215 220 225 230 tgc aag gtc tcc aac aaa gcc ctc cca gcc ccc atc gag aaa acc atc 816 Cys Lys Val Ser Asn Lys Ala Leu Pro Ala Pro Ile Glu Lys Thr Ile 235 240 245 tcc aaa gcc aaa ggg cag ccc cga gaa cca cag gtg tac acc ctg ccc 864 Ser Lys Ala Lys Gly Gln Pro Arg Glu Pro Gln Val Tyr Thr Leu Pro 250 255 260 cca tcc cgg gat gag ctg acc aag aac cag gtc agc ctg acc tgc ctg 912 Pro Ser Arg Asp Glu Leu Thr Lys Asn Gln Val Ser Leu Thr Cys Leu 265 270 275 gtc aaa ggc ttc tat ccc agc gac atc gcc gtg gag tgg gag agc aat 960 Val Lys Gly Phe Tyr Pro Ser Asp Ile Ala Val Glu Trp Glu Ser Asn 280 285 290 ggg cag ccg gag aac aac tac aag acc acg cct ccc gtg ctg gac tcc 1008 Gly Gln Pro Glu Asn Asn Tyr Lys Thr Thr Pro Pro Val Leu Asp Ser 295 300 305 310 gac ggc tcc ttc ttc ctc tac agc aag ctc acc gtg gac aag agc agg 1056 Asp Gly Ser Phe Phe Leu Tyr Ser Lys Leu Thr Val Asp Lys Ser Arg 315 320 325 tgg cag cag ggg aac gtc ttc tca tgc tcc gtg atg cat gag gct ctg 1104 Trp Gln Gln Gly Asn Val Phe Ser Cys Ser Val Met His Glu Ala Leu 330 335 340 cac aac cac tac acg cag aag agc ctc tcc ctg tct ccg ggt aaa tga 1152 His Asn His Tyr Thr Gln Lys Ser Leu Ser Leu Ser Pro Gly Lys 345 350 355 13 383 PRT Artificial L104EA29TIg 13 Met Gly Val Leu Leu Thr Gln Arg Thr Leu Leu Ser Leu Val Leu Ala -25 -20 -15 Leu Leu Phe Pro Ser Met Ala Ser Met Ala Met His Val Ala Gln Pro -10 -5 -1 1 5 Ala Val Val Leu Ala Ser Ser Arg Gly Ile Ala Ser Phe Val Cys Glu 10 15 20 Tyr Ala Ser Pro Gly Lys Thr Thr Glu Val Arg Val Thr Val Leu Arg 25 30 35 Gln Ala Asp Ser Gln Val Thr Glu Val Cys Ala Ala Thr Tyr Met Met 40 45 50 Gly Asn Glu Leu Thr Phe Leu Asp Asp Ser Ile Cys Thr Gly Thr Ser 55 60 65 70 Ser Gly Asn Gln Val Asn Leu Thr Ile Gln Gly Leu Arg Ala Met Asp 75 80 85 Thr Gly Leu Tyr Ile Cys Lys Val Glu Leu Met Tyr Pro Pro Pro Tyr 90 95 100 Tyr Glu Gly Ile Gly Asn Gly Thr Gln Ile Tyr Val Ile Asp Pro Glu 105 110 115 Pro Cys Pro Asp Ser Asp Gln Glu Pro Lys Ser Ser Asp Lys Thr His 120 125 130 Thr Ser Pro Pro Ser Pro Ala Pro Glu Leu Leu Gly Gly Ser Ser Val 135 140 145 150 Phe Leu Phe Pro Pro Lys Pro Lys Asp Thr Leu Met Ile Ser Arg Thr 155 160 165 Pro Glu Val Thr Cys Val Val Val Asp Val Ser His Glu Asp Pro Glu 170 175 180 Val Lys Phe Asn Trp Tyr Val Asp Gly Val Glu Val His Asn Ala Lys 185 190 195 Thr Lys Pro Arg Glu Glu Gln Tyr Asn Ser Thr Tyr Arg Val Val Ser 200 205 210 Val Leu Thr Val Leu His Gln Asp Trp Leu Asn Gly Lys Glu Tyr Lys 215 220 225 230 Cys Lys Val Ser Asn Lys Ala Leu Pro Ala Pro Ile Glu Lys Thr Ile 235 240 245 Ser Lys Ala Lys Gly Gln Pro Arg Glu Pro Gln Val Tyr Thr Leu Pro 250 255 260 Pro Ser Arg Asp Glu Leu Thr Lys Asn Gln Val Ser Leu Thr Cys Leu 265 270 275 Val Lys Gly Phe Tyr Pro Ser Asp Ile Ala Val Glu Trp Glu Ser Asn 280 285 290 Gly Gln Pro Glu Asn Asn Tyr Lys Thr Thr Pro Pro Val Leu Asp Ser 295 300 305 310 Asp Gly Ser Phe Phe Leu Tyr Ser Lys Leu Thr Val Asp Lys Ser Arg 315 320 325 Trp Gln Gln Gly Asn Val Phe Ser Cys Ser Val Met His Glu Ala Leu 330 335 340 His Asn His Tyr Thr Gln Lys Ser Leu Ser Leu Ser Pro Gly Lys 345 350 355 14 1152 DNA Artificial L104EA29WIg 14 atg ggt gta ctg ctc aca cag agg acg ctg ctc agt ctg gtc ctt gca 48 Met Gly Val Leu Leu Thr Gln Arg Thr Leu Leu Ser Leu Val Leu Ala -25 -20 -15 ctc ctg ttt cca agc atg gcg agc atg gca atg cac gtg gcc cag cct 96 Leu Leu Phe Pro Ser Met Ala Ser Met Ala Met His Val Ala Gln Pro -10 -5 -1 1 5 gct gtg gta ctg gcc agc agc cga ggc atc gct agc ttt gtg tgt gag 144 Ala Val Val Leu Ala Ser Ser Arg Gly Ile Ala Ser Phe Val Cys Glu 10 15 20 tat gca tct cca ggc aaa tgg act gag gtc cgg gtg aca gtg ctt cgg 192 Tyr Ala Ser Pro Gly Lys Trp Thr Glu Val Arg Val Thr Val Leu Arg 25 30 35 cag gct gac agc cag gtg act gaa gtc tgt gcg gca acc tac atg atg 240 Gln Ala Asp Ser Gln Val Thr Glu Val Cys Ala Ala Thr Tyr Met Met 40 45 50 ggg aat gag ttg acc ttc cta gat gat tcc atc tgc acg ggc acc tcc 288 Gly Asn Glu Leu Thr Phe Leu Asp Asp Ser Ile Cys Thr Gly Thr Ser 55 60 65 70 agt gga aat caa gtg aac ctc act atc caa gga ctg agg gcc atg gac 336 Ser Gly Asn Gln Val Asn Leu Thr Ile Gln Gly Leu Arg Ala Met Asp 75 80 85 acg gga ctc tac atc tgc aag gtg gag ctc atg tac cca ccg cca tac 384 Thr Gly Leu Tyr Ile Cys Lys Val Glu Leu Met Tyr Pro Pro Pro Tyr 90 95 100 tac gag ggc ata ggc aac gga acc cag att tat gta att gat cca gaa 432 Tyr Glu Gly Ile Gly Asn Gly Thr Gln Ile Tyr Val Ile Asp Pro Glu 105 110 115 ccg tgc cca gat tct gat cag gag ccc aaa tct tct gac aaa act cac 480 Pro Cys Pro Asp Ser Asp Gln Glu Pro Lys Ser Ser Asp Lys Thr His 120 125 130 aca tcc cca ccg tcc cca gca cct gaa ctc ctg ggg gga tcg tca gtc 528 Thr Ser Pro Pro Ser Pro Ala Pro Glu Leu Leu Gly Gly Ser Ser Val 135 140 145 150 ttc ctc ttc ccc cca aaa ccc aag gac acc ctc atg atc tcc cgg acc 576 Phe Leu Phe Pro Pro Lys Pro Lys Asp Thr Leu Met Ile Ser Arg Thr 155 160 165 cct gag gtc aca tgc gtg gtg gtg gac gtg agc cac gaa gac cct gag 624 Pro Glu Val Thr Cys Val Val Val Asp Val Ser His Glu Asp Pro Glu 170 175 180 gtc aag ttc aac tgg tac gtg gac ggc gtg gag gtg cat aat gcc aag 672 Val Lys Phe Asn Trp Tyr Val Asp Gly Val Glu Val His Asn Ala Lys 185 190 195 aca aag ccg cgg gag gag cag tac aac agc acg tac cgt gtg gtc agc 720 Thr Lys Pro Arg Glu Glu Gln Tyr Asn Ser Thr Tyr Arg Val Val Ser 200 205 210 gtc ctc acc gtc ctg cac cag gac tgg ctg aat ggc aag gag tac aag 768 Val Leu Thr Val Leu His Gln Asp Trp Leu Asn Gly Lys Glu Tyr Lys 215 220 225 230 tgc aag gtc tcc aac aaa gcc ctc cca gcc ccc atc gag aaa acc atc 816 Cys Lys Val Ser Asn Lys Ala Leu Pro Ala Pro Ile Glu Lys Thr Ile 235 240 245 tcc aaa gcc aaa ggg cag ccc cga gaa cca cag gtg tac acc ctg ccc 864 Ser Lys Ala Lys Gly Gln Pro Arg Glu Pro Gln Val Tyr Thr Leu Pro 250 255 260 cca tcc cgg gat gag ctg acc aag aac cag gtc agc ctg acc tgc ctg 912 Pro Ser Arg Asp Glu Leu Thr Lys Asn Gln Val Ser Leu Thr Cys Leu 265 270 275 gtc aaa ggc ttc tat ccc agc gac atc gcc gtg gag tgg gag agc aat 960 Val Lys Gly Phe Tyr Pro Ser Asp Ile Ala Val Glu Trp Glu Ser Asn 280 285 290 ggg cag ccg gag aac aac tac aag acc acg cct ccc gtg ctg gac tcc 1008 Gly Gln Pro Glu Asn Asn Tyr Lys Thr Thr Pro Pro Val Leu Asp Ser 295 300 305 310 gac ggc tcc ttc ttc ctc tac agc aag ctc acc gtg gac aag agc agg 1056 Asp Gly Ser Phe Phe Leu Tyr Ser Lys Leu Thr Val Asp Lys Ser Arg 315 320 325 tgg cag cag ggg aac gtc ttc tca tgc tcc gtg atg cat gag gct ctg 1104 Trp Gln Gln Gly Asn Val Phe Ser Cys Ser Val Met His Glu Ala Leu 330 335 340 cac aac cac tac acg cag aag agc ctc tcc ctg tct ccg ggt aaa tga 1152 His Asn His Tyr Thr Gln Lys Ser Leu Ser Leu Ser Pro Gly Lys 345 350 355 15 383 PRT Artificial L104EA29WIg 15 Met Gly Val Leu Leu Thr Gln Arg Thr Leu Leu Ser Leu Val Leu Ala -25 -20 -15 Leu Leu Phe Pro Ser Met Ala Ser Met Ala Met His Val Ala Gln Pro -10 -5 -1 1 5 Ala Val Val Leu Ala Ser Ser Arg Gly Ile Ala Ser Phe Val Cys Glu 10 15 20 Tyr Ala Ser Pro Gly Lys Trp Thr Glu Val Arg Val Thr Val Leu Arg 25 30 35 Gln Ala Asp Ser Gln Val Thr Glu Val Cys Ala Ala Thr Tyr Met Met 40 45 50 Gly Asn Glu Leu Thr Phe Leu Asp Asp Ser Ile Cys Thr Gly Thr Ser 55 60 65 70 Ser Gly Asn Gln Val Asn Leu Thr Ile Gln Gly Leu Arg Ala Met Asp 75 80 85 Thr Gly Leu Tyr Ile Cys Lys Val Glu Leu Met Tyr Pro Pro Pro Tyr 90 95 100 Tyr Glu Gly Ile Gly Asn Gly Thr Gln Ile Tyr Val Ile Asp Pro Glu 105 110 115 Pro Cys Pro Asp Ser Asp Gln Glu Pro Lys Ser Ser Asp Lys Thr His 120 125 130 Thr Ser Pro Pro Ser Pro Ala Pro Glu Leu Leu Gly Gly Ser Ser Val 135 140 145 150 Phe Leu Phe Pro Pro Lys Pro Lys Asp Thr Leu Met Ile Ser Arg Thr 155 160 165 Pro Glu Val Thr Cys Val Val Val Asp Val Ser His Glu Asp Pro Glu 170 175 180 Val Lys Phe Asn Trp Tyr Val Asp Gly Val Glu Val His Asn Ala Lys 185 190 195 Thr Lys Pro Arg Glu Glu Gln Tyr Asn Ser Thr Tyr Arg Val Val Ser 200 205 210 Val Leu Thr Val Leu His Gln Asp Trp Leu Asn Gly Lys Glu Tyr Lys 215 220 225 230 Cys Lys Val Ser Asn Lys Ala Leu Pro Ala Pro Ile Glu Lys Thr Ile 235 240 245 Ser Lys Ala Lys Gly Gln Pro Arg Glu Pro Gln Val Tyr Thr Leu Pro 250 255 260 Pro Ser Arg Asp Glu Leu Thr Lys Asn Gln Val Ser Leu Thr Cys Leu 265 270 275 Val Lys Gly Phe Tyr Pro Ser Asp Ile Ala Val Glu Trp Glu Ser Asn 280 285 290 Gly Gln Pro Glu Asn Asn Tyr Lys Thr Thr Pro Pro Val Leu Asp Ser 295 300 305 310 Asp Gly Ser Phe Phe Leu Tyr Ser Lys Leu Thr Val Asp Lys Ser Arg 315 320 325 Trp Gln Gln Gly Asn Val Phe Ser Cys Ser Val Met His Glu Ala Leu 330 335 340 His Asn His Tyr Thr Gln Lys Ser Leu Ser Leu Ser Pro Gly Lys 345 350 355 16 636 DNA Homo sapiens CDS (1)..(636) mat_peptide (79)..() 16 atg ggt gta ctg ctc aca cag agg acg ctg ctc agt ctg gtc ctt gca 48 Met Gly Val Leu Leu Thr Gln Arg Thr Leu Leu Ser Leu Val Leu Ala -25 -20 -15 ctc ctg ttt cca agc atg gcg agc atg gca atg cac gtg gcc cag cct 96 Leu Leu Phe Pro Ser Met Ala Ser Met Ala Met His Val Ala Gln Pro -10 -5 -1 1 5 gct gtg gta ctg gcc agc agc cga ggc atc gcc agc ttt gtg tgt gag 144 Ala Val Val Leu Ala Ser Ser Arg Gly Ile Ala Ser Phe Val Cys Glu 10 15 20 tat gca tct cca ggc aaa gcc act gag gtc cgg gtg aca gtg ctt cgg 192 Tyr Ala Ser Pro Gly Lys Ala Thr Glu Val Arg Val Thr Val Leu Arg 25 30 35 cag gct gac agc cag gtg act gaa gtc tgt gcg gca acc tac atg atg 240 Gln Ala Asp Ser Gln Val Thr Glu Val Cys Ala Ala Thr Tyr Met Met 40 45 50 ggg aat gag ttg acc ttc cta gat gat tcc atc tgc acg ggc acc tcc 288 Gly Asn Glu Leu Thr Phe Leu Asp Asp Ser Ile Cys Thr Gly Thr Ser 55 60 65 70 agt gga aat caa gtg aac ctc act atc caa gga ctg agg gcc atg gac 336 Ser Gly Asn Gln Val Asn Leu Thr Ile Gln Gly Leu Arg Ala Met Asp 75 80 85 acg gga ctc tac atc tgc aag gtg gag ctc atg tac cca ccg cca tac 384 Thr Gly Leu Tyr Ile Cys Lys Val Glu Leu Met Tyr Pro Pro Pro Tyr 90 95 100 tac ctg ggc ata ggc aac gga acc cag att tat gta att gat cca gaa 432 Tyr Leu Gly Ile Gly Asn Gly Thr Gln Ile Tyr Val Ile Asp Pro Glu 105 110 115 ccg tgc cca gat tct gac ttc ctc ctc tgg atc ctt gca gca gtt agt 480 Pro Cys Pro Asp Ser Asp Phe Leu Leu Trp Ile Leu Ala Ala Val Ser 120 125 130 tcg ggg ttg ttt ttt tat agc ttt ctc ctc aca gct gtt tct ttg agc 528 Ser Gly Leu Phe Phe Tyr Ser Phe Leu Leu Thr Ala Val Ser Leu Ser 135 140 145 150 aaa atg cta aag aaa aga agc cct ctt aca aca ggg gtc tat gtg aaa 576 Lys Met Leu Lys Lys Arg Ser Pro Leu Thr Thr Gly Val Tyr Val Lys 155 160 165 atg ccc cca aca gag cca gaa tgt gaa aag caa ttt cag cct tat ttt 624 Met Pro Pro Thr Glu Pro Glu Cys Glu Lys Gln Phe Gln Pro Tyr Phe 170 175 180 att ccc atc aat 636 Ile Pro Ile Asn 185 17 212 PRT Homo sapiens 17 Met Gly Val Leu Leu Thr Gln Arg Thr Leu Leu Ser Leu Val Leu Ala -25 -20 -15 Leu Leu Phe Pro Ser Met Ala Ser Met Ala Met His Val Ala Gln Pro -10 -5 -1 1 5 Ala Val Val Leu Ala Ser Ser Arg Gly Ile Ala Ser Phe Val Cys Glu 10 15 20 Tyr Ala Ser Pro Gly Lys Ala Thr Glu Val Arg Val Thr Val Leu Arg 25 30 35 Gln Ala Asp Ser Gln Val Thr Glu Val Cys Ala Ala Thr Tyr Met Met 40 45 50 Gly Asn Glu Leu Thr Phe Leu Asp Asp Ser Ile Cys Thr Gly Thr Ser 55 60 65 70 Ser Gly Asn Gln Val Asn Leu Thr Ile Gln Gly Leu Arg Ala Met Asp 75 80 85 Thr Gly Leu Tyr Ile Cys Lys Val Glu Leu Met Tyr Pro Pro Pro Tyr 90 95 100 Tyr Leu Gly Ile Gly Asn Gly Thr Gln Ile Tyr Val Ile Asp Pro Glu 105 110 115 Pro Cys Pro Asp Ser Asp Phe Leu Leu Trp Ile Leu Ala Ala Val Ser 120 125 130 Ser Gly Leu Phe Phe Tyr Ser Phe Leu Leu Thr Ala Val Ser Leu Ser 135 140 145 150 Lys Met Leu Lys Lys Arg Ser Pro Leu Thr Thr Gly Val Tyr Val Lys 155 160 165 Met Pro Pro Thr Glu Pro Glu Cys Glu Lys Gln Phe Gln Pro Tyr Phe 170 175 180 Ile Pro Ile Asn 185 18 1152 DNA Artificial CTLA4Ig 18 atg ggt gta ctg ctc aca cag agg acg ctg ctc agt ctg gtc ctt gca 48 Met Gly Val Leu Leu Thr Gln Arg Thr Leu Leu Ser Leu Val Leu Ala -25 -20 -15 ctc ctg ttt cca agc atg gcg agc atg gca atg cac gtg gcc cag cct 96 Leu Leu Phe Pro Ser Met Ala Ser Met Ala Met His Val Ala Gln Pro -10 -5 -1 1 5 gct gtg gta ctg gcc agc agc cga ggc atc gct agc ttt gtg tgt gag 144 Ala Val Val Leu Ala Ser Ser Arg Gly Ile Ala Ser Phe Val Cys Glu 10 15 20 tat gca tct cca ggc aaa gcc act gag gtc cgg gtg aca gtg ctt cgg 192 Tyr Ala Ser Pro Gly Lys Ala Thr Glu Val Arg Val Thr Val Leu Arg 25 30 35 cag gct gac agc cag gtg act gaa gtc tgt gcg gca acc tac atg atg 240 Gln Ala Asp Ser Gln Val Thr Glu Val Cys Ala Ala Thr Tyr Met Met 40 45 50 ggg aat gag ttg acc ttc cta gat gat tcc atc tgc acg ggc acc tcc 288 Gly Asn Glu Leu Thr Phe Leu Asp Asp Ser Ile Cys Thr Gly Thr Ser 55 60 65 70 agt gga aat caa gtg aac ctc act atc caa gga ctg agg gcc atg gac 336 Ser Gly Asn Gln Val Asn Leu Thr Ile Gln Gly Leu Arg Ala Met Asp 75 80 85 acg gga ctc tac atc tgc aag gtg gag ctc atg tac cca ccg cca tac 384 Thr Gly Leu Tyr Ile Cys Lys Val Glu Leu Met Tyr Pro Pro Pro Tyr 90 95 100 tac ctg ggc ata ggc aac gga acc cag att tat gta att gat cca gaa 432 Tyr Leu Gly Ile Gly Asn Gly Thr Gln Ile Tyr Val Ile Asp Pro Glu 105 110 115 ccg tgc cca gat tct gat cag gag ccc aaa tct tct gac aaa act cac 480 Pro Cys Pro Asp Ser Asp Gln Glu Pro Lys Ser Ser Asp Lys Thr His 120 125 130 aca tcc cca ccg tcc cca gca cct gaa ctc ctg ggt gga tcg tca gtc 528 Thr Ser Pro Pro Ser Pro Ala Pro Glu Leu Leu Gly Gly Ser Ser Val 135 140 145 150 ttc ctc ttc ccc cca aaa ccc aag gac acc ctc atg atc tcc cgg acc 576 Phe Leu Phe Pro Pro Lys Pro Lys Asp Thr Leu Met Ile Ser Arg Thr 155 160 165 cct gag gtc aca tgc gtg gtg gtg gac gtg agc cac gaa gac cct gag 624 Pro Glu Val Thr Cys Val Val Val Asp Val Ser His Glu Asp Pro Glu 170 175 180 gtc aag ttc aac tgg tac gtg gac ggc gtg gag gtg cat aat gcc aag 672 Val Lys Phe Asn Trp Tyr Val Asp Gly Val Glu Val His Asn Ala Lys 185 190 195 aca aag ccg cgg gag gag cag tac aac agc acg tac cgg gtg gtc agc 720 Thr Lys Pro Arg Glu Glu Gln Tyr Asn Ser Thr Tyr Arg Val Val Ser 200 205 210 gtc ctc acc gtc ctg cac cag gac tgg ctg aat ggc aag gag tac aag 768 Val Leu Thr Val Leu His Gln Asp Trp Leu Asn Gly Lys Glu Tyr Lys 215 220 225 230 tgc aag gtc tcc aac aaa gcc ctc cca gcc ccc atc gag aaa acc atc 816 Cys Lys Val Ser Asn Lys Ala Leu Pro Ala Pro Ile Glu Lys Thr Ile 235 240 245 tcc aaa gcc aaa ggg cag ccc cga gaa cca cag gtg tac acc ctg ccc 864 Ser Lys Ala Lys Gly Gln Pro Arg Glu Pro Gln Val Tyr Thr Leu Pro 250 255 260 cca tcc cgg gat gag ctg acc aag aac cag gtc agc ctg acc tgc ctg 912 Pro Ser Arg Asp Glu Leu Thr Lys Asn Gln Val Ser Leu Thr Cys Leu 265 270 275 gtc aaa ggc ttc tat ccc agc gac atc gcc gtg gag tgg gag agc aat 960 Val Lys Gly Phe Tyr Pro Ser Asp Ile Ala Val Glu Trp Glu Ser Asn 280 285 290 ggg cag ccg gag aac aac tac aag acc acg cct ccc gtg ctg gac tcc 1008 Gly Gln Pro Glu Asn Asn Tyr Lys Thr Thr Pro Pro Val Leu Asp Ser 295 300 305 310 gac ggc tcc ttc ttc ctc tac agc aag ctc acc gtg gac aag agc agg 1056 Asp Gly Ser Phe Phe Leu Tyr Ser Lys Leu Thr Val Asp Lys Ser Arg 315 320 325 tgg cag cag ggg aac gtc ttc tca tgc tcc gtg atg cat gag gct ctg 1104 Trp Gln Gln Gly Asn Val Phe Ser Cys Ser Val Met His Glu Ala Leu 330 335 340 cac aac cac tac acg cag aag agc ctc tcc ctg tct ccg ggt aaa tga 1152 His Asn His Tyr Thr Gln Lys Ser Leu Ser Leu Ser Pro Gly Lys 345 350 355 19 383 PRT Artificial CTLA4Ig 19 Met Gly Val Leu Leu Thr Gln Arg Thr Leu Leu Ser Leu Val Leu Ala -25 -20 -15 Leu Leu Phe Pro Ser Met Ala Ser Met Ala Met His Val Ala Gln Pro -10 -5 -1 1 5 Ala Val Val Leu Ala Ser Ser Arg Gly Ile Ala Ser Phe Val Cys Glu 10 15 20 Tyr Ala Ser Pro Gly Lys Ala Thr Glu Val Arg Val Thr Val Leu Arg 25 30 35 Gln Ala Asp Ser Gln Val Thr Glu Val Cys Ala Ala Thr Tyr Met Met 40 45 50 Gly Asn Glu Leu Thr Phe Leu Asp Asp Ser Ile Cys Thr Gly Thr Ser 55 60 65 70 Ser Gly Asn Gln Val Asn Leu Thr Ile Gln Gly Leu Arg Ala Met Asp 75 80 85 Thr Gly Leu Tyr Ile Cys Lys Val Glu Leu Met Tyr Pro Pro Pro Tyr 90 95 100 Tyr Leu Gly Ile Gly Asn Gly Thr Gln Ile Tyr Val Ile Asp Pro Glu 105 110 115 Pro Cys Pro Asp Ser Asp Gln Glu Pro Lys Ser Ser Asp Lys Thr His 120 125 130 Thr Ser Pro Pro Ser Pro Ala Pro Glu Leu Leu Gly Gly Ser Ser Val 135 140 145 150 Phe Leu Phe Pro Pro Lys Pro Lys Asp Thr Leu Met Ile Ser Arg Thr 155 160 165 Pro Glu Val Thr Cys Val Val Val Asp Val Ser His Glu Asp Pro Glu 170 175 180 Val Lys Phe Asn Trp Tyr Val Asp Gly Val Glu Val His Asn Ala Lys 185 190 195 Thr Lys Pro Arg Glu Glu Gln Tyr Asn Ser Thr Tyr Arg Val Val Ser 200 205 210 Val Leu Thr Val Leu His Gln Asp Trp Leu Asn Gly Lys Glu Tyr Lys 215 220 225 230 Cys Lys Val Ser Asn Lys Ala Leu Pro Ala Pro Ile Glu Lys Thr Ile 235 240 245 Ser Lys Ala Lys Gly Gln Pro Arg Glu Pro Gln Val Tyr Thr Leu Pro 250 255 260 Pro Ser Arg Asp Glu Leu Thr Lys Asn Gln Val Ser Leu Thr Cys Leu 265 270 275 Val Lys Gly Phe Tyr Pro Ser Asp Ile Ala Val Glu Trp Glu Ser Asn 280 285 290 Gly Gln Pro Glu Asn Asn Tyr Lys Thr Thr Pro Pro Val Leu Asp Ser 295 300 305 310 Asp Gly Ser Phe Phe Leu Tyr Ser Lys Leu Thr Val Asp Lys Ser Arg 315 320 325 Trp Gln Gln Gly Asn Val Phe Ser Cys Ser Val Met His Glu Ala Leu 330 335 340 His Asn His Tyr Thr Gln Lys Ser Leu Ser Leu Ser Pro Gly Lys 345 350 355 20 6 PRT Artificial MYPPPY amino acid sequence 20 Met Tyr Pro Pro Pro Tyr 1 5 21 1152 DNA Artificial CTLA4Ig 21 atg ggt gta ctg ctc aca cag agg acg ctg ctc agt ctg gtc ctt gca 48 Met Gly Val Leu Leu Thr Gln Arg Thr Leu Leu Ser Leu Val Leu Ala -25 -20 -15 ctc ctg ttt cca agc atg gcg agc atg gca atg cac gtg gcc cag cct 96 Leu Leu Phe Pro Ser Met Ala Ser Met Ala Met His Val Ala Gln Pro -10 -5 -1 1 5 gct gtg gta ctg gcc agc agc cga ggc atc gcc agc ttt gtg tgt gag 144 Ala Val Val Leu Ala Ser Ser Arg Gly Ile Ala Ser Phe Val Cys Glu 10 15 20 tat gca tct cca ggc aaa gcc act gag gtc cgg gtg aca gtg ctt cgg 192 Tyr Ala Ser Pro Gly Lys Ala Thr Glu Val Arg Val Thr Val Leu Arg 25 30 35 cag gct gac agc cag gtg act gaa gtc tgt gcg gca acc tac atg atg 240 Gln Ala Asp Ser Gln Val Thr Glu Val Cys Ala Ala Thr Tyr Met Met 40 45 50 ggg aat gag ttg acc ttc cta gat gat tcc atc tgc acg ggc acc tcc 288 Gly Asn Glu Leu Thr Phe Leu Asp Asp Ser Ile Cys Thr Gly Thr Ser 55 60 65 70 agt gga aat caa gtg aac ctc act atc caa gga ctg agg gcc atg gac 336 Ser Gly Asn Gln Val Asn Leu Thr Ile Gln Gly Leu Arg Ala Met Asp 75 80 85 acg gga ctc tac atc tgc aag gtg gag ctc atg tac cca ccg cca tac 384 Thr Gly Leu Tyr Ile Cys Lys Val Glu Leu Met Tyr Pro Pro Pro Tyr 90 95 100 tac ctg ggc ata ggc aac gga acc cag att tat gta att gat cca gaa 432 Tyr Leu Gly Ile Gly Asn Gly Thr Gln Ile Tyr Val Ile Asp Pro Glu 105 110 115 ccg tgc cca gat tct gat cag gag ccc aaa tct tct gac aaa act cac 480 Pro Cys Pro Asp Ser Asp Gln Glu Pro Lys Ser Ser Asp Lys Thr His 120 125 130 aca tcc cca ccg tcc cca gca cct gaa ctc ctg ggg gga tcg tca gtc 528 Thr Ser Pro Pro Ser Pro Ala Pro Glu Leu Leu Gly Gly Ser Ser Val 135 140 145 150 ttc ctc ttc ccc cca aaa ccc aag gac acc ctc atg atc tcc cgg acc 576 Phe Leu Phe Pro Pro Lys Pro Lys Asp Thr Leu Met Ile Ser Arg Thr 155 160 165 cct gag gtc aca tgc gtg gtg gtg gac gtg agc cac gaa gac cct gag 624 Pro Glu Val Thr Cys Val Val Val Asp Val Ser His Glu Asp Pro Glu 170 175 180 gtc aag ttc aac tgg tac gtg gac ggc gtg gag gtg cat aat gcc aag 672 Val Lys Phe Asn Trp Tyr Val Asp Gly Val Glu Val His Asn Ala Lys 185 190 195 aca aag ccg cgg gag gag cag tac aac agc acg tac cgt gtg gtc agc 720 Thr Lys Pro Arg Glu Glu Gln Tyr Asn Ser Thr Tyr Arg Val Val Ser 200 205 210 gtc ctc acc gtc ctg cac cag gac tgg ctg aat ggc aag gag tac aag 768 Val Leu Thr Val Leu His Gln Asp Trp Leu Asn Gly Lys Glu Tyr Lys 215 220 225 230 tgc aag gtc tcc aac aaa gcc ctc cca gcc ccc atc gag aaa acc atc 816 Cys Lys Val Ser Asn Lys Ala Leu Pro Ala Pro Ile Glu Lys Thr Ile 235 240 245 tcc aaa gcc aaa ggg cag ccc cga gaa cca cag gtg tac acc ctg ccc 864 Ser Lys Ala Lys Gly Gln Pro Arg Glu Pro Gln Val Tyr Thr Leu Pro 250 255 260 cca tcc cgg gat gag ctg acc aag aac cag gtc agc ctg acc tgc ctg 912 Pro Ser Arg Asp Glu Leu Thr Lys Asn Gln Val Ser Leu Thr Cys Leu 265 270 275 gtc aaa ggc ttc tat ccc agc gac atc gcc gtg gag tgg gag agc aat 960 Val Lys Gly Phe Tyr Pro Ser Asp Ile Ala Val Glu Trp Glu Ser Asn 280 285 290 ggg cag ccg gag aac aac tac aag acc acg cct ccc gtg ctg gac tcc 1008 Gly Gln Pro Glu Asn Asn Tyr Lys Thr Thr Pro Pro Val Leu Asp Ser 295 300 305 310 gac ggc tcc ttc ttc ctc tac agc aag ctc acc gtg gac aag agc agg 1056 Asp Gly Ser Phe Phe Leu Tyr Ser Lys Leu Thr Val Asp Lys Ser Arg 315 320 325 tgg cag cag ggg aac gtc ttc tca tgc tcc gtg atg cat gag gct ctg 1104 Trp Gln Gln Gly Asn Val Phe Ser Cys Ser Val Met His Glu Ala Leu 330 335 340 cac aac cac tac acg cag aag agc ctc tcc ctg tct ccg ggt aaa tga 1152 His Asn His Tyr Thr Gln Lys Ser Leu Ser Leu Ser Pro Gly Lys 345 350 355 22 383 PRT Artificial CTLA4Ig 22 Met Gly Val Leu Leu Thr Gln Arg Thr Leu Leu Ser Leu Val Leu Ala -25 -20 -15 Leu Leu Phe Pro Ser Met Ala Ser Met Ala Met His Val Ala Gln Pro -10 -5 -1 1 5 Ala Val Val Leu Ala Ser Ser Arg Gly Ile Ala Ser Phe Val Cys Glu 10 15 20 Tyr Ala Ser Pro Gly Lys Ala Thr Glu Val Arg Val Thr Val Leu Arg 25 30 35 Gln Ala Asp Ser Gln Val Thr Glu Val Cys Ala Ala Thr Tyr Met Met 40 45 50 Gly Asn Glu Leu Thr Phe Leu Asp Asp Ser Ile Cys Thr Gly Thr Ser 55 60 65 70 Ser Gly Asn Gln Val Asn Leu Thr Ile Gln Gly Leu Arg Ala Met Asp 75 80 85 Thr Gly Leu Tyr Ile Cys Lys Val Glu Leu Met Tyr Pro Pro Pro Tyr 90 95 100 Tyr Leu Gly Ile Gly Asn Gly Thr Gln Ile Tyr Val Ile Asp Pro Glu 105 110 115 Pro Cys Pro Asp Ser Asp Gln Glu Pro Lys Ser Ser Asp Lys Thr His 120 125 130 Thr Ser Pro Pro Ser Pro Ala Pro Glu Leu Leu Gly Gly Ser Ser Val 135 140 145 150 Phe Leu Phe Pro Pro Lys Pro Lys Asp Thr Leu Met Ile Ser Arg Thr 155 160 165 Pro Glu Val Thr Cys Val Val Val Asp Val Ser His Glu Asp Pro Glu 170 175 180 Val Lys Phe Asn Trp Tyr Val Asp Gly Val Glu Val His Asn Ala Lys 185 190 195 Thr Lys Pro Arg Glu Glu Gln Tyr Asn Ser Thr Tyr Arg Val Val Ser 200 205 210 Val Leu Thr Val Leu His Gln Asp Trp Leu Asn Gly Lys Glu Tyr Lys 215 220 225 230 Cys Lys Val Ser Asn Lys Ala Leu Pro Ala Pro Ile Glu Lys Thr Ile 235 240 245 Ser Lys Ala Lys Gly Gln Pro Arg Glu Pro Gln Val Tyr Thr Leu Pro 250 255 260 Pro Ser Arg Asp Glu Leu Thr Lys Asn Gln Val Ser Leu Thr Cys Leu 265 270 275 Val Lys Gly Phe Tyr Pro Ser Asp Ile Ala Val Glu Trp Glu Ser Asn 280 285 290 Gly Gln Pro Glu Asn Asn Tyr Lys Thr Thr Pro Pro Val Leu Asp Ser 295 300 305 310 Asp Gly Ser Phe Phe Leu Tyr Ser Lys Leu Thr Val Asp Lys Ser Arg 315 320 325 Trp Gln Gln Gly Asn Val Phe Ser Cys Ser Val Met His Glu Ala Leu 330 335 340 His Asn His Tyr Thr Gln Lys Ser Leu Ser Leu Ser Pro Gly Lys 345 350 355

Claims (115)

What is claimed:
1. A method for blocking B7 interactions with CTLA4 and/or CD28 comprising administering to a subject an effective amount of a first agent and a second agent, wherein
a) the first agent is a soluble CTLA4 molecule, and
b) the second agent is selected from a group consisting of immunosuppressive agents, corticosteroids, nonsteroidal antiinflammatory drugs, prednisone, azathioprine, methotrexate, TNFα: blockers or antagonists, infliximab, any biological agent targeting an inflammatory cytokine, hydroxychloroquine, sulphasalazopryine, gold salts, etanercept, anakinra, cyclophosphamide, leflunomide, collagen, dnaJ, a molecule that blocks TNF receptors, pegsunercept, a molecule that blocks cytokine function, AMG719, a molecule that blocks LFA-1 function, efalizumab, acetyl salicylic acid, choline magnesium salicylate, diflunisal, magnesium salicylate, salsalate, sodium salicylate, diclofenac, etodolac, fenoprofen, flurbiprofen, indomethacin, ketoprofen, ketorolac, meclofenamate, naproxen, nabumetone, phenylbutazone, piroxicam, sulindac, tolmetin, acetaminophen, ibuprofen, Cox-2 inhibitors, meloxicam, codeine phosphate, propoxyphene napsylate, oxycodone hydrochloride, oxycodone bitartrate and tramadol.
2. A method for treating an immune system disease comprising administering to a subject an effective amount of a first agent and a second agent, wherein
a) the first agent is a soluble CTLA4 molecule, and
b) the second agent is selected from a group consisting of immunosuppressive agents, corticosteroids, nonsteroidal antiinflammatory drugs, prednisone, azathioprine, methotrexate, TNFα blockers or antagonists, infliximab, any biological agent targeting an inflammatory cytokine, hydroxychloroquine, sulphasalazopryine, gold salts, etanercept, anakinra, cyclophosphamide, leflunomide, collagen, dnaJ, a molecule that blocks TNF receptors, pegsunercept, a molecule that blocks cytokine function, AMG719, a molecule that blocks LFA-1 function, efalizumab, acetyl salicylic acid, choline magnesium salicylate, diflunisal, magnesium salicylate, salsalate, sodium salicylate, diclofenac, etodolac, fenoprofen, flurbiprofen, indomethacin, ketoprofen, ketorolac, meclofenamate, naproxen, nabumetone, phenylbutazone, piroxicam, sulindac, tolmetin, acetaminophen, ibuprofen, Cox-2 inhibitors, meloxicam, codeine phosphate, propoxyphene napsylate, oxycodone hydrochloride, oxycodone bitartrate and tramadol.
3. The method of claim 1 or 2, wherein cytokine production is regulated.
4. The method of claim 1 or 2, wherein ACR 20, 50 and/or 70 response rates are improved.
5. A method for blocking B7 interactions with CTLA4 and/or CD28 comprising administering to a subject an effective amount of a soluble CTLA4 molecule, wherein the effective amount of soluble CTLA4 is about 0.1 to 100 mg/kg weight of the subject, about 0.5 to 5 mg/kg weight of a subject, about 5 to 10 mg/kg weight of a subject, about 10 to 15 mg/kg weight of a subject, about 15 to 20 mg/kg weight of a subject, about 20 to 25 mg/kg weight of a subject, about 25 to 30 mg/kg weight of a subject, about 30 to 35 mg/kg weight of a subject, about 35 to 40 mg/kg weight of a subject, about 40 to 45 mg/kg of a subject, about 45 to 50 mg/kg weight of a subject, about 50 to 55 mg/kg weight of a subject, about 55 to 60 mg/kg weight of a subject, about 60 to 65 mg/kg weight of a subject, about 65 to 70 mg/kg weight of a subject, about 70 to 75 mg/kg weight of a subject, about 75 to 80 mg/kg weight of a subject, about 80 to 85 mg/kg weight of a subject, about 85 to 90 mg/kg weight of a subject, about 90 to 95 mg/kg weight of a subject, about 95 to 100 mg/kg weight of a subject, about 2 to 10 mg/kg weight of a subject, about 0.1 to 4 mg/kg weight of a subject, about 0.1 to 0.5 mg/kg weight of a subject, about 0.5 to 1.0 mg/kg weight of a subject, about 1.0 to 1.5 mg/kg weight of a subject, about 1.5 to 2.0 mg/kg weight of a subject, about 2.0 to 2.5 mg/kg weight of a subject, about 2.5 to 3.0 mg/kg weight of a subject, about 3.0 to 3.5 mg/kg weight of a subject, about 3.5 to 4.0 mg/kg weight of a subject, about 4.0 to 4.5 mg/kg weight of a subject, about 4.5 to 5.0 mg/kg weight of a subject, about 5.0 to 5.5 mg/kg weight of a subject, about 5.5 to 6.0 mg/kg weight of a subject, about 6.0 to 6.5 mg/kg weight of a subject, about 6.5 to 7.0 mg/kg weight of a subject, about 7.0 to 7.5 mg/kg weight of a subject, about 7.5 to 8.0 mg/kg weight of a subject, about 8.0 to 8.5 mg/kg weight of a subject, about 8.5 to 9.0 mg/kg weight of a subject, about 9.0 to 9.5 mg/kg weight of a subject, about 9.5 to 10.0 mg/kg weight of a subject, about 0.1 to 2 mg/kg weight of a subject, about 2 to 4 mg/kg weight of a subject, about 4 to 6 mg/kg weight of a subject, about 6 to 8 mg/kg weight of a subject, about 8 to 10 mg/kg weight of a subject, about 10 to 12 mg/kg weight of a subject, about 12 to 14 mg/kg weight of a subject, about 14 to 16 mg/kg weight of a subject, about 16 to 18 mg/kg weight of a subject, about 18 to 20 mg/kg weight of a subject, about 0.5 mg/kg weight of the subject, 2 mg/kg weight of the subject, 10 mg/kg weight of the subject, about 0.5 mg/kg to 100 weight of the subject, about 0.5 to 10 mg/kg weight of a subject, about 0.1 to 20 mg/kg weight of a subject, about 500 mg for a subject weighing less than 60 kg, 750 mg for a subject weighing between 60-100 kg or 1000 mg for a subject weighing more than 100 kg).
6. A method for treating an immune system disease comprising administering to a subject an effective amount of a soluble CTLA4 molecule, wherein the effective amount of soluble CTLA4 is about 0.1 to 100 mg/kg weight of the subject, about 0.5 to 5 mg/kg weight of a subject, about 5 to 10 mg/kg weight of a subject, about 10 to 15 mg/kg weight of a subject, about 15 to 20 mg/kg weight of a subject, about. 20 to 25 mg/kg weight of a subject, about 25 to 30 mg/kg weight of a subject, about 30 to 35 mg/kg weight of a subject, about 35 to 40 mg/kg weight of a subject, about 40 to 45 mg/kg weight of a subject, about 45 to 50 mg/kg weight of a subject, about 50 to 55 mg/kg weight of a subject, about 55 to 60 mg/kg weight of a subject, about 60 to 65 mg/kg weight of a subject, about 65 to 70 mg/kg weight of a subject, about 70 to 75 mg/kg weight of a subject, about 75 to 80 mg/kg weight of a subject, about 80 to 85 mg/kg weight of a subject, about 85 to 90 mg/kg weight of a subject, about 90 to 95 mg/kg weight of a subject, about 95 to 100 mg/kg weight of a subject, about 2 to 10 mg/kg weight of a subject, about 0.1 to 4 mg/kg weight of a subject, about 0.1 to 0.5 mg/kg weight of a subject, about 0.5 to 1.0 mg/kg weight of a subject, about 1.0 to 1.5 mg/kg weight of a subject, about 1.5 to 2.0 mg/kg weight of a subject, about 2.0 to 2.5 mg/kg weight of a subject, about 2.5 to 3.0 mg/kg weight of a subject, about 3.0 to 3.5 mg/kg weight of a subject, about 3.5 to 4.0 mg/kg weight of a subject, about 4.0 to 4.5 mg/kg weight of a subject, about 4.5 to 5.0 mg/kg weight of a subject, about 5.0 to 5.5 mg/kg weight of a subject, about 5.5 to 6.0 mg/kg weight of a subject, about 6.0 to 6.5 mg/kg weight of a subject, about 6.5 to 7.0 mg/kg weight of a subject, about 7.0 to 7.5 mg/kg weight of a subject, about 7.5 to 8.0 mg/kg weight of a subject, about 8.0 to 8.5 mg/kg weight of a subject, about 8.5 to 9.0 mg/kg weight of a subject, about 9.0 to 9.5 mg/kg weight of a subject, about 9.5 to 10.0 mg/kg weight of a subject, about 0.1 to 2 mg/kg weight of a subject, about 2 to 4 mg/kg weight of a subject, about 4 to 6 mg/kg weight of a subject, about 6 to 8 mg/kg weight of a subject, about 8 to 10 mg/kg weight of a subject, about 10 to 12 mg/kg weight of a subject, about 12 to 14 mg/kg weight of a subject, about 14 to 16 mg/kg weight of a subject, about 16 to 18 mg/kg weight of a subject, about 18 to 20 mg/kg weight of a subject, about 0.5 mg/kg weight of the subject, 2 mg/kg weight of the subject, 10 mg/kg weight of the subject, about 0.5 mg/kg to 100 weight of the subject, about 0.5 to 10 mg/kg weight of a subject, about 0.1 to 20 mg/kg weight of a subject, about 500 mg for a subject weighing less than 60 kg, 750 mg for a subject weighing between 60-100 kg or 1000 mg for a subject weighing more than 100 kg.
7. The method of claim 5 or 6, wherein cytokine production is regulated.
8. The method of claim 5 or 6, wherein ACR 20, 50 and/or 70 response rates are improved.
9. The method of claim 1, 2, 5, or 6, wherein the soluble CTLA4 molecule comprises an extracellular domain of a CTLA4 molecule, or portion thereof, which binds a B7 antigen expressed on activated B cells.
10. The method of claim 9, wherein the extracellular domain of the CTLA4 molecule comprises the amino acids shown in FIG. 23 (SEQ ID NO: 17) beginning with methionine at position 1 and or with alanine at position −1 and ending with aspartic acid at position 124.
11. The method of claim 1, 2, 5, or 6, wherein the soluble CTLA4 molecule is a CTLA4 fusion molecule.
12. The method of claim 11, wherein the CTLA4 fusion molecule comprises an extracellular domain of a CTLA4 molecule which binds a B7 antigen expressed on activated B cells joined to a non-CTLA4 molecule.
13. The method of 12, wherein the non-CTLA4 molecule comprises an amino acid sequence which alters the solubility or affinity of the soluble CTLA4 molecule.
14. The method of claim 13, wherein the amino acid sequence which alters the solubility or affinity comprises an immunoglobulin moiety.
15. The method of claim 14, wherein the immunoglubulin moiety comprises one or more mutations to reduce effector function.
16. The method of claim 14, wherein the immunoglubulin moiety is an immunoglubulin constant region or portion thereof.
17. The method of claim 15, wherein the immunoglubulin constant region or portion thereof is mutated to reduce effector function.
18. The method of claim 15, wherein the immunoglubulin constant region comprises a hinge, CH2 and CH3 regions of an immunoglobulin molecule.
19. The method of claim 15, wherein the immunoglubulin constant region or portion thereof is a human or monkey immunoglobulin constant region.
20. The method of claim 1, 2, 5, or 6, wherein the soluble CTLA4 further comprises an amino acid sequence which permits secretion of the soluble CTLA4 molecule.
21. The method of claim 20, wherein the amino acid sequence which permits secretion comprises an oncostatin M signal peptide.
22. The method of claim 11, wherein the CTLA4 fusion molecule is CTLA4Ig.
23. The method of claim 22, wherein the CTLA4Ig is shown in FIG. 24 (SEQ ID NO:19) beginning with methionine at position +1 or with alanine at position −1 and ending with lysine at position +357.
24. The method of claim 1, 2, 5, or 6, wherein the soluble CTLA4 molecule is a soluble CTLA4 mutant molecule.
25. The method of claim 24, wherein the soluble CTLA4 mutant molecule binds a B7 antigen expressed on activated B cells and comprises a mutation in the extracellular domain of a CTLA4 molecule.
26. The method of claim 24, wherein the soluble CTLA4 mutant molecule comprises an extracellular domain of CTLA4,
a) wherein the extracellular domain of CTLA4 begins with methionine at position +1 or with alanine at position −1 and ends with aspartic acid at position +124 as shown in FIG. 23 or 24 (SEQ ID NO: 17 or 19), and
b) wherein at position +104 of the extracellular domain of CTLA4, leucine is substituted with any other amino acid.
27. The method of claim 24, wherein the soluble CTLA4 mutant molecule comprises an extracellular domain of CTLA4,
a) wherein the extracellular domain of CTLA4 begins with methionine at position +1 or with alanine at position −1 and ends with aspartic acid at position +124 as shown in FIG. 23 or 24 (SEQ ID NO: 17 or 19),
b) wherein at position +104 of the extracellular domain of CTLA4, leucine is substituted with glutamic acid, and
c) wherein at position +29 of the extracellular domain of CTLA4, alanine is substituted with tyrosine.
28. The method of claim 24, wherein the soluble CTLA4 mutant molecule is L104EA29YIg as shown in FIG. 19 (SEQ ID NO: 9) beginning with methionine at position +1 or with alanine at position −1 and ending with lysine at position +357 as shown in FIG. 19 (SEQ ID NO: 9).
29. A method for treating an immune system disease comprising administering to a subject a combination of an effective amount of a soluble CTLA4 and a second agent, wherein
a) the soluble CTLA4 is a CTLA4Ig beginning with methionine at position +1 or with alanine at position −1 and ending with lysine as position +357 as shown in FIG. 24 (SEQ ID NO: 19), and
b) the second agent is selected from a group consisting of immunosuppressive agents, corticosteroids, nonsteroidal antiinflammatory drugs, prednisone, azathioprine, methotrexate, TNFα blockers or antagonists, infliximab, any biological agent targeting an inflammatory cytokine, hydroxychloroquine, sulphasalazopryine, gold salts, etanercept, anakinra, cyclophosphamide, leflunomide, collagen, dnaJ, a molecule that blocks TNF receptors, pegsunercept, a molecule that blocks cytokine function, AMG719, a molecule that blocks LFA-1 function, efalizumab, acetyl salicylic acid, choline magnesium salicylate, diflunisal, magnesium salicylate, salsalate, sodium salicylate, diclofenac, etodolac, fenoprofen, flurbiprofen, indomethacin, ketoprofen, ketorolac, meclofenamate, naproxen, nabumetone, phenylbutazone, piroxicam, sulindac, tolmetin, acetaminophen, ibuprofen, Cox-2 inhibitors, meloxicam, codeine phosphate, propoxyphene napsylate, oxycodone hydrochloride, oxycodone bitartrate and tramadol.
30. A method for treating an immune system disease comprising administering to a subject a combination of an effective amount of a soluble CTLA4 and a second agent, wherein
a) the soluble CTLA4 comprises the extracellular domain of a CTLA4 molecule as shown in FIG. 23 (SEQ ID NO: 17) beginning with methionine at position +1 or with alanine at position −1 and ending with aspartic acid at position +124, and
b) the second agent is selected from a group consisting of immunosuppressive agents, corticosteroids, nonsteroidal antiinflammatory drugs, prednisone, azathioprine, methotrexate, TNFα blockers or antagonists, infliximab, any biological agent targeting an inflammatory cytokine, hydroxychloroquine, sulphasalazopryine, gold salts, etanercept, anakinra, cyclophosphamide, leflunomide, collagen, dnaJ, a molecule that blocks TNF receptors, pegsunercept, a molecule that blocks cytokine function, AMG719, a molecule that blocks LFA-1 function, efalizumab, acetyl salicylic acid, choline magnesium salicylate, diflunisal, magnesium salicylate, salsalate, sodium salicylate, diclofenac, etodolac, fenoprofen, flurbiprofen, indomethacin, ketoprofen, ketorolac, meclofenamate, naproxen, nabumetone, phenylbutazone, piroxicam, sulindac, tolmetin, acetaminophen, ibuprofen, Cox-2 inhibitors, meloxicam, codeine phosphate, propoxyphene napsylate, oxycodone hydrochloride, oxycodone bitartrate and tramadol.
31. A method for treating an immune system disease comprising administering to a subject a combination of an effective amount of a soluble CTLA4 and a second agent, wherein
a) the soluble CTLA4 mutant molecule comprises an extracellular domain of CTLA4,
i) wherein the extracellular domain of CTLA4 begins with methionine at position +1 or with alanine at position −1 and ends with aspartic acid at position +124 as shown in FIG. 23 or 24 (SEQ ID NO: 17 or 19), and
ii) wherein at position +104 of the extracellular domain of CTLA4, leucine is substituted with any other amino acid.
b) the second agent is selected from a group consisting of immunosuppressive agents, corticosteroids, nonsteroidal antiinflammatory drugs, prednisone, azathioprine, methotrexate, TNFα blockers or antagonists, infliximab, any biological agent targeting an inflammatory cytokine, hydroxychloroquine, sulphasalazopryine, gold salts, etanercept, anakinra, cyclophosphamide, leflunomide, collagen, dnaJ, a molecule that blocks TNF receptors, pegsunercept, a molecule that blocks cytokine function, AMG719, a molecule that blocks LFA-1 function, efalizumab, acetyl salicylic acid, choline magnesium salicylate, diflunisal, magnesium salicylate, salsalate, sodium salicylate, diclofenac, etodolac, fenoprofen, flurbiprofen, indomethacin, ketoprofen, ketorolac, meclofenamate, naproxen, nabumetone, phenylbutazone, piroxicam, sulindac, tolmetin, acetaminophen, ibuprofen, Cox-2 inhibitors, meloxicam, codeine phosphate, propoxyphene napsylate, oxycodone hydrochloride, oxycodone bitartrate and tramadol.
32. The method of claim 31, wherein the soluble CTLA4 mutant molecule comprises an extracellular domain of CTLA4,
a) wherein the extracellular domain of CTLA4 begins with methionine at position +1 or with alanine at position −1 and ends with aspartic acid at position +124 as shown in FIG. 23 or 24 (SEQ ID NO: 17 or 19),
b) wherein at position +104 of the extracellular domain of CTLA4, leucine is substituted with glutamic acid, and
c) wherein at position +29 of the extracellular domain of CTLA4, alanine is substituted with tyrosine.
33. The method of claim 31, wherein the soluble CTLA4 mutant molecule is L104EA29YIg as shown in FIG. 19 (SEQ ID NO: 9) beginning with methionine at position +1 or with alanine at position −1 and ending with lysine at position +357 as shown in FIG. 19 (SEQ ID NO: 9).
34. A method for treating an immune system disease comprising administering to a subject a combination of an effective amount of a soluble CTLA4 and a second agent, wherein
a) the soluble CTLA4 is L104EA29YIg beginning with methionine at position +1 or with alanine at position −1 and ending with lysine as position +357 as shown in FIG. 19 (SEQ ID NO: 9), and
b) the second agent is selected from a group consisting of immunosuppressive agents, corticosteroids, nonsteroidal antiinflammatory drugs, prednisone, azathioprine, methotrexate, TNFα blockers or antagonists, infliximab, any biological agent targeting an inflammatory cytokine, hydroxychloroquine, sulphasalazopryine, gold salts, etanercept, anakinra, cyclophosphamide, leflunomide, collagen, dnaJ, a molecule that blocks TNF receptors, pegsunercept, a molecule that blocks cytokine function, AMG719, a molecule that blocks LFA-1 function, efalizumab, acetyl salicylic acid, choline magnesium salicylate, diflunisal, magnesium salicylate, salsalate, sodium salicylate, diclofenac, etodolac, fenoprofen, flurbiprofen, indomethacin, ketoprofen, ketorolac, meclofenamate, naproxen, nabumetone, phenylbutazone, piroxicam, sulindac, tolmetin, acetaminophen, ibuprofen, Cox-2 inhibitors, meloxicam, codeine phosphate, propoxyphene napsylate, oxycodone hydrochloride, oxycodone bitartrate and tramadol.
35. A method for blocking B7 interactions with CTLA4 and/or CD28 comprising administering to a subject an effective amount of a first agent and a second agent, wherein
a) the first agent is a soluble CTLA4 molecule, and
b) the second agent is Methotrexate,
wherein the effective amount of Methotrexate is about 0.1 to 40 mg per week, about 5 to 30 mg per week, about 0.1 to 5 mg/week, about 5 to 10 mg/week, about 10 to 15 mg/week, about 15 to 20 mg/week, about 20 to 25 mg/week, about 25 to 30 mg/week, about 30 to 35 mg/week, about 35 to 40 mg/week or about 10 to 30 mg/week.
36. A method for treating an immune system disease comprising administering to a subject an effective amount of a first agent and a second agent, wherein
a) the first agent is a soluble CTLA4 molecule, and
b) the second agent is Methotrexate,
wherein the effective amount of Methotrexate is about 0.1 to 40 mg per week, about 5 to 30 mg per week, about 0.1 to 5 mg/week, about 5 to 10 mg/week, about 10 to 15 mg/week, about 15 to 20 mg/week, about 20 to 25 mg/week, about 25 to 30 mg/week, about 30 to 35 mg/week, about 35 to 40 mg/week or about 10 to 30 mg/week.
37. A method for blocking B7 interactions with CTLA4 and/or CD28 comprising administering to a subject an effective amount of a first agent and a second agent, wherein
a) the first agent is a soluble CTLA4 molecule, and
b) the second agent is Etanercept,
wherein the effective amount of Etanercept is about 10 to 100 mg per week, about 50 mg per week, about 0.1 to 50 mg/kg body weight per week
38. A method for treating an immune system disease comprising administering to a subject an effective amount of a first agent and a second agent, wherein
a) the first agent is a soluble CTLA4 molecule, and
b) the second agent is Etanercept,
wherein the effective amount of Etanercept is about 10 to 100 mg per week, about 50 mg per week, about 0.1 to 50 mg/kg body weight per week
39. A method for blocking B7 interactions with CTLA4 and/or CD28 comprising administering to a subject an effective amount of a first agent and a second agent, wherein
a) the first agent is a soluble CTLA4 molecule, and
b) the second agent is a DMARD,
wherein the effective amount of the DMARD is about 1 to about 5000 mg/day, about 1 to 10 mg/day, about 10 to 50 mg/day, about 50 to 100 mg/day, about 100 to 150 mg/day, about 150 to 200 mg/day, about 200 to 250 mg/day, about 250 to 300 mg/day, about 300 to 350 mg/day, about 350 to 400 mg/day, about 400 to 450 mg/day, about 450 to 500 mg/day, about 500 to 550 mg/day, about 550 to 600 mg/day, about 600 to 650 mg/day, about 650 to 700 mg/day, about 700 to 750 mg/day, about 750 to 800 mg/day, about 800 to 850 mg/day, about 850 to 900 mg/day, about 900 to 950 mg/day, about 950 to 1000 mg/day, about 1000 to 1100 mg/day, about 1100 to 1200 mg/day, about 1200 to 1300 mg/day, about 1300 to 1400 mg/day, about 1400 to 1500 mg/day, about 1500 to 1600 mg/day, about 1600 to 1700 mg/day, about 1700 to 1800 mg/day, about 1800 to 1900 mg/day, about 1900 to 2000 mg/day, about 2000 to 2500 mg/day, about 2500 to 3000 mg/day, about 3000 to 3500 mg/day, about 3500 to 4000 mg/day, about 4000 to 4500 mg/day or about 4500 to 5000 mg/day.
40. A method for treating an immune system disease administering to a subject an effective amount of a first agent and a second agent, wherein
a) the first agent is a soluble CTLA4 molecule, and
b) the second agent is a DMARD,
wherein the effective amount of the DMARD is about 1 to about 5000 mg/day, about 1 to 10 mg/day, about 10 to 50 mg/day, about 50 to 100 mg/day, about 100 to 150 mg/day, about 150 to 200 mg/day, about 200 to 250 mg/day, about 250 to 300 mg/day, about 300 to 350 mg/day, about 350 to 400 mg/day, about 400 to 450 mg/day, about 450 to 500 mg/day, about 500 to 550 mg/day, about 550 to 600 mg/day, about 600 to 650 mg/day, about 650 to 700 mg/day, about 700 to 750 mg/day, about 750 to 800 mg/day, about 800 to 850 mg/day, about 850 to 900 mg/day, about 900 to 950 mg/day, about 950 to 1000 mg/day, about 1000 to 1100 mg/day, about 1100 to 1200 mg/day, about 1200 to 1300 mg/day, about 1300 to 1400 mg/day, about 1400 to 1500 mg/day, about 1500 to 1600 mg/day, about 1600 to 1700 mg/day, about 1700 to 1800 mg/day, about 1800 to 1900 mg/day, about 1900 to 2000 mg/day, about 2000 to 2500 mg/day, about 2500 to 3000 mg/day, about 3000 to 3500 mg/day, about 3500 to 4000 mg/day, about 4000 to 4500 mg/day or about 4500 to 5000 mg/day.
41. The method of claim 40, wherein the DMARD is selected from a group consisting of a dihydrofolic acid reductase inhibitor, cyclophosphamide, cyclosporine, cyclosporin A, chloroquine, hydroxychloroquine, sulfasalazine (sulphasalazopyrine), gold salts, D-penicillamine, leflunomide, azathioprine, anakinra, a TNF blocker, a biological agent that targets an inflammatory cytokine, methotrexate or etanercept.
42. The method of claim 1, 2, 29, 30, 31, 34, 35, 36, 37, 38, 39 or 40, wherein the effective amount of soluble CTLA4 is between about 0.5 through 100 mg/kg weight of the subject, about 0.1 to 100 mg/kg weight of the subject, about 0.5 to 10 mg/kg weight of a subject, about 0.5 to 5 mg/kg weight of a subject, about 5 to 10 mg/kg weight of a subject, about 10 to 15 mg/kg weight of a subject, about 15 to 20 mg/kg weight of a subject, about 20 to 25 mg/kg weight of a subject, about 25 to 30 mg/kg weight of a subject, about 30 to 35 mg/kg weight of a subject, about 35 to 40 mg/kg weight of a subject, about 40 to 45 mg/kg weight of a subject, about 45 to 50 mg/kg weight of a subject, about 50 to 55 mg/kg weight of a subject, about 55 to 60 mg/kg weight of a subject, about 60 to 65 mg/kg weight of a subject, about 65 to 70 mg/kg weight of a subject, about 70 to 75 mg/kg weight of a subject, about 75 to 80 mg/kg weight of a subject, about 80 to 85 mg/kg weight of a subject, about 85 to 90 mg/kg weight of a subject, about 90 to 95 mg/kg weight of a subject, about 95 to 100 mg/kg weight of a subject, about 2 to 10 mg/kg weight of a subject, about 0.1 to 4 mg/kg weight of a subject, about 0.1 to 0.5 mg/kg weight of a subject, about 0.5 to 1.0 mg/kg weight of a subject, about 1.0 to 1.5 mg/kg weight of a subject, about 1.5 to 2.0 mg/kg weight of a subject, about 2.0 to 2.5 mg/kg weight of a subject, about 2.5 to 3.0 mg/kg weight of a subject, about 3.0 to 3.5 mg/kg weight of a subject, about 3.5 to 4.0 mg/kg about 4.0 to 4.5 mg/kg weight of a subject, about 4.5 to 5.0 mg/kg weight of a subject, about 5.0 to 5.5 mg/kg weight of a subject, about 5.5 to 6.0 mg/kg weight of a subject, about 6.0 to 6.5 mg/kg weight of a subject, about 6.5 to 7.0 mg/kg weight of a subject, about 7.0 to 7.5 mg/kg weight of a subject, about 7.5 to 8.0 mg/kg weight of a subject, about 8.0 to 8.5 mg/kg weight of a subject, about 8.5 to 9.0 mg/kg weight of a subject, about 9.0 to 9.5 mg/kg weight of a subject, about 9.5 to 10.0 mg/kg weight of a subject, weight of a subject, about 0.1 to 20 mg/kg weight of a subject, about 0.1 to 2 mg/kg weight of a subject, about 2 to 4 mg/kg weight of a subject, about 4 to 6 mg/kg weight of a subject, about 6 to 8 mg/kg weight of a subject, about 8 to 10 mg/kg weight of a subject, about 10 to 12 mg/kg weight of a subject, about 12 to 14 mg/kg weight of a subject, about 14 to 16 mg/kg weight of a subject, about 16 to 18 mg/kg weight of a subject, about 18 to 20 mg/kg weight of a subject, 0.5 mg/kg weight of the subject, 2 mg/kg weight of a subject, 10 mg/kg weight of a subject, about 0.5 mg/kg weight of the subject, 2 mg/kg weight of the subject, 10 mg/kg weight of the subject, about 0.5 mg/kg to 100 weight of the subject, about 0.5 to 10 mg/kg weight of a subject, about 0.1 to 20 mg/kg weight of a subject, about 500 mg for a subject weighing less than 60 kg, 750 mg for a subject weighing between 60-100 kg or 1000 mg for a subject weighing more than 100 kg.
43. The method of claim 2, 6, 29, 30, 31, 34, 36, 38 or 40, wherein the immune system disease is a rheumatic disease.
44. The method of claim 43, wherein the rheumatic disease is rheumatoid arthritis.
45. The method of claim 2, 6, 29, 30, 31, 34, 36, 38 or 40,, wherein the immune system disease is selected from autoimmune diseases, psoriasis, immune disorders associated with graft transplantation rejection, T cell lymphoma, T cell acute lymphoblastic leukemia, testicular angiocentric T cell lymphoma, benign lymphocytic angiitis, lupus erythematosus, Hashimoto's thyroiditis, primary myxedema, Graves' disease, pernicious anemia, autoimmune atrophic gastritis, Addison's disease, insulin dependent diabetes mellitis, good pasture's syndrome, myasthenia gravis, pemiphigus, Crohn's disease, sympathetic ophthalmia, autoimmune uveitis, multiple sclerosis, autoimmune hemolytic anemia, idiopathic thrombocytopenia, primary biliary cirrhosis, chronic action hepatitis, ulceratis colitis, Sjogren's syndrome, rheumatic disease, rheumatoid arthritis, polymyositis, scleroderma, mixed connective tissue disease, inflammatory rheumatism, degenerative rheumatism, extra-articular rheumatism, collagen diseases, chronic polyarthritis, psoriasis arthropathica, ankylosing spondylitis, juvenile rheumatoid arthritis, periarthritis humeroscapularis, panarteriitis nodosa, systemic lupus erythematosus, progressive systemic scleroderma, arthritis uratica, dermatomyositis, muscular rheumatism, myositis, myogelosis and chondrocalcinosis.
46. The method of claim 2, 6, 29, 30, 31, 34, 36, 38 or 40,, wherein the immune system disease is selected from graft related disorders, chronic rejection, tissue or cell allo- or xenografts, skin allo- or xenografts, islet allo- or xenografts, muscle allo- or xenografts, hepatocyte allo- or xenografts and neuron allo- or xenografts.
47. The method of claim 2, 6, 29, 30, 31, 34, 36, 38 or 40,, wherein a symptom associated with the immune system disease is alleviated and wherein the symptom is selected from the group consisting of joint swelling, pain, tenderness, morning stiffness, structural damage, an elevated level of serum C-reactive protein, an elevated level of soluble IL-2r, an elevated level of soluble ICAM-1, an elevated level of soluble E-selectin and an elevated erythrocyte sedimentation rate.
48. The method of claim 2, 6, 29, 30, 31, 34, 36, 38 or 40, wherein treating a subject suffering from an immune system disease induces a pathophysiological change.
49. The method of claim 48, wherein the pathophysiological change associated with the immune system disease is reduced structural damage.
50. A method for treating a disease selected from autoimmune diseases, psoriasis, immune disorders associated with graft transplantation rejection, T cell lymphoma, T cell acute lymphoblastic leukemia, testicular angiocentric T cell lymphoma, benign lymphocytic angiitis, lupus erythematosus, Hashimoto's thyroiditis, primary myxedema, Graves' disease, pernicious anemia, autoimmune atrophic gastritis, Addison's disease, insulin dependent diabetes mellitis, good pasture's syndrome, myasthenia gravis, pemphigus, Crohn's disease, sympathetic ophthalmia, autoimmune uveitis, multiple sclerosis, autoimmune hemolytic anemia, idiopathic thrombocytopenia, primary biliary cirrhosis, chronic action hepatitis, ulceratis colitis, Sjogren's syndrome, rheumatic disease, rheumatoid arthritis, polymyositis, scleroderma, mixed connective tissue disease, inflammatory rheumatism, degenerative rheumatism, extra-articular rheumatism, collagen diseases, chronic polyarthritis, psoriasis arthropathica, ankylosing spondylitis, juvenile rheumatoid arthritis, periarthritis humeroscapularis, panarteriitis nodosa, systemic lupus erythematosus, progressive systemic scleroderma, arthritis uratica, dermatomyositis, muscular rheumatism, myositis, myogelosis and chondrocalcinosis, by administering to a subject an effective amount of a soluble CTLA4 molecule comprising the sequence shown in FIG. 19 (SEQ ID NO: 9) beginning with methionine at position +1 or with alanine at position −1 and ending with aspartic acid at position +124, and wherein the effective amount is about 0.1 to 100 mg/kg weight of the subject, about 0.5 to 5 mg/kg weight of a subject, about 5 to 10 mg/kg weight of a subject, about 10 to 15 mg/kg weight of a subject, about 15 to 20 mg/kg weight of a subject, about 20 to 25 mg/kg weight of a subject, about 25 to 30 mg/kg weight of a subject, about 30 to 35 mg/kg weight of a subject, about 35 to 40 mg/kg weight of a subject, about 40 to 45 mg/kg weight of a subject, about 45 to 50 mg/kg weight of a subject, about 50 to 55 mg/kg weight of a subject, about 55 to 60 mg/kg weight of a subject, about 60 to 65 mg/kg weight of a subject, about 65 to 70 mg/kg weight of a subject, about 70 to 75 mg/kg weight of a subject, about 75 to 80 mg/kg weight of a subject, about 80 to 85 mg/kg weight of a subject, about 85 to 90 mg/kg weight of a subject, about 90 to 95 mg/kg weight of a subject, about 95 to 100 mg/kg weight of a subject, about 2 to 10 mg/kg weight of a subject, about 0.1 to 4 mg/kg weight of a subject, about 0.1 to 0.5 mg/kg weight of a subject, about 0.5 to 1.0 mg/kg weight of a subject, about 1.0 to 1.5 mg/kg weight of a subject, about 1.5 to 2.0 mg/kg weight of a subject, about 2.0 to 2.5 mg/kg weight of a subject, about 2.5 to 3.0 mg/kg weight of a subject, about 3.0 to 3.5 mg/kg weight of a subject, about 3.5 to 4.0 mg/kg weight of a subject, about 4.0 to 4.5 mg/kg weight of a subject, about 4.5 to 5.0 mg/kg weight of a subject, about 5.0 to 5.5 mg/kg weight of a subject, about 5.5 to 6.0 mg/kg weight of a subject, about 6.0 to 6.5 mg/kg weight of a subject, about 6.5 to 7.0 mg/kg weight of a subject, about 7.0 to 7.5 mg/kg weight of a subject, about 7.5 to 8.0 mg/kg weight of a subject, about 8.0 to 8.5 mg/kg weight of a subject, about 8.5 to 9.0 mg/kg weight of a subject, about 9.0 to 9.5 mg/kg weight of a subject, about 9.5 to 10.0 mg/kg weight of a subject, about 0.1 to 2 mg/kg weight of a subject, about 2 to 4 mg/kg weight of a subject, about 4 to 6 mg/kg weight of a subject, about 6 to 8 mg/kg weight of a subject, about 8 to 10 mg/kg weight of a subject, about 10 to 12 mg/kg weight of a subject, about 12 to 14 mg/kg weight of a subject, about 14 to 16 mg/kg weight of a subject, about 16 to 18 mg/kg weight of a subject, about 18 to 20 mg/kg weight of a subject about 0.5 mg/kg weight of the subject, 2 mg/kg weight of the subject, 10 mg/kg weight of the subject, about 0.5 mg/kg to 100 weight of the subject, about 0.5 to 10 mg/kg weight of a subject, about 0.1 to 20 mg/kg weight of a subject, about 500 mg for a subject weighing less than 60 kg, 750 mg for a subject weighing between 60-100 kg or 1000 mg for a subject weighing more than 100 kg.
51. A method for treating a disease selected from autoimmune diseases, psoriasis, immune disorders associated with graft transplantation rejection, T cell lymphoma, T cell acute lymphoblastic leukemia, testicular angiocentric T cell lymphoma, benign lymphocytic angiitis, lupus erythematosus, Hashimoto's thyroiditis, primary myxedema, Graves' disease, pernicious anemia, autoimmune atrophic gastritis, Addison's disease, insulin dependent diabetes mellitis, good pasture's syndrome, myasthenia gravis, pemphigus, Crohn's disease, sympathetic ophthalmia, autoimmune uveitis, multiple sclerosis, autoimmune hemolytic anemia, idiopathic thrombocytopenia, primary biliary cirrhosis, chronic action hepatitis, ulceratis colitis, Sjogren's syndrome, rheumatic disease, rheumatoid arthritis, polymyositis, scleroderma, mixed connective tissue disease, inflammatory rheumatism, degenerative rheumatism, extra-articular rheumatism, collagen diseases, chronic polyarthritis, psoriasis arthropathica, ankylosing spondylitis, juvenile rheumatoid arthritis, periarthritis humeroscapularis, panarteriitis nodosa, systemic lupus erythematosus, progressive systemic scleroderma, arthritis uratica, dermatomyositis, muscular rheumatism, myositis, myogelosis and chondrocalcinosis, by administering to a subject an effective amount of a soluble CTLA4 molecule L104EA29YIg having the sequence shown in FIG. 19 (SEQ ID NO: 9) beginning with methionine at position +1 or with alanine at position −1 and ending with lysine as position +357, and wherein the effective amount is about 0.1 to 100 mg/kg weight of the subject, about 0.5 to 5 mg/kg weight of a subject, about 5 to 10 mg/kg weight of a subject, about 10 to 15 mg/kg weight of a subject, about 15 to 20 mg/kg weight of a subject, about 20 to 25 mg/kg weight of a subject, about 25 to 30 mg/kg weight of a subject, about 30 to 35 mg/kg weight of a subject, about 35 to 40 mg/kg weight of a subject, about 40 to 45 mg/kg weight of a subject, about 45 to 50 mg/kg weight of a subject, about 50 to 55 mg/kg weight of a subject, about 55 to 60 mg/kg weight of a subject, about 60 to 65 mg/kg weight of a subject, about 65 to 70 mg/kg weight of a subject, about 70 to 75 mg/kg weight of a subject, about 75 to 80 mg/kg weight of a subject, about 80 to 85 mg/kg weight of a subject, about 85 to 90 mg/kg weight of a subject, about 90 to 95 mg/kg weight of a subject, about 95 to 100 mg/kg weight of a subject, about 2 to 10 mg/kg weight of a subject, about 0.1 to 4 mg/kg weight of a subject, about 0.1 to 0.5 mg/kg weight of a subject, about 0.5 to 1.0 mg/kg weight of a subject, about 1.0 to 1.5 mg/kg weight of a subject, about 1.5 to 2.0 mg/kg weight of a subject, about 2.0 to 2.5 mg/kg weight of a subject, about 2.5 to 3.0 mg/kg weight of a subject, about 3.0 to 3.5 mg/kg weight of a subject, about 3.5 to 4.0 mg/kg weight of a subject, about 4.0 to 4.5 mg/kg weight of a subject, about 4.5 to 5.0 mg/kg weight of a subject, about 5.0 to 5.5 mg/kg weight of a subject, about 5.5 to 6.0 mg/kg weight of a subject, about 6.0 to 6.5 mg/kg weight of a subject, about 6.5 to 7.0 mg/kg weight of a subject, about 7.0 to 7.5 mg/kg weight of a subject, about 7.5 to 8.0 mg/kg weight of a subject, about 8.0 to 8.5 mg/kg weight of a subject, about 8.5 to 9.0 mg/kg weight of a subject, about 9.0 to 9.5 mg/kg weight of a subject, about 9.5 to 10.0 mg/kg weight of a subject, about 0.1 to 2 mg/kg weight of a subject, about 2 to 4 mg/kg weight of a subject, about 4 to 6 mg/kg weight of a subject, about 6 to 8 mg/kg weight of a subject, about 8 to 10 mg/kg weight of a subject, about 10 to 12 mg/kg weight of a subject, about 12 to 14 mg/kg weight of a subject, about 14 to 16 mg/kg weight of a subject, about 16 to 18 mg/kg weight of a subject, about 18 to 20 mg/kg weight of a subject, about 0.5 mg/kg weight of the subject, 2 mg/kg weight of the subject, 10 mg/kg weight of the subject, about 0.5 mg/kg to 100 weight of the subject, about 0.5 to 10 mg/kg weight of a subject, about 0.1 to 20 mg/kg weight of a subject, about 500 mg for a subject weighing less than 60 kg, 750 mg for a subject weighing between 60-100 kg or 1000 mg for a subject weighing more than 100 kg.
52. A method for treating a disease selected from graft related disorders, chronic rejection, tissue or cell allo- or xenografts, skin allo- or xenografts, islet allo- or xenografts, muscle allo- or xenografts, hepatocyte allo- or xenografts and neuron allo- or xenografts, by administering to a subject an effective amount of a soluble CTLA4 molecule comprising the sequence shown in FIG. 19 (SEQ ID NO: 9) beginning with methionine at position +1 or with alanine at position −1 and ending with aspartic acid at position +124, and wherein the effective amount is about 0.1 to 100 mg/kg weight of the subject, about 0.5 to 5 mg/kg weight of a subject, about 5 to 10 mg/kg weight of a subject, about 10 to 15 mg/kg weight of a subject, about 15 to 20 mg/kg weight of a subject, about 20 to 25 mg/kg weight of a subject, about 25 to 30 mg/kg weight of a subject, about 30 to 35 mg/kg weight of a subject, about 35 to 40 mg/kg weight of a subject, about 40 to 45 mg/kg weight of a subject, about 45 to 50 mg/kg weight of a subject, about 50 to 55 mg/kg weight of a subject, about 55 to 60 mg/kg weight of a subject, about 60 to 65 mg/kg weight of a subject, about 65 to 70 mg/kg weight of a subject, about 70 to 75 mg/kg weight of a subject, about 75 to 80 mg/kg weight of a subject, about 80 to 85 mg/kg weight of a subject, about 85 to 90 mg/kg weight of a subject, about 90 to 95 mg/kg weight of a subject, about 95 to 100 mg/kg weight of a subject, about 2 to 10 mg/kg weight of a subject, about 0.1 to 4 mg/kg weight of a subject, about 0.1 to 0.5 mg/kg weight of a subject, about 0.5 to 1.0 mg/kg weight of a subject, about 1.0 to 1.5 mg/kg weight of a subject, about 1.5 to 2.0 mg/kg weight of a subject, about 2.0 to 2.5 mg/kg weight of a subject, about 2.5 to 3.0 mg/kg weight of a subject, about 3.0 to 3.5 mg/kg weight of a subject, about 3.5 to 4.0 mg/kg about 4.0 to 4.5 mg/kg weight of a subject, about 4.5 to 5.0 mg/kg weight of a subject, about 5.0 to 5.5 mg/kg weight of a subject, about 5.5 to 6.0 mg/kg weight of a subject, about 6.0 to 6.5 mg/kg weight of a subject, about 6.5 to 7.0 mg/kg weight of a subject, about 7.0 to 7.5 mg/kg weight of a subject, about 7.5 to 8.0 mg/kg weight of a subject, about 8.0 to 8.5 mg/kg weight of a subject, about 8.5 to 9.0 mg/kg weight of a subject, about 9.0 to 9.5 mg/kg weight of a subject, about 9.5 to 10.0 mg/kg weight of a subject, weight of a subject, about 0.1 to 2 mg/kg weight of a subject, about 2 to 4 mg/kg weight of a subject, about 4 to 6 mg/kg weight of a subject, about 6 to 8 mg/kg weight of a subject, about 8 to 10 mg/kg weight of a subject, about 10 to 12 mg/kg weight of a subject, about 12 to 14 mg/kg weight of a subject, about 14 to 16 mg/kg weight of a subject, about 16 to 18 mg/kg weight of a subject, about 18 to 20 mg/kg weight of a subject, about 0.5 mg/kg weight of the subject, 2 mg/kg weight of the subject, 10 mg/kg weight of the subject, about 0.5 mg/kg to 100 weight of the subject, about 0.5 to 10 mg/kg weight of a subject, about 0.1 to 20 mg/kg weight of a subject, about 500 mg for a subject weighing less than 60 kg, 750 mg for a subject weighing between 60-100 kg or 1000 mg for a subject weighing more than 100 kg.
53. A method for treating a disease selected from graft related disorders, chronic rejection, tissue or cell allo- or xenografts, skin allo- or xenografts, islet allo- or xenografts, muscle allo- or xenografts, hepatocyte allo- or xenografts and neuron allo- or xenografts, by administering to a subject an effective amount of a soluble CTLA4 molecule L104EA29YIg having the sequence shown in FIG. 19 (SEQ ID NO: 9) beginning with methionine at position +1 or with alanine at position −1 and ending with lysine as position +357, and wherein the effective amount is about 0.1 to 100 mg/kg weight of the subject, about 0.5 to 5 mg/kg weight of a subject, about 5 to 10 mg/kg weight of a subject, about 10 to 15 mg/kg weight of a subject, about 15 to 20 mg/kg weight of a subject, about 20 to 25 mg/kg weight of a subject, about 25 to 30 mg/kg weight of a subject, about 30 to 35 mg/kg weight of a subject, about 35 to 40 mg/kg weight of a subject, about 40 to 45 mg/kg weight of a subject, about 45 to 50 mg/kg weight of a subject, about 50 to 55 mg/kg weight of a subject, about 55 to 60 mg/kg weight of a subject, about 60 to 65 mg/kg weight of a subject, about 65 to 70 mg/kg weight of a subject, about 70 to 75 mg/kg weight of a subject, about 75 to 80 mg/kg weight of a subject, about 80 to 85 mg/kg weight of a subject, about 85 to 90 mg/kg weight of a subject, about 90 to 95 mg/kg weight of a subject, about 95 to 100 mg/kg weight of a subject, about 2 to 10 mg/kg weight of a subject, about 0.1 to 4 mg/kg weight of a subject, about 0.1 to 0.5 mg/kg weight of a subject, about 0.5 to 1.0 mg/kg weight of a subject, about 1.0 to 1.5 mg/kg weight of a subject, about 1.5 to 2.0 mg/kg weight of a subject, about 2.0 to 2.5 mg/kg weight of a subject, about 2.5 to 3.0 mg/kg weight of a subject, about 3.0 to 3.5 mg/kg weight of a subject, about 3.5 to 4.0 mg/kg weight of a subject, about 4.0 to 4.5 mg/kg weight of a subject, about 4.5 to 5.0 mg/kg weight of a subject, about 5.0 to 5.5 mg/kg weight of a subject, about 5.5 to 6.0 mg/kg weight of a subject, about 6.0 to 6.5 mg/kg weight of a subject, about 6.5 to 7.0 mg/kg weight of a subject, about 7.0 to 7.5 mg/kg weight of a subject, about 7.5 to 8.0 mg/kg weight of a subject, about 8.0 to 8.5 mg/kg weight of a subject, about 8.5 to 9.0 mg/kg weight of a subject, about 9.0 to 9.5 mg/kg weight of a subject, about 9.5 to 10.0 mg/kg weight of a subject, about 0.9 to 2 mg/kg weight of a subject, about 2 to 4 mg/kg weight of a subject, about 4 to 6 mg/kg weight of a subject, about 6 to 8 mg/kg weight of a subject, about 8 to 10 mg/kg weight of a subject, about 10 to 12 mg/kg weight of a subject, about 12 to 14 mg/kg weight of a subject, about 14 to 16 mg/kg weight of a subject, about 16 to 18 mg/kg weight of a subject, about 18 to 20 mg/kg weight of a subject, about 0.5 mg/kg weight of the subject, 2 mg/kg weight of the subject, 10 mg/kg weight of the subject, about 0.5 mg/kg to 100 weight of the subject, about 0.5 to 10 mg/kg weight of a subject, about 0.1 to 20 mg/kg weight of a subject, about 500 mg for a subject weighing less than 60 kg, 750 mg for a subject weighing between 60-100 kg or 1000 mg for a subject weighing more than 100 kg.
54. A method for treating rheumatic disease comprising administering to a subject an effective amount of a soluble CTLA4 molecule L104EA29YIg having the sequence shown in FIG. 19 (SEQ ID NO: 9) beginning with methionine at position +1 or with alanine at position −1 and ending with lysine as position +357, and wherein the effective amount is about 0.1 to 100 mg/kg weight of the subject, about 0.5 to 5 mg/kg weight of a subject, about 5 to 10 mg/kg weight of a subject, about 10 to 15 mg/kg weight of a subject, about 15 to 20 mg/kg weight of a subject, about 20 to 25 mg/kg weight of a subject, about 25 to 30 mg/kg weight of a subject, about 30 to 35 mg/kg weight of a subject, about 35 to 40 mg/kg weight of a subject, about 40 to 45 mg/kg weight of a subject, about 45 to 50 mg/kg weight of a subject, about 50 to 55 mg/kg weight of a subject, about 55 to 60 mg/kg weight of a subject, about 60 to 65 mg/kg weight of a subject, about 65 to 70 mg/kg weight of a subject, about 70 to 75 mg/kg weight of a subject, about 75 to 80 mg/kg weight of a subject, about 80 to 85 mg/kg weight of a subject, about 85 to 90 mg/kg weight of a subject, about 90 to 95 mg/kg weight of a subject, about 95 to 100 mg/kg weight of a subject, about 2 to 10 mg/kg weight of a subject, about 0.1 to 4 mg/kg weight of a subject, about 0.1 to 0.5 mg/kg weight of a subject, about 0.5 to 1.0 mg/kg weight of a subject, about 1.0 to 1.5 mg/kg weight of a subject, about 1.5 to 2.0 mg/kg weight of a subject, about 2.0 to 2.5 mg/kg weight of a subject, about 2.5 to 3.0 mg/kg weight of a subject, about 3.0 to 3.5 mg/kg weight of a subject, about 3.5 to 4.0 mg/kg weight of a subject, about 4.0 to 4.5 mg/kg weight of a subject, about 4.5 to 5.0 mg/kg weight of a subject, about 5.0 to 5.5 mg/kg weight of a subject, about 5.5 to 6.0 mg/kg weight of a subject, about 6.0 to 6.5 mg/kg weight of a subject, about 6.5 to 7.0 mg/kg weight of a subject, about 7.0 to 7.5 mg/kg weight of a subject, about 7.5 to 8.0 mg/kg weight of a subject, about 8.0 to 8.5 mg/kg weight of a subject, about 8.5 to 9.0 mg/kg weight of a subject, about 9.0 to 9.5 mg/kg weight of a subject, about 9.5 to 10.0 mg/kg weight of a subject, about 0.1 to 2 mg/kg weight of a subject, about 2 to 4 mg/kg weight of a subject, about 4 to 6 mg/kg weight of a subject, about 6 to 8 mg/kg weight of a subject, about 8 to 10 mg/kg weight of a subject, about 10 to 12 mg/kg weight of a subject, about 12 to 14 mg/kg weight of a subject, about 14 to 16 mg/kg weight of a subject, about 16 to 18 mg/kg weight of a subject, about 18 to 20 mg/kg weight of a subject, about 0.5 mg/kg weight of the subject, 2 mg/kg weight of the subject, 10 mg/kg weight of the subject, about 0.5 mg/kg to 100 weight of the subject, about 0.5 to 10 mg/kg weight of a subject, about 0.1 to 20 mg/kg weight of a subject, about 500 mg for a subject weighing less than 60 kg, 750 mg for a subject weighing between 60-100 kg or 1000 mg for a subject weighing more than 100 kg.
55. The method of claim 35, 37, or 39, cytokine production is regulated.
56. The method of claim 35, 37, or 39, wherein improves ACR 20, 50 and/or 70 response rates is improved.
57. The method of claim 29, 30, 31, 34, 35, 36, 37, 38, 39, 40, 50, 51, 52, 53 or 54, wherein the soluble CTLA4 molecule comprises an amino acid sequence which permits secretion of the soluble CTLA4 molecule.
58. The method of claim 57, wherein the amino acid sequence which permits secretion comprises an oncostatin M signal peptide.
59. The method of claim 1, 2, 5, 6, 29, 30, 31, 34, 35, 36, 37, 38, 39, 40, 50, 51, 52, 53 or 54, wherein administration of the agents is effected locally or systemically.
60. The method of claim 59, wherein administration is selected from the group consisting of, intravenous, intramuscular, intraperitoneal, oral, inhalation, subcutaneous, implantable pump, continuous infusion, gene therapy, liposomes, suppositories, topical contact, vesicles, capsules, biodegradable polymers, hydrogels, controlled release patch and injection methods.
61. The method of claim 1, 2, 5, 6, 29, 30, 31, 34, 35, 36, 37, 38, 39, 40, 50, 51, 52, 53 or 54, wherein the subject is selected from the group consisting of human, monkey, ape, dog, cat, cow, horse, rabbit, mouse, and rat.
62. The method of claim 22 or 29, wherein the CTLA4Ig is encoded by a nucleic acid molecule designated ATCC No. 68629.
63. The method of claim 28, 32, 51, 53 or 54, wherein L104EA29YIg is encoded by a DNA sequence designated ATCC PTA-2104.
64. A pharmaceutical composition comprising a pharmaceutically acceptable carrier and an effective amount of a first agent and a second agent, wherein
a) the first agent is soluble CTLA4, and
b) the second agent is selected from a group consisting of immunosuppressive agents, corticosteroids, nonsteroidal antiinflammatory drugs, prednisone, azathioprine, methotrexate, TNFα blockers or antagonists, infliximab, any biological agent targeting an inflammatory cytokine, hydroxychloroquine, sulphasalazopryine, gold salts, etanercept, anakinra, cyclophosphamide, leflunomide, collagen, dnaJ, a molecule that blocks TNF receptors, pegsunercept, a molecule that blocks cytokine function, AMG719, a molecule that blocks LFA-1 function, efalizumab, acetyl salicylic acid, choline magnesium salicylate, diflunisal, magnesium salicylate, salsalate, sodium salicylate, diclofenac, etodolac, fenoprofen, flurbiprofen, indomethacin, ketoprofen, ketorolac, meclofenamate, naproxen, nabumetone, phenylbutazone, piroxicam, sulindac, tolmetin, acetaminophen, ibuprofen, Cox-2 inhibitors, meloxicam, codeine phosphate, propoxyphene napsylate, oxycodone hydrochloride, oxycodone bitartrate and tramadol.
65. The pharmaceutical composition of claim 64, wherein the soluble CTLA4 comprises the extracellular domain of a CTLA4 molecule, or portion thereof, which binds a B7 antigen expressed on activated B cells.
66. The pharmaceutical composition of claim 64, wherein the soluble CTLA4 is a CTLA4 fusion molecule.
67. The pharmaceutical composition of claim 66, wherein the CTLA4 fusion molecule comprises an extracellular domain of a CTLA4 molecule which binds a B7 antigen expressed on activated B cells joined to a non-CTLA4 molecule.
68. The pharmaceutical composition of claim 64, wherein the soluble CTLA4 is a soluble CTLA4 mutant molecule.
69. The pharmaceutical composition of claim 68, wherein the soluble CTLA4 mutant molecule comprises an extracellular domain of CTLA4,
a) wherein the extracellular domain of CTLA4 begins with methionine at position +1 or with alanine at position −1 and ends with aspartic acid at position +124 as shown in FIG. 23 or 24 (SEQ ID NO: 17 or 19), and
b) wherein at position +104 of the extracellular domain of CTLA4, leucine is substituted with any other amino acid.
70. The pharmaceutical composition of claim 68, wherein the soluble CTLA4 mutant molecule comprises an extracellular domain of CTLA4,
a) wherein the extracellular domain of CTLA4 begins with methionine at position +1 or with alanine at position −1 and ends with aspartic acid at position +124 as shown in FIG. 23 or 24 (SEQ ID NO: 17 or 19),
b) wherein at position +104 of the extracellular domain of CTLA4, leucine is substituted with glutamic acid, and
c) wherein at position +29 of the extracellular domain of CTLA4, alanine is substituted with tyrosine.
71. The pharmaceutical composition of claim 68, wherein the soluble CTLA4 mutant molecule is L104EA29YIg as shown in FIG. 19 (SEQ ID NO: 9), beginning with methionine at position +1 or with alanine at position −1 and ending with lysine at position +357.
72. The pharmaceutical composition of claim 64, wherein the pharmaceutically acceptable carrier is selected ion exchangers, alumina, aluminum stearate, lecithin, serum proteins, such as human serum albumin, buffer substances, glycine, sorbic acid, potassium sorbate, partial glyceride mixtures of saturated vegetable fatty acids, phosphate buffered saline solution, water, emulsions, salts or electrolytes, colloidal silica, magnesium trisilicate, polyvinyl pyrrolidone, cellulose-based substances, polyethylene glycol, sterile solutions, tablets, excipients, sucrose, glucose, maltose, flavor and color additives, lipid compositions and polymeric compositions.
73. Combination of a pharmaceutical composition comprising a soluble CTLA4 with a pharmaceutical composition comprising an agent selected from a group consisting of immunosuppressive agents, corticosteroids, nonsteroidal antiinflammatory drugs, prednisone, azathioprine, methotrexate, TNFα blockers or antagonists, infliximab, any biological agent targeting an inflammatory cytokine, hydroxychloroquine, sulphasalazopryine, gold salts, etanercept, anakinra, cyclophosphamide, leflunomide, collagen, dnaJ, a molecule that blocks TNF receptors, pegsunercept, a molecule that blocks cytokine function, AMG719, a molecule that blocks LFA-1 function, efalizumab, acetyl salicylic acid, choline magnesium salicylate, diflunisal, magnesium salicylate, salsalate, sodium salicylate, diclofenac, etodolac, fenoprofen, flurbiprofen, indomethacin, ketoprofen, ketorolac, meclofenamate, naproxen, nabumetone, phenylbutazone, piroxicam, sulindac, tolmetin, acetaminophen, ibuprofen, Cox-2 inhibitors, meloxicam, codeine phosphate, propoxyphene napsylate, oxycodone hydrochloride, oxycodone bitartrate and tramadol, for use in therapy.
74. A kit comprising an effective amount of a first agent and a second agent, wherein
a) the first agent is soluble CTLA4, and
b) the second agent is selected from a group consisting of immunosuppressive agents, corticosteroids, nonsteroidal antiinflammatory drugs, prednisone, azathioprine, methotrexate, TNFα blockers or antagonists, infliximab, any biological agent targeting an inflammatory cytokine, hydroxychloroquine, sulphasalazopryine, gold salts, etanercept, anakinra, cyclophosphamide, leflunomide, corticosteroids, nonsteroidal antiinflammatory drugs, prednisone, azathioprine, methotrexate, TNFα blockers or antagonists, infliximab, any biological agent targeting an inflammatory cytokine, hydroxychloroquine, sulphasalazopryine, gold salts, etanercept, anakinra, cyclophosphamide, leflunomide, collagen, dnaJ, a molecule that blocks TNF receptors, pegsunercept, a molecule that blocks cytokine function, AMG719, a molecule that blocks LFA-1 function, efalizumab, acetyl salicylic acid, choline magnesium salicylate, diflunisal, magnesium salicylate, salsalate, sodium salicylate, diclofenac, etodolac, fenoprofen, flurbiprofen, indomethacin, ketoprofen, ketorolac, meclofenamate, naproxen, nabumetone, phenylbutazone, piroxicam, sulindac, tolmetin, acetaminophen, ibuprofen, Cox-2 inhibitors, meloxicam, codeine phosphate, propoxyphene napsylate, oxycodone hydrochloride, oxycodone bitartrate and tramadol.
75. The kit of claim 74, further comprising a label indicating that the first and second agents are useful to treat an immune system disease.
76. The kit of claim 75, wherein the label indicates an effective amount for the first agent being about 0.1 to 100 mg/kg weight of the subject, about 0.5 to 5 mg/kg weight of a subject, about 5 to 10 mg/kg weight of a subject, about 10 to 15 mg/kg weight of a subject, about 15 to 20 mg/kg weight of a subject, about 20 to 25 mg/kg weight of a subject, about 25 to 30 mg/kg weight of a subject, about 30 to 35 mg/kg weight of a subject, about 35 to 40 mg/kg weight of a subject, about 40 to 45 mg/kg weight of a subject, about 45 to 50 mg/kg weight of a subject, about 50 to 55 mg/kg weight of a subject, about 55 to 60 mg/kg weight of a subject, about 60 to 65 mg/kg weight of a subject, about 65 to 70 mg/kg weight of a subject, about 70 to 75 mg/kg weight of a subject, about 75 to 80 mg/kg weight of a subject, about 80 to 85 mg/kg weight of a subject, about 85 to 90 mg/kg weight of a subject, about 90 to 95 mg/kg weight of a subject, about 95 to 100 mg/kg weight of a subject, about 2 to 10 mg/kg weight of a subject, about 0.1 to 4 mg/kg weight of a subject, about 0.1 to 0.5 mg/kg weight of a subject, about 0.5 to 1.0 mg/kg weight of a subject, about 1.0 to 1.5 mg/kg weight of a subject, about 1.5 to 2.0 mg/kg weight of a subject, about 2.0 to 2.5 mg/kg weight of a subject, about 2.5 to 3.0 mg/kg weight of a subject, about 3.0 to 3.5 mg/kg weight of a subject, about 3.5 to 4.0 mg/kg weight of a subject, about 4.0 to 4.5 mg/kg weight of a subject, about 4.5 to 5.0 mg/kg weight of a subject, about 5.0 to 5.5 mg/kg weight of a subject, about 5.5 to 6.0 mg/kg weight of a subject, about 6.0 to 6.5 mg/kg weight of a subject, about 6.5 to 7.0 mg/kg weight of a subject, about 7.0 to 7.5 mg/kg weight of a subject, about 7.5 to 8.0 mg/kg weight of a subject, about 8.0 to 8.5 mg/kg weight of a subject, about 8.5 to 9.0 mg/kg weight of a subject, about 9.0 to 9.5 mg/kg weight of a subject, about 9.5 to 10.0 mg/kg weight of a subject, about 0.1 to 2 mg/kg weight of a subject, about 2 to 4 mg/kg weight of a subject, about 4 to 6 mg/kg weight of a subject, about 6 to 8 mg/kg weight of a subject, about 8 to 10 mg/kg weight of a subject, about 10 to 12 mg/kg weight of a subject, about 12 to 14 mg/kg weight of a subject, about 14 to 16 mg/kg weight of a subject, about 16 to 18 mg/kg weight of a subject, about 18 to 20 mg/kg weight of a subject, about 500 mg for a subject weighing less than 60 kg, 750 mg for a subject weighing between 60-100 kg or 1000 mg for a subject weighing more than 100 kg.
77. The kit of claim 75, wherein the label indicates an effective amount for the second agent being about 0.1 to 40 mg per week, about 5 to 30 mg per week, about 0.1 to 5 mg/week, about 5 to 10 mg/week, about 10 to 15 mg/week, about 15 to 20 mg/week, about 20 to 25 mg/week, about 25 to 30 mg/week, about 30 to 35 mg/week, about 35 to 40 mg/week, about 10 to 30 mg/week, about 10 to 100 mg/week, about 50 mg/week, about 0.1 to 50 mg/kg body weight per week, about 1 to about 5000 mg/day, about 1 to 10 mg/day, about 10 to 50 mg/day, about 50 to 100 mg/day, about 100 to 150 mg/day, about 150 to 200 mg/day, about 200 to 250 mg/day, about 250 to 300 mg/day, about 300 to 350 mg/day, about 350 to 400 mg/day, about 400 to 450 mg/day, about 450 to 500 mg/day, about 500 to 550 mg/day, about 550 to 600 mg/day, about 600 to 650 mg/day, about 650 to 700 mg/day, about 700 to 750 mg/day, about 750 to 800 mg/day, about 800 to 850 mg/day, about 850 to 900 mg/day, about 900 to 950 mg/day, about 950 to 1000 mg/day, about 1000 to 1100 mg/day, about 1100 to 1200 mg/day, about 1200 to 1300 mg/day, about 1300 to 1400 mg/day, about 1400 to 1500 mg/day, about 1500 to 1600 mg/day, about 1600 to 1700 mg/day, about 1700 to 1800 mg/day, about 1800 to 1900 mg/day, about 1900 to 2000 mg/day, about 2000 to 2500 mg/day, about 2500 to 3000 mg/day, about 3000 to 3500 mg/day, about 3500 to 4000 mg/day, about 4000 to 4500 mg/day or about 4500 to 5000 mg/day.
78. The kit of claim 75, wherein the label indicates an effective amount for the first and second agent
a. the effective amount of the first agent being about 0.1 to 100 mg/kg weight of the subject, about 0.5 to 5 mg/kg weight of a subject, about 5 to 10 mg/kg weight of a subject, about 10 to 15 mg/kg weight of a subject, about 15 to 20 mg/kg weight of a subject, about 20 to 25 mg/kg weight of a subject, about 25 to 30 mg/kg weight of a subject, about 30 to 35 mg/kg weight of a subject, about 35 to 40 mg/kg weight of a subject, about 40 to 45 mg/kg weight of a subject, about 45 to 50 mg/kg weight of a subject, about 50 to 55 mg/kg weight of a subject, about 55 to 60 mg/kg weight of a subject, about 60 to 65 mg/kg weight of a subject, about 65 to 70 mg/kg weight of a subject, about 70 to 75 mg/kg weight of a subject, about 75 to 80 mg/kg weight of a subject, about 80 to 85 mg/kg weight of a subject, about 85 to 90 mg/kg weight of a subject, about 90 to 95 mg/kg weight of a subject, about 95 to 100 mg/kg weight of a subject, about 2 to 10 mg/kg weight of a subject, about 0.1 to 4 mg/kg weight of a subject, about 0.1 to 0.5 mg/kg weight of a subject, about 0.5 to 1.0 mg/kg weight of a subject, about 1.0 to 1.5 mg/kg weight of a subject, about 1.5 to 2.0 mg/kg weight of a subject, about 2.0 to 2.5 mg/kg weight of a subject, about 2.5 to 3.0 mg/kg weight of a subject, about 3.0 to 3.5 mg/kg weight of a subject, about 3.5 to 4.0 mg/kg weight of a subject, about 4.0 to 4.5 mg/kg weight of a subject, about 4.5 to 5.0 mg/kg weight of a subject, about 5.0 to 5.5 mg/kg weight of a subject, about 5.5 to 6.0 mg/kg weight of a subject, about 6.0 to 6.5 mg/kg weight of a subject, about 6.5 to 7.0 mg/kg weight of a subject, about 7.0 to 7.5 mg/kg weight of a subject, about 7.5 to 8.0 mg/kg weight of a subject, about 8.0 to 8.5 mg/kg weight of a subject, about 8.5 to 9.0 mg/kg weight of a subject, about 9.0 to 9.5 mg/kg weight of a subject, about 9.5 to 10.0 mg/kg weight of a subject, about 0.1 to 2 mg/kg weight of a subject, about 2 to 4 mg/kg weight of a subject, about 4 to 6 mg/kg weight of a subject, about 6 to 8 mg/kg weight of a subject, about 8 to 10 mg/kg weight of a subject, about 10 to 12 mg/kg weight of a subject, about 12 to 14 mg/kg weight of a subject, about 14 to 16 mg/kg weight of a subject, about 16 to 18 mg/kg weight of a subject, about 18 to 20 mg/kg weight of a subject, about 500 mg for a subject weighing less than 60 kg, 750 mg for a subject weighing between 60-100 kg or 1000 mg for a subject weighing more than 100 kg, and
b. the effective amount of the second agent is about 0.1 to 40 mg per week, about 5 to 30 mg per week, about 0.1 to 5 mg/week, about 5 to 10 mg/week, about 10 to 15 mg/week, about 15 to 20 mg/week, about 20 to 25 mg/week, about 25 to 30 mg/week, about 30 to 35 mg/week, about 35 to 40 mg/week, about 10 to 30 mg/week, about 10 to 100 mg/week, about 50 mg/week, about 0.1 to 50 mg/kg body weight per week, about 1 to about 5000 mg/day, about 1 to 10 mg/day, about 10 to 50 mg/day, about 50 to 100 mg/day, about 100 to 150 mg/day, about 150 to 200 mg/day, about 200 to 250 mg/day, about 250 to 300 mg/day, about 300 to 350 mg/day, about 350 to 400 mg/day, about 400 to 450 mg/day, about 450 to 500 mg/day, about 500 to 550 mg/day, about 550 to 600 mg/day, about 600 to 650 mg/day, about 650 to 700 mg/day, about 700 to 750 mg/day, about 750 to 800 mg/day, about 800 to 850 mg/day, about 850 to 900 mg/day, about 900 to 950 mg/day, about 950 to 1000 mg/day, about 1000 to 1100 mg/day, about 1100 to 1200 mg/day, about 1200 to 1300 mg/day, about 1300 to 1400 mg/day, about 1400 to 1500 mg/day, about 1500 to 1600 mg/day, about 1600 to 1700 mg/day, about 1700 to 1800 mg/day, about 1800 to 1900 mg/day, about 1900 to 2000 mg/day, about 2000 to 2500 mg/day, about 2500 to 3000 mg/day, about 3000 to 3500 mg/day, about 3500 to 4000 mg/day, about 4000 to 4500 mg/day or about 4500 to 5000 mg/day.
79. The kit of claim 75, wherein the immune system disease is selected from autoimmune diseases, psoriasis, immune disorders associated with graft transplantation rejection, T cell lymphoma, T cell acute lymphoblastic leukemia, testicular angiocentric T cell lymphoma, benign lymphocytic angiitis, lupus erythematosus, Hashimoto's thyroiditis, primary myxedema, Graves' disease, pernicious anemia, autoimmune atrophic gastritis, Addison's disease, insulin dependent diabetes mellitis, good pasture's syndrome, myasthenia gravis, pemphigus, Crohn's disease, sympathetic ophthalmia, autoimmune uveitis, multiple sclerosis, autoimmune hemolytic anemia, idiopathic thrombocytopenia, primary biliary cirrhosis, chronic action hepatitis, ulceratis colitis, Sjogren's syndrome, rheumatic disease, rheumatoid arthritis, polymyositis, scleroderma, mixed connective tissue disease, inflammatory rheumatism, degenerative rheumatism, extra-articular rheumatism, collagen diseases, chronic polyarthritis, psoriasis arthropathica, ankylosing spondylitis, juvenile rheumatoid arthritis, periarthritis humeroscapularis, panarteriitis nodosa, systemic lupus erythematosus, progressive systemic scleroderma, arthritis uratica, dermatomyositis, muscular rheumatism, myositis, myogelosis and chondrocalcinosis.
80. The kit of claim 74, further comprising a container.
81. The kit of claim 74, wherein the soluble CTLA4 comprises the extracellular domain of a CTLA4 molecule, or portion thereof, which binds a B7 antigen expressed on activated B cells.
82. The kit of claim 74, wherein the soluble CTLA4 comprises the extracellular domain of a CTLA4 molecule as shown in FIG. 23 (SEQ ID NO: 17) beginning with methionine at position +1 or with alanine at position −1 and ending with aspartic acid at position +124.
83. The kit of claim 74, wherein the soluble CTLA4 is a CTLA4 fusion molecule.
84. The kit of claim 83, wherein the CTLA4 fusion molecule comprises an extracellular domain of a CTLA4 molecule which binds a B7 antigen expressed on activated B cells joined to a non-CTLA4 molecule.
85. The kit of claim 84, wherein the non-CTLA4 molecule comprises an amino acid sequence which alters the solubility or affinity which comprises an immunoglobulin moiety.
86. The kit of claim 85, wherein the immunoglubulin moiety is an immunoglubulin constant region or portion thereof.
87. The kit of claim 86, wherein the immunoglubulin constant region or portion thereof is mutated to reduce effector function.
88. The kit of claim 83, wherein the CTLA4 fusion molecule is CTLA4Ig.
89. The kit of claim 88, wherein the CTLA4Ig is shown in FIG. 24 (SEQ ID NO:19) beginning with methionine at position +1 or with alanine at position −1 and ending with lysine at position +357.
90. The kit of claim 74, wherein the soluble CTLA4 is a soluble CTLA4 mutant molecule.
91. The kit of claim 90, wherein the soluble CTLA4 mutant molecule comprises an extracellular domain of CTLA4,
a) wherein the extracellular domain of CTLA4 begins with methionine at position +1 or with alanine at position −1 and ends with aspartic acid at position +124 as shown in FIG. 23 or 24 (SEQ ID NO: 17 or 19), and
b) wherein at position +104 of the extracellular domain of CTLA4, leucine is substituted with any other amino acid.
92. The kit of claim 90, wherein the soluble CTLA4 mutant molecule comprises an extracellular domain of CTLA4,
a) wherein the extracellular domain of CTLA4 begins with methionine at position +1 or with alanine at position −1 and ends with aspartic acid at position +124 as shown in FIG. 23 or 24 (SEQ ID NO: 17 or 19),
b) wherein at position +104 of the extracellular domain of CTLA4, leucine is substituted with glutamic acid, and
c) wherein at position +29 of the extracellular domain of CTLA4, alanine is substituted with tyrosine.
93. The kit of claim 90, wherein the soluble CTLA4 mutant molecule is L104EA29YIg as shown in FIG. 19 (SEQ. ID NO: 9) beginning with methionine at position +1 or with alanine at position −1 and ending with lysine at position +357 as shown in FIG. 19 (SEQ ID NO: 9).
94. Use of a kit comprising the pharmaceutical composition of claim 68, a container and a label.
95. A kit comprising an effective amount of a first agent and a label, wherein
a) the first agent is soluble CTLA4, and
b) the label indicates that the first agent can be used with a second agent selected from a group consisting of immunosuppressive agents, corticosteroids, nonsteroidal antiinflammatory drugs, prednisone, azathioprine, methotrexate, TNFα blockers or antagonists, infliximab, any biological agent targeting an inflammatory cytokine, hydroxychloroquine, sulphasalazopryine, gold salts, etanercept, anakinra, cyclophosphamide, leflunomide, collagen, dnaJ, a molecule that blocks TNF receptors, pegsunercept, a molecule that blocks cytokine function, AMG719, a molecule that blocks LFA-1 function, efalizumab, acetyl salicylic acid, choline magnesium salicylate, diflunisal, magnesium salicylate, salsalate, sodium salicylate, diclofenac, etodolac, fenoprofen, flurbiprofen, indomethacin, ketoprofen, ketorolac, meclofenamate, naproxen, nabumetone, phenylbutazone, piroxicam, sulindac, tolmetin, acetaminophen, ibuprofen, Cox-2 inhibitors, meloxicam, codeine phosphate, propoxyphene napsylate, oxycodone hydrochloride, oxycodone bitartrate and tramadol.
96. The kit of claim 95, wherein the label further indicates an effective amount for the first agent being about 0.1 to 100 mg/kg weight of the subject, about 0.5 to 5 mg/kg weight of a subject, about 5 to 10 mg/kg weight of a subject, about 10 to 15 mg/kg weight of a subject, about 15 to 20 mg/kg weight of a subject, about 20 to 25 mg/kg weight of a subject, about 25 to 30 mg/kg weight of a subject, about 30 to 35 mg/kg weight of a subject, about 35 to 40 mg/kg weight of a subject, about 40 to 45 mg/kg weight of a subject, about 45 to 50 mg/kg weight of a subject, about 50 to 55 mg/kg weight of a subject, about 55 to 60 mg/kg weight of a subject, about 60 to 65 mg/kg weight of a subject, about 65 to 70 mg/kg weight of a subject, about 70 to 75 mg/kg weight of a subject, about 75 to 80 mg/kg weight of a subject, about 80 to 85 mg/kg weight of a subject, about 85 to 90 mg/kg weight of a subject, about 90 to 95 mg/kg weight of a subject, about 95 to 100 mg/kg weight of a subject, about 2 to 10 mg/kg weight of a subject, about 0.1 to 4 mg/kg weight of a subject, about 0.1 to 0.5 mg/kg weight of a subject, about 0.5 to 1.0 mg/kg weight of a subject, about 1.0 to 1.5 mg/kg weight of a subject, about 1.5 to 2.0 mg/kg weight of a subject, about 2.0 to 2.5 mg/kg weight of a subject, about 2.5 to 3.0 mg/kg weight of a subject, about 3.0 to 3.5 mg/kg weight of a subject, about 3.5 to 4.0 mg/kg weight of a subject, about 4.0 to 4.5 mg/kg weight of a subject, about 4.5 to 5.0 mg/kg weight of a subject, about 5.0 to 5.5 mg/kg weight of a subject, about 5.5 to 6.0 mg/kg weight of a subject, about 6.0 to 6.5 mg/kg weight of a subject, about 6.5 to 7.0 mg/kg weight of a subject, about 7.0 to 7.5 mg/kg weight of a subject, about 7.5 to 8.0 mg/kg weight of a subject, about 8.0 to 8.5 mg/kg weight of a subject, about 8.5 to 9.0 mg/kg weight of a subject, about 9.0 to 9.5 mg/kg weight of a subject, about 9.5 to 10.0 mg/kg weight of a subject, about 0.1 to 2 mg/kg weight of a subject, about 2 to 4 mg/kg weight of a subject, about 4 to 6 mg/kg weight of a subject, about 6 to 8 mg/kg weight of a subject, about 8 to 10 mg/kg weight of a subject, about 10 to 12 mg/kg weight of a subject, about 12 to 14 mg/kg weight of a subject, about 14 to 16 mg/kg weight of a subject, about 16 to 18 mg/kg weight of a subject, about 18 to 20 mg/kg weight of a subject, about 500 mg for a subject weighing less than 60 kg, 750 mg for a subject weighing between 60-100 kg or 1000 mg for a subject weighing more than 100 kg.
97. The kit of claim 95, wherein the label indicates an effective amount for the second agent being about 0.1 to 40 mg per week, about 5 to 30 mg per week, about 0.1 to 5 mg/week, about 5 to 10 mg/week, about 10 to 15 mg/week, about 15 to 20 mg/week, about 20 to 25 mg/week, about 25 to 30 mg/week, about 30 to 35 mg/week, about 35 to 40 mg/week, about 10 to 30 mg/week, about 10 to 100 mg/week, about 50 mg/week, about 0.1 to 50 mg/kg body weight per week, about 1 to about 5000 mg/day, about 1 to 10 mg/day, about 10 to 50 mg/day, about 50 to 100 mg/day, about 100 to 150 mg/day, about 150 to 200 mg/day, about 200 to 250 mg/day, about 250 to 300 mg/day, about 300 to 350 mg/day, about 350 to 400 mg/day, about 400 to 450 mg/day, about 450 to 500 mg/day, about 500 to 550 mg/day, about 550 to 600 mg/day, about 600 to 650 mg/day, about 650 to 700 mg/day, about 700 to 750 mg/day, about 750 to 800 mg/day, about 800 to 850 mg/day, about 850 to 900 mg/day, about 900 to 950 mg/day, about 950 to 1000 mg/day, about 1000 to 1100 mg/day, about 1100 to 1200 mg/day, about 1200 to 1300 mg/day, about 1300 to 1400 mg/day, about 1400 to 1500 mg/day, about 1500 to 1600 mg/day, about 1600 to 1700 mg/day, about 1700 to 1800 mg/day, about 1800 to 1900 mg/day, about 1900 to 2000 mg/day, about 2000 to 2500 mg/day, about 2500 to 3000 mg/day, about 3000 to 3500 mg/day, about 3500 to 4000 mg/day, about 4000 to 4500 mg/day or about 4500 to 5000 mg/day.
98. The kit of claim 95, wherein the label indicates an effective amount for the first and second agent,
a. wherein the effective amount of the first agent being about 0.1 to 100 mg/kg weight of the subject, about 0.5 to 5 mg/kg weight of a subject, about 5 to 10 mg/kg weight of a subject, about 10 to 15 mg/kg weight of a subject, about 15 to 20 mg/kg weight of a subject, about 20 to 25 mg/kg weight of a subject, about 25 to 30 mg/kg weight of a subject, about 30 to 35 mg/kg weight of a subject, about 35 to 40 mg/kg weight of a subject, about 40 to 45 mg/kg weight of a subject, about 45 to 50 mg/kg weight of a subject, about 50 to 55 mg/kg weight of a subject, about 55 to 60 mg/kg weight of a subject, about 60 to 65 mg/kg weight of a subject, about 65 to 70 mg/kg weight of a subject, about 70 to 75 mg/kg weight of a subject, about 75 to 80 mg/kg weight of a subject, about 80 to 85 mg/kg weight of a subject, about 85 to 90 mg/kg weight of a subject, about 90 to 95 mg/kg weight of a subject, about 95 to 100 mg/kg weight of a subject, about 2 to 10 mg/kg weight of a subject, about 0.1 to 4 mg/kg weight of a subject, about 0.1 to 0.5 mg/kg weight of a subject, about 0.5 to 1.0 mg/kg weight of a subject, about 1.0 to 1.5 mg/kg weight of a subject, about 1.5 to 2.0 mg/kg weight of a subject, about 2.0 to 2.5 mg/kg weight of a subject, about 2.5 to 3.0 mg/kg weight of a subject, about 3.0 to 3.5 mg/kg weight of a subject, about 3.5 to 4.0 mg/kg weight of a subject, about 4.0 to 4.5 mg/kg weight of a subject, about 4.5 to 5.0 mg/kg weight of a subject, about 5.0 to 5.5 mg/kg weight of a subject, about 5.5 to 6.0 mg/kg weight of a subject, about 6.0 to 6.5 mg/kg weight of a subject, about 6.5 to 7.0 mg/kg weight of a subject, about 7.0 to 7.5 mg/kg weight of a subject, about 7.5 to 8.0 mg/kg weight of a subject, about 8.0 to 8.5 mg/kg weight of a subject, about 8.5 to 9.0 mg/kg weight of a subject, about 9.0 to 9.5 mg/kg weight of a subject, about 9.5 to 10.0 mg/kg weight of a subject, about 0.1 to 2 mg/kg weight of a subject, about 2 to 4 mg/kg weight of a subject, about 4 to 6 mg/kg weight of a subject, about 6 to 8 mg/kg weight of a subject, about 8 to 10 mg/kg weight of a subject, about 10 to 12 mg/kg weight of a subject, about 12 to 14 mg/kg weight of a subject, about 14 to 16 mg/kg weight of a subject, about 16 to 18 mg/kg weight of a subject, about 18 to 20 mg/kg weight of a subject, about 500 mg for a subject weighing less than 60 kg, 750 mg for a subject weighing between 60-100 kg or 1000 mg for a subject weighing more than 100 kg, and
b. wherein the effective amount of the second agent is about 0.1 to 40 mg per week, about 5 to 30 mg per week, about 0.1 to 5 mg/week, about 5 to 10 mg/week, about 10 to 15 mg/week, about 15 to 20 mg/week, about 20 to 25 mg/week, about 25 to 30 mg/week, about 30 to 35 mg/week, about 35 to 40 mg/week, about 10 to 30 mg/week, about 10 to 100 mg/week, about 50 mg/week, about 0.1 to 50 mg/kg body weight per week, about 1 to about 5000 mg/day, about 1 to 10 mg/day, about 10 to 50 mg/day, about 50 to 100 mg/day, about 100 to 150 mg/day, about 150 to 200 mg/day, about 200 to 250 mg/day, about 250 to 300 mg/day, about 300 to 350 mg/day, about 350 to 400 mg/day, about 400 to 450 mg/day, about 450 to 500 mg/day, about 500 to 550 mg/day, about 550 to 600 mg/day, about 600 to 650 mg/day, about 650 to 700 mg/day, about 700 to 750 mg/day, about 750 to 800 mg/day, about 800 to 850 mg/day, about 850 to 900 mg/day, about 900 to 950 mg/day, about 950 to 1000 mg/day, about 1000 to 1100 mg/day, about 1100 to 1200 mg/day, about 1200 to 1300 mg/day, about 1300 to 1400 mg/day, about 1400 to 1500 mg/day, about 1500 to 1600 mg/day, about 1600 to 1700 mg/day, about 1700 to 1800 mg/day, about 1800 to 1900 mg/day, about 1900 to 2000 mg/day, about 2000 to 2500 mg/day, about 2500 to 3000 mg/day, about 3000 to 3500 mg/day, about 3500 to 4000 mg/day, about 4000 to 4500 mg/day or about 4500 to 5000 mg/day.
99. The kit of claim 95, further comprising a label indicating that the first and second agents are useful to treat an immune system disease.
100. The kit of claim 99, wherein the immune system disease is selected from autoimmune diseases, psoriasis, immune disorders associated with graft transplantation rejection, T cell lymphoma, T cell acute lymphoblastic leukemia, testicular angiocentric T cell lymphoma, benign lymphocytic angiitis, lupus erythematosus, Hashimoto's thyroiditis, primary myxedema, Graves' disease, pernicious anemia, autoimmune atrophic gastritis, Addison's disease, insulin dependent diabetes mellitis, good pasture's syndrome, myasthenia gravis, pemphigus, Crohn's disease, sympathetic ophthalmia, autoimmune uveitis, multiple sclerosis, autoimmune hemolytic anemia, idiopathic thrombocytopenia, primary biliary cirrhosis, chronic action hepatitis, ulceratis colitis, Sjogren's syndrome, rheumatic disease, rheumatoid arthritis, polymyositis, scleroderma, mixed connective tissue disease, inflammatory rheumatism, degenerative rheumatism, extra-articular rheumatism, collagen diseases, chronic polyarthritis, psoriasis arthropathica, ankylosing spondylitis, juvenile rheumatoid arthritis, periarthritis humeroscapularis, panarteriitis nodosa, systemic lupus erythematosus, progressive systemic sclerodermia, arthritis uratica, dermatomyositis, muscular rheumatism, myositis, myogelosis and chondrocalcinosis.
101. A kit comprising an effective amount of a first agent and a label, wherein
a) the first agent is soluble CTLA4, and
b) the label indicates that the first agent can be used with a second agent selected from a group consisting of immunosuppressive agents, corticosteroids, nonsteroidal antiinflammatory drugs, prednisone, azathioprine, methotrexate, TNFα blockers or antagonists, infliximab, any biological agent targeting an inflammatory cytokine, hydroxychloroquine, sulphasalazopryine, gold salts, etanercept, anakinra, cyclophosphamide, leflunomide, collagen, dnaJ, a molecule that blocks TNF receptors, pegsunercept, a molecule that blocks cytokine function, AMG719, a molecule that blocks LFA-1 function, efalizumab, acetyl salicylic acid, choline magnesium salicylate, diflunisal, magnesium salicylate, salsalate, sodium salicylate, diclofenac, etodolac, fenoprofen, flurbiprofen, indomethacin, ketoprofen, ketorolac, meclofenamate, naproxen, nabumetone, phenylbutazone, piroxicam, sulindac, tolmetin, acetaminophen, ibuprofen, Cox-2 inhibitors, meloxicam, codeine phosphate, propoxyphene napsylate, oxycodone hydrochloride, oxycodone bitartrate and tramadol,
wherein the label further indicates an effective amount for the first agent being about 0.1 to 100 mg/kg weight of the subject, about 0.5 to 5 mg/kg weight of a subject, about 5 to 10 mg/kg weight of a subject, about 10 to 15 mg/kg weight of a subject, about 15 to 20 mg/kg weight of a subject, about 20 to 25 mg/kg weight of a subject, about 25 to 30 mg/kg weight of a subject, about 30 to 35 mg/kg weight of a subject, about 35 to 40 mg/kg weight of a subject, about 40 to 45 mg/kg weight of a subject, about 45 to 50 mg/kg weight of a subject, about 50 to 55 mg/kg weight of a subject, about 55 to 60 mg/kg weight of a subject, about 60 to 65 mg/kg weight of a subject, about 65 to 70 mg/kg weight of a subject, about 70 to 75 mg/kg weight of a subject, about 75 to 80 mg/kg weight of a subject, about 80 to 85 mg/kg weight of a subject, about 85 to 90 mg/kg weight of a subject, about 90 to 95 mg/kg weight of a subject, about 95 to 100 mg/kg weight of a subject, about 2 to 10 mg/kg weight of a subject, about 0.1 to 4 mg/kg weight of a subject, about 0.1 to 0.5 mg/kg weight of a subject, about 0.5 to 1.0 mg/kg weight of a subject, about 1.0 to 1.5 mg/kg weight of a subject, about 1.5 to 2.0 mg/kg weight of a subject, about 2.0 to 2.5 mg/kg weight of a subject, about 2.5 to 3.0 mg/kg weight of a subject, about 3.0 to 3.5 mg/kg weight of a subject, about 3.5 to 4.0 mg/kg weight of a subject, about 4.0 to 4.5 mg/kg weight of a subject, about 4.5 to 5.0 mg/kg weight of a subject, about 5.0 to 5.5 mg/kg weight of a subject, about 5.5 to 6.0 mg/kg weight of a subject, about 6.0 to 6.5 mg/kg weight of a subject, about 6.5 to 7.0 mg/kg weight of a subject, about 7.0 to 7.5 mg/kg weight of a subject, about 7.5 to 8.0 mg/kg weight of a subject, about 8.0 to 8.5 mg/kg weight of a subject, about 8.5 to 9.0 mg/kg weight of a subject, about 9.0 to 9.5 mg/kg weight of a subject, about 9.5 to 10.0 mg/kg weight of a subject, about 0.1 to 2 mg/kg weight of a subject, about 2 to 4 mg/kg weight of a subject, about 4 to 6 mg/kg weight of a subject, about 6 to 8 mg/kg weight of a subject, about 8 to 10 mg/kg weight of a subject, about 10 to 12 mg/kg weight of a subject, about 12 to 14 mg/kg weight of a subject, about 14 to 16 mg/kg weight of a subject, about 16 to 18 mg/kg weight of a subject, about 18 to 20 mg/kg weight of a subject, about 500 mg for a subject weighing less than 60 kg, 750 mg for a subject weighing between 60-100 kg or 1000 mg for a subject weighing more than 100 kg, and
wherein the label indicates an effective amount for the second agent being about 0.1 to 40 mg per week, about 5 to 30 mg per week, about 0.1 to 5 mg/week, about 5 to 10 mg/week, about 10 to 15 mg/week, about 15 to 20 mg/week, about 20 to 25 mg/week, about 25 to 30 mg/week, about 30 to 35 mg/week, about 35 to 40 mg/week, about 10 to 30 mg/week, about 10 to 100 mg/week, about 50 mg/week, about 0.1 to 50 mg/kg body weight per week, about 1 to about 5000 mg/day, about 1 to 10 mg/day, about 10 to 50 mg/day, about 50 to 100 mg/day, about 100 to 150 mg/day, about 150 to 200 mg/day, about 200 to 250 mg/day, about 250 to 300 mg/day, about 300 to 350 mg/day, about 350 to 400 mg/day, about 400 to 450 mg/day, about 450 to 500 mg/day, about 500 to 550 mg/day, about 550 to 600 mg/day, about 600 to 650 mg/day, about 650 to 700 mg/day, about 700 to 750 mg/day, about 750 to 800 mg/day, about 800 to 850 mg/day, about 850 to 900 mg/day, about 900 to 950 mg/day, about 950 to 1000 mg/day, about 1000 to 1100 mg/day, about 1100 to 1200 mg/day, about 1200 to 1300 mg/day, about 1300 to 1400 mg/day, about 1400 to 1500 mg/day, about 1500 to 1600 mg/day, about 1600 to 1700 mg/day, about 1700 to 1800 mg/day, about 1800 to 1900 mg/day, about 1900 to 2000 mg/day, about 2000 to 2500 mg/day, about 2500 to 3000 mg/day, about 3000 to 3500 mg/day, about 3500 to 4000 mg/day, about 4000 to 4500 mg/day or about 4500 to 5000 mg/day.
102. The kit of claim 101, wherein the label further indicates that the first and second agents are useful to treat an immune system disease selected from autoimmune diseases, psoriasis, immune disorders associated with graft transplantation rejection, T cell lymphoma, T cell acute lymphoblastic leukemia, testicular angiocentric T cell lymphoma, benign lymphocytic angiitis, lupus erythematosus, Hashimoto's thyroiditis, primary myxedema, Graves' disease, pernicious anemia, autoimmune atrophic gastritis, Addison's disease, insulin dependent diabetes mellitis, good pasture's syndrome, myasthenia gravis, pemphigus, Crohn's disease, sympathetic ophthalmia, autoimmune uveitis, multiple sclerosis, autoimmune hemolytic anemia, idiopathic thrombocytopenia, primary biliary cirrhosis, chronic action hepatitis, ulceratis colitis, Sjogren's syndrome, rheumatic disease, rheumatoid arthritis, polymyositis, scleroderma, mixed connective tissue disease, inflammatory rheumatism, degenerative rheumatism, extra-articular rheumatism, collagen diseases, chronic polyarthritis, psoriasis arthropathica, ankylosing spondylitis, juvenile rheumatoid arthritis, periarthritis humeroscapularis, panarteriitis nodosa, systemic lupus erythematosus, progressive systemic scleroderma, arthritis uratica, dermatomyositis, muscular rheumatism, myositis, myogelosis and chondrocalcinosis.
103. The kit of claim 101, further comprising a container.
104. The kit of claim 101, wherein the soluble CTLA4 comprises the extracellular domain of a CTLA4 molecule, or portion thereof, which binds a B7 antigen expressed on activated B cells.
105. The kit of claim 101, wherein the soluble CTLA4 is a CTLA4 fusion molecule.
106. The kit of claim 105, wherein the CTLA4 fusion molecule comprises an extracellular domain of a CTLA4 molecule which binds a B7 antigen expressed on activated B cells joined to a non-CTLA4 molecule.
107. The kit of claim 106, wherein the non-CTLA4 molecule comprises an amino acid sequence which alters the solubility or affinity comprises an immunoglobulin moiety.
108. The kit of claim 107, wherein the immunoglubulin moiety is an immunoglubulin constant region or portion thereof.
109. The kit of claim 108, wherein the immunoglubulin constant region or portion thereof is mutated to reduce effector function.
110. The kit of claim 105, wherein the CTLA4 fusion molecule is CTLA4Ig.
111. The kit of claim 110, wherein the CTLA4Ig is shown in FIG. 24 (SEQ ID NO:19) beginning with methionine at position +1 or with alanine at position −1 and ending with lysine at position +357.
112. The kit of claim 101, wherein the soluble CTLA4 is a soluble CTLA4 mutant molecule.
113. The kit of claim 112, wherein the soluble CTLA4 mutant molecule comprises an extracellular domain of CTLA4,
a) wherein the extracellular domain of CTLA4 begins with methionine at position +1 or with alanine at position −1 and ends with aspartic acid at position +124 as shown in FIG. 23 or 24 (SEQ ID NO: 17 or 19), and
b) wherein at position +104 of the extracellular domain of CTLA4, leucine is substituted with any other amino acid.
114. The kit of claim 112, wherein the soluble CTLA4 mutant molecule comprises an extracellular domain of CTLA4,
a) wherein the extracellular domain of CTLA4 begins with methionine at position +1 or with alanine at position −1 and ends with aspartic acid at position +124 as shown in FIG. 23 or 24 (SEQ ID NO: 17 or 19),
b) wherein at position +104 of the extracellular domain of CTLA4, leucine is substituted with glutamic acid, and
c) wherein at position +29 of the extracellular domain of CTLA4, alanine is substituted with tyrosine.
115. The kit of claim 101, wherein the soluble CTLA4 mutant molecule is L104EA29YIg as shown in FIG. 19 (SEQ ID NO: 9) beginning with methionine at position +1 or with alanine at position −1 and ending with lysine at position +357 as shown in FIG. 19 (SEQ ID NO: 9).
US10/419,008 2000-07-03 2003-04-18 Methods for treating an autoimmune disease using a soluble CTLA4 molecule and a DMARD or NSAID Abandoned US20040022787A1 (en)

Priority Applications (8)

Application Number Priority Date Filing Date Title
US10/419,008 US20040022787A1 (en) 2000-07-03 2003-04-18 Methods for treating an autoimmune disease using a soluble CTLA4 molecule and a DMARD or NSAID
US12/720,064 US8227420B2 (en) 2000-07-03 2010-03-09 Method for treating an autoimmune disease using a soluble CTLA4 molecule and a DMARD or NSAID
US13/191,923 US8148332B2 (en) 2000-07-03 2011-07-27 Method for treating a rheumatic disease using a soluble TLA4 molecule
US13/404,384 US8497247B2 (en) 2000-07-03 2012-02-24 Methods for treating type I diabetes mellitus by administering a soluble CTLA4 molecule
US13/788,970 US8722632B2 (en) 2000-07-03 2013-03-07 Methods for treating Sjogrens syndrome by administering a soluble CTLA4 molecule
US13/795,545 US8703718B2 (en) 2000-07-03 2013-03-12 Methods for treating juvenile rheumatoid arthritis by administering a soluble CTLA4 molecule
US14/193,687 US9296808B2 (en) 2000-07-03 2014-02-28 Methods for treating scleroderma by administering a soluble CTLA4 molecule
US15/047,849 US10052360B2 (en) 2000-07-03 2016-02-19 Methods for treating dermatomyositis or polymyositis by administering a soluble CTLA4 molecule

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
US21591300P 2000-07-03 2000-07-03
US09/898,195 US7455835B2 (en) 2000-07-03 2001-07-02 Methods for treating immune system diseases using a soluble CTLA4 molecule
US10/419,008 US20040022787A1 (en) 2000-07-03 2003-04-18 Methods for treating an autoimmune disease using a soluble CTLA4 molecule and a DMARD or NSAID

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
US09/898,195 Continuation-In-Part US7455835B2 (en) 2000-07-03 2001-07-02 Methods for treating immune system diseases using a soluble CTLA4 molecule

Related Child Applications (2)

Application Number Title Priority Date Filing Date
US12/720,064 Continuation US8227420B2 (en) 2000-07-03 2010-03-09 Method for treating an autoimmune disease using a soluble CTLA4 molecule and a DMARD or NSAID
US13/191,923 Continuation US8148332B2 (en) 2000-07-03 2011-07-27 Method for treating a rheumatic disease using a soluble TLA4 molecule

Publications (1)

Publication Number Publication Date
US20040022787A1 true US20040022787A1 (en) 2004-02-05

Family

ID=46299178

Family Applications (8)

Application Number Title Priority Date Filing Date
US10/419,008 Abandoned US20040022787A1 (en) 2000-07-03 2003-04-18 Methods for treating an autoimmune disease using a soluble CTLA4 molecule and a DMARD or NSAID
US12/720,064 Expired - Lifetime US8227420B2 (en) 2000-07-03 2010-03-09 Method for treating an autoimmune disease using a soluble CTLA4 molecule and a DMARD or NSAID
US13/191,923 Expired - Lifetime US8148332B2 (en) 2000-07-03 2011-07-27 Method for treating a rheumatic disease using a soluble TLA4 molecule
US13/404,384 Expired - Lifetime US8497247B2 (en) 2000-07-03 2012-02-24 Methods for treating type I diabetes mellitus by administering a soluble CTLA4 molecule
US13/788,970 Expired - Fee Related US8722632B2 (en) 2000-07-03 2013-03-07 Methods for treating Sjogrens syndrome by administering a soluble CTLA4 molecule
US13/795,545 Expired - Fee Related US8703718B2 (en) 2000-07-03 2013-03-12 Methods for treating juvenile rheumatoid arthritis by administering a soluble CTLA4 molecule
US14/193,687 Expired - Lifetime US9296808B2 (en) 2000-07-03 2014-02-28 Methods for treating scleroderma by administering a soluble CTLA4 molecule
US15/047,849 Expired - Fee Related US10052360B2 (en) 2000-07-03 2016-02-19 Methods for treating dermatomyositis or polymyositis by administering a soluble CTLA4 molecule

Family Applications After (7)

Application Number Title Priority Date Filing Date
US12/720,064 Expired - Lifetime US8227420B2 (en) 2000-07-03 2010-03-09 Method for treating an autoimmune disease using a soluble CTLA4 molecule and a DMARD or NSAID
US13/191,923 Expired - Lifetime US8148332B2 (en) 2000-07-03 2011-07-27 Method for treating a rheumatic disease using a soluble TLA4 molecule
US13/404,384 Expired - Lifetime US8497247B2 (en) 2000-07-03 2012-02-24 Methods for treating type I diabetes mellitus by administering a soluble CTLA4 molecule
US13/788,970 Expired - Fee Related US8722632B2 (en) 2000-07-03 2013-03-07 Methods for treating Sjogrens syndrome by administering a soluble CTLA4 molecule
US13/795,545 Expired - Fee Related US8703718B2 (en) 2000-07-03 2013-03-12 Methods for treating juvenile rheumatoid arthritis by administering a soluble CTLA4 molecule
US14/193,687 Expired - Lifetime US9296808B2 (en) 2000-07-03 2014-02-28 Methods for treating scleroderma by administering a soluble CTLA4 molecule
US15/047,849 Expired - Fee Related US10052360B2 (en) 2000-07-03 2016-02-19 Methods for treating dermatomyositis or polymyositis by administering a soluble CTLA4 molecule

Country Status (1)

Country Link
US (8) US20040022787A1 (en)

Cited By (49)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20020031510A1 (en) * 1996-03-20 2002-03-14 Bristol-Myers Squibb Company Methods for inhibiting an immune response by blocking the GP39/CD40 and CTLA4/CD28/B7 pathways and compositions for use therewith
US20020039577A1 (en) * 2000-06-09 2002-04-04 Townsend Robert M. Methods for regulating a lymphocyte-mediated immune response by blocking costimulatory signals and blocking LFA-1 mediated adhesion in lymphocytes
US20020182211A1 (en) * 2000-05-26 2002-12-05 Peach Robert J. Soluble CTLA4 mutant molecules and uses thereof
US20030007968A1 (en) * 2001-01-26 2003-01-09 Larsen Christian P. Methods of inducing organ transplant tolerance and correcting hemoglobinopathies
US20030022836A1 (en) * 2001-05-23 2003-01-30 Larsen Christian P. Methods for protecting allogeneic islet transplant using soluble CTLA4 mutant molecules
US20030083246A1 (en) * 2000-07-03 2003-05-01 Robert Cohen Methods for treating rheumatic diseases using a soluble CTLA4 molecule
US20030219863A1 (en) * 1997-01-31 2003-11-27 Bristol-Myers Squibb Company Soluble CTLA4 mutant molecules and uses thereof
US20040014171A1 (en) * 1997-01-31 2004-01-22 Bristol-Myers Squibb Company Soluble CTLA4 mutant molecules and uses thereof
US20050019859A1 (en) * 2002-12-23 2005-01-27 Schilling Bernhard M. Mammalian ceII culture processes for protein production
US20050084933A1 (en) * 2002-12-23 2005-04-21 Schilling Bernhard M. Product quality enhancement in mammalian cell culture processes for protein production
US20050123539A1 (en) * 2003-08-04 2005-06-09 James Rusnak Methods for treating cardiovascular disease using a soluble CTLA4 molecule
US20050137262A1 (en) * 2003-12-22 2005-06-23 Hu Patrick C. Highly concentrated pourable aqueous solutions of potassium ibuprofen, their preparation and their uses
US20050169919A1 (en) * 1991-06-27 2005-08-04 Bristol-Myers Squibb Company CTLA4 molecules and IL4-binding molecules and uses thereof
US20050191276A1 (en) * 2003-11-25 2005-09-01 Gregory Gurtner Treatment of inflammatory bowel disease through induction of indoleamine 2.3-dioxygenase
US20050238651A1 (en) * 2003-11-25 2005-10-27 Gurtner Gregory J Treatment of inflammatory bowel disease
US20060024306A1 (en) * 2002-06-14 2006-02-02 Warren Strober Methods of treating and preventing colitis involving il-13 and nk-t cells
WO2007076354A2 (en) 2005-12-20 2007-07-05 Bristol-Myers Squibb Company Stable protein formulations
WO2007076032A2 (en) 2005-12-20 2007-07-05 Bristol-Myers Squibb Company Compositions and methods for producing a composition
US20070172895A1 (en) * 2006-01-24 2007-07-26 Bristol-Myers Squibb Company CD86 and CD80 receptor competition assays
US20070274960A1 (en) * 2003-10-08 2007-11-29 Vet-Stem Inc. Methods of Preparing and Using Novel Stem Cell Compositions and Kits Comprising the Same
US20080019999A1 (en) * 2006-05-12 2008-01-24 Bristol-Myers Squibb Company Method of vaccinating subjects receiving immune modulating therapy
US20080135164A1 (en) * 2004-04-01 2008-06-12 Swei Mu Wang Method for forming laminated synthetic leather
US20090258031A1 (en) * 2007-11-01 2009-10-15 Maxygen, Inc. Immunosuppressive polypeptides and nucleic acids
WO2009137424A1 (en) * 2008-05-05 2009-11-12 Bristol-Myers Squibb Company Method of preventing the development of rheumatoid arthritis in subjects with undifferentiated arthritis
US20110059972A1 (en) * 2008-03-26 2011-03-10 Centre National De La Recherche Scientifique 1,4-naphthoquinones derivatives and therapeutic use thereof
US7915395B2 (en) 1991-06-27 2011-03-29 Bristol-Myers Squibb Company Expression vectors encoding bispecific fusion proteins and methods of producing biologically active bispecific fusion proteins in a mammalian cell
US20110212071A1 (en) * 2008-10-30 2011-09-01 Yeda Research And Development Co. Ltd. Anti third party central memory t cells, methods of producing same and use of same in transplantation and disease treatment
WO2012032525A2 (en) 2010-09-08 2012-03-15 Yeda Research And Development Co. Ltd. An immunosuppressive drug combination for a stable and long term engraftment
US20130235806A1 (en) * 2010-11-15 2013-09-12 Telefonaktiebolaget L M Ericsson (Publ) Antenna architecture for maintaining beam shape in a reconfigurable antenna
US20140065644A1 (en) * 2003-10-24 2014-03-06 Immunaid Pty. Ltd. Method of therapy
WO2014151230A2 (en) 2013-03-15 2014-09-25 Bristol-Myers Squibb Company Method of treating granulomatosis with polyangiitis
AU2014213571B2 (en) * 2008-05-05 2015-12-03 Bristol-Myers Squibb Company Method of preventing the development of rheumatoid arthritis in subjects with un-differentiated arthritis
US9309316B2 (en) 2005-12-20 2016-04-12 Bristol-Myers Squibb Company Stable subcutaneous protein formulations and uses thereof
US9421228B2 (en) 2008-10-30 2016-08-23 Yeda Research And Development Co. Ltd. Use of anti third party central memory T cells for anti-leukemia/lymphoma treatment
US9492444B2 (en) 2013-12-17 2016-11-15 Pharmaceutical Manufacturing Research Services, Inc. Extruded extended release abuse deterrent pill
US20160380009A1 (en) * 2015-06-24 2016-12-29 Shenzhen China Star Optoelectronics Technology Co. Ltd Thin film transistor array substrate and manufacturing method thereof
US9707184B2 (en) 2014-07-17 2017-07-18 Pharmaceutical Manufacturing Research Services, Inc. Immediate release abuse deterrent liquid fill dosage form
EP3192805A1 (en) 2016-01-15 2017-07-19 Humanitas Mirasole S.p.A. Inhibitors of t cell activation or stimulation and uses thereof
WO2018081624A1 (en) 2016-10-27 2018-05-03 The United States Government As Represented By The Department Of Veterans Affairs Microrna in t cell activation
US10172797B2 (en) 2013-12-17 2019-01-08 Pharmaceutical Manufacturing Research Services, Inc. Extruded extended release abuse deterrent pill
US10195153B2 (en) 2013-08-12 2019-02-05 Pharmaceutical Manufacturing Research Services, Inc. Extruded immediate release abuse deterrent pill
US20190175657A1 (en) * 2004-03-22 2019-06-13 Mesoblast International Sárl Mesenchymal stem cells and uses therefor
US10714208B2 (en) 2009-05-27 2020-07-14 Biotempus Pty Ltd Computer systems for treating diseases
US10751368B2 (en) 2017-01-18 2020-08-25 Yeda Research And Development Co. Ltd. Methods of transplantation and disease treatment
US10851150B2 (en) 2005-12-20 2020-12-01 Bristol-Myers Squibb Company Carbohydrate content of CTLA4 molecules
US10933124B2 (en) 2015-07-16 2021-03-02 Yeda Research And Development Co. Ltd. Methods of transplantation and disease treatment
US10959958B2 (en) 2014-10-20 2021-03-30 Pharmaceutical Manufacturing Research Services, Inc. Extended release abuse deterrent liquid fill dosage form
US11324777B2 (en) 2011-09-08 2022-05-10 Yeda Research And Development Co. Ltd. Anti third party central memory T cells, methods of producing same and use of same in transplantation and disease treatment
US11555178B2 (en) 2017-01-18 2023-01-17 Yeda Research And Development Co. Ltd. Genetically modified veto cells and use of same in immunotherapy

Families Citing this family (21)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20040022787A1 (en) 2000-07-03 2004-02-05 Robert Cohen Methods for treating an autoimmune disease using a soluble CTLA4 molecule and a DMARD or NSAID
KR20080009111A (en) 2005-04-11 2008-01-24 새비언트 파마수티컬즈 인크. Variant forms of urate oxidase and use thereof
BRPI1010069A2 (en) 2009-06-25 2016-03-15 Savient Pharmaceuticals Inc "method for preventing infusion reactions during pegylated uricase therapy in patients; and method for diagnosing whether a patient treated with pegylated uricase will develop infusion reactions or develop antibody-mediated pegylated uricase release without measuring anti-peg antibody titers and pegylated anti-uricase "
ES2933198T3 (en) 2010-10-21 2023-02-02 Rtu Pharmaceuticals Llc Ready-to-use ketorolac formulations
WO2012142434A2 (en) * 2011-04-15 2012-10-18 THE UNITED STATES OF AMERICA, as represented by the secretary, Depart.of Healtth and Human Services Aav mediated ctla-4 gene transfer to treat sjogren's syndrome
US9511103B2 (en) 2011-04-20 2016-12-06 The United States Of America, As Represented By The Secretary, Department Of Health And Human Services AAV mediated exendin-4 gene transfer to salivary glands to protect subjects from diabetes or obesity
US8735359B2 (en) 2012-05-24 2014-05-27 Orban Biotech Llc Combinations of modalities for the treatment of diabetes
JP2015522026A (en) 2012-06-27 2015-08-03 オルバン バイオテック エルエルシー CTLA4 fusion protein for treating diabetes
AU2013308470B2 (en) 2012-08-31 2017-04-20 The United States Of America, As Represented By The Secretary, Department Of Health And Human Services AAV mediated aquaporin gene transfer to treat Sjogren's syndrome
GB2523399B (en) 2014-02-25 2019-03-13 Orban Tihamer A composition comprising ten overlapping peptide fragments of the entire preproinsulin sequence
WO2016081522A1 (en) 2014-11-17 2016-05-26 Rhode Island Hospital Nanomaterial compositions, synthesis, and assembly
ES2870991T3 (en) 2015-04-17 2021-10-28 Alpine Immune Sciences Inc Immunomodulatory proteins with adjustable affinities
US10874741B2 (en) 2017-08-28 2020-12-29 Spectrix Therapeutics, LLC Compound to treat Sjogren's syndrome
EP4219540A3 (en) 2017-10-10 2023-12-06 Alpine Immune Sciences, Inc. Ctla-4 variant immunomodulatory proteins and uses thereof
TW201925223A (en) 2017-10-18 2019-07-01 美商艾爾潘免疫科學有限公司 Variant ICOS ligand immunomodulatory proteins and related compositions and methods
US20210024603A1 (en) 2018-03-16 2021-01-28 INSERM (Institut National de la Santé et de la Recherche Médicale) Antigenic peptides deriving from urocortin 3 and uses thereof for the diagnosis and treatment of type 1 diabetes
US20210070819A1 (en) 2018-03-16 2021-03-11 INSERM (Institut National de la Santé et de la Recherche Médicale) Antigenic peptides deriving from secretogranin v and uses thereof for the diagnosis and treatment of type 1 diabetes
WO2019175381A1 (en) 2018-03-16 2019-09-19 INSERM (Institut National de la Santé et de la Recherche Médicale) Antigenic peptides deriving from pcsk2 and uses thereof for the diagnosis and treatment of type 1 diabetes
US20200237879A1 (en) * 2019-01-30 2020-07-30 Horizon Pharma Rheumatology Llc Tolerization reduces intolerance to pegloticase and prolongs the urate lowering effect (triple)
US20220125880A1 (en) 2019-02-04 2022-04-28 Assistance Publique - Hôpitaux De Paris Method for treating checkpoint inhibitors induced adverse events
WO2020172391A1 (en) * 2019-02-22 2020-08-27 The Children's Medical Center Methods and compositions relating to the treatment of gvhd

Citations (43)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4399216A (en) * 1980-02-25 1983-08-16 The Trustees Of Columbia University Processes for inserting DNA into eucaryotic cells and for producing proteinaceous materials
US4603102A (en) * 1984-07-06 1986-07-29 Agfa-Gevaert Aktiengesellschaft Photographic silver halide recording material with cellulose dicarboxylic acid semiester particles in outer layer
US5110802A (en) * 1987-07-14 1992-05-05 City Of Hope Oligonucleotide phosphonates and method of inhibiting a human immunodeficiency virus in vitro utilizing said oligonucleotide phosphonates
US5194428A (en) * 1986-05-23 1993-03-16 Worcester Foundation For Experimental Biology Inhibition of influenza virus replication by oligonucleotide phosphorothioates
US5354678A (en) * 1990-10-30 1994-10-11 Applied Immune Sciences, Inc. Production of recombinant adeno-associated virus vectors
US5397703A (en) * 1992-07-09 1995-03-14 Cetus Oncology Corporation Method for generation of antibodies to cell surface molecules
US5434131A (en) * 1991-06-27 1995-07-18 Bristol Myers Squibb Co. Chimeric CTLA4 receptor and methods for its use
US5521288A (en) * 1990-03-26 1996-05-28 Bristol-Myers Squibb Company CD28IG fusion protein
US5539082A (en) * 1993-04-26 1996-07-23 Nielsen; Peter E. Peptide nucleic acids
US5624823A (en) * 1991-11-22 1997-04-29 The General Hospital Corporation DNA encoding procine interleukin-10
US5637481A (en) * 1993-02-01 1997-06-10 Bristol-Myers Squibb Company Expression vectors encoding bispecific fusion proteins and methods of producing biologically active bispecific fusion proteins in a mammalian cell
US5708037A (en) * 1993-12-01 1998-01-13 Sankyo Company, Limited Inhibitor of inflammatory cytokine formation comprising polyprenyl derivatives as the active ingredient
US5747034A (en) * 1992-07-09 1998-05-05 Chiron Corporation Methods and materials for the induction of T cell anergy
US5770197A (en) * 1991-06-27 1998-06-23 Bristol-Myers Squibb Company Methods for regulating the immune response using B7 binding molecules and IL4-binding molecules
US5773253A (en) * 1993-01-22 1998-06-30 Bristol-Myers Squibb Company MYPPPY variants of CTL A4 and uses thereof
US5824655A (en) * 1995-02-15 1998-10-20 The University Of Utah Anti-transforming growth factor-β gene therapy
US5844095A (en) * 1991-06-27 1998-12-01 Bristol-Myers Squibb Company CTLA4 Ig fusion proteins
US5851795A (en) * 1991-06-27 1998-12-22 Bristol-Myers Squibb Company Soluble CTLA4 molecules and uses thereof
US5883223A (en) * 1988-11-23 1999-03-16 Gray; Gary S. CD9 antigen peptides and antibodies thereto
US5916560A (en) * 1996-03-20 1999-06-29 Bristol-Myers Squibb Company Methods for inhibiting an immune response by blocking the GP39/CD40 and CTLA4/CD28/B7 pathways and compositions for use therewith
US5958403A (en) * 1992-02-28 1999-09-28 Beth Israel Hospital Association Methods and compounds for prevention of graft rejection
US5993800A (en) * 1995-06-05 1999-11-30 Bristol-Myers Squibb Company Methods for prolonging the expression of a heterologous gene of interest using soluble CTLA4 molecules and an antiCD40 ligand
US6040292A (en) * 1999-06-04 2000-03-21 Celtrix Pharmaceuticals, Inc. Methods for treating diabetes
US6090914A (en) * 1991-06-27 2000-07-18 Bristol-Myers Squibb Company CTLA4/CD28Ig hybrid fusion proteins and uses thereof
US6113898A (en) * 1995-06-07 2000-09-05 Idec Pharmaceuticals Corporation Human B7.1-specific primatized antibodies and transfectomas expressing said antibodies
US20010053361A1 (en) * 1988-11-23 2001-12-20 The Regents Of The University Of Michigan Immunotherapy involving cd28 stimulation
US6352694B1 (en) * 1994-06-03 2002-03-05 Genetics Institute, Inc. Methods for inducing a population of T cells to proliferate using agents which recognize TCR/CD3 and ligands which stimulate an accessory molecule on the surface of the T cells
US20020039577A1 (en) * 2000-06-09 2002-04-04 Townsend Robert M. Methods for regulating a lymphocyte-mediated immune response by blocking costimulatory signals and blocking LFA-1 mediated adhesion in lymphocytes
US6444792B1 (en) * 1996-02-02 2002-09-03 Repligen Corporation CTLA4-Cγ4 fusion proteins
US20020182211A1 (en) * 2000-05-26 2002-12-05 Peach Robert J. Soluble CTLA4 mutant molecules and uses thereof
US20030007968A1 (en) * 2001-01-26 2003-01-09 Larsen Christian P. Methods of inducing organ transplant tolerance and correcting hemoglobinopathies
US20030022836A1 (en) * 2001-05-23 2003-01-30 Larsen Christian P. Methods for protecting allogeneic islet transplant using soluble CTLA4 mutant molecules
US6534055B1 (en) * 1988-11-23 2003-03-18 Genetics Institute, Inc. Methods for selectively stimulating proliferation of T cells
US20030083246A1 (en) * 2000-07-03 2003-05-01 Robert Cohen Methods for treating rheumatic diseases using a soluble CTLA4 molecule
US20030219863A1 (en) * 1997-01-31 2003-11-27 Bristol-Myers Squibb Company Soluble CTLA4 mutant molecules and uses thereof
US20040014171A1 (en) * 1997-01-31 2004-01-22 Bristol-Myers Squibb Company Soluble CTLA4 mutant molecules and uses thereof
US6685941B1 (en) * 1988-11-23 2004-02-03 The Regents Of The University Of Michigan Methods of treating autoimmune disease via CTLA-4Ig
US6719972B1 (en) * 1994-06-03 2004-04-13 Repligen Corporation Methods of inhibiting T cell proliferation or IL-2 accumulation with CTLA4- specific antibodies
US6830937B1 (en) * 1990-03-26 2004-12-14 Bristol-Myers Squibb Company Method for generating and identifying antibodies directed against a B7
US20050019859A1 (en) * 2002-12-23 2005-01-27 Schilling Bernhard M. Mammalian ceII culture processes for protein production
US20050084933A1 (en) * 2002-12-23 2005-04-21 Schilling Bernhard M. Product quality enhancement in mammalian cell culture processes for protein production
US6887471B1 (en) * 1991-06-27 2005-05-03 Bristol-Myers Squibb Company Method to inhibit T cell interactions with soluble B7
US20050123539A1 (en) * 2003-08-04 2005-06-09 James Rusnak Methods for treating cardiovascular disease using a soluble CTLA4 molecule

Family Cites Families (35)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6905680B2 (en) 1988-11-23 2005-06-14 Genetics Institute, Inc. Methods of treating HIV infected subjects
EP0497922B1 (en) 1989-10-24 2002-01-30 Chiron Corporation Infective protein delivery system
FR2689013B1 (en) 1992-03-30 1995-05-05 Rhone Poulenc Rorer Sa New therapeutic application of phenothiazine derivatives.
CA2133075A1 (en) 1992-04-07 1993-10-14 Craig B. Thompson CD28 Pathway Immunoregulation
JPH08511166A (en) 1993-06-04 1996-11-26 アメリカ合衆国 Method for selectively stimulating T cell proliferation
AU7107794A (en) 1993-06-10 1995-01-03 Regents Of The University Of Michigan, The Cd28 pathway immunosuppression
EP0705377B1 (en) 1993-06-23 2002-01-30 Schlegel Pty Limited Method for making an elongate barrier
WO1995006481A1 (en) 1993-09-02 1995-03-09 Trustees Of Dartmouth College Methods for inducing antigen-specific t cell tolerance
US5683693A (en) 1994-04-25 1997-11-04 Trustees Of Dartmouth College Method for inducing T cell unresponsiveness to a tissue or organ graft with anti-CD40 ligand antibody or soluble CD40
SK136296A3 (en) 1994-04-27 1997-05-07 Henkel Kgaa Refillable container for delivering a spreading compound, in particular an adhesive compound
CA2191586A1 (en) 1994-06-03 1995-12-14 Carl H. June Methods for selectively stimulating proliferation of t cells
JPH10501815A (en) 1994-06-07 1998-02-17 リージェンツ・オブ・ザ・ユニバーシティ・オブ・ミネソタ Methods for inhibiting antigen-specific T cell responses
AU4158396A (en) 1994-11-10 1996-06-06 Dana-Farber Cancer Institute Methods for inhibiting graft versus host disease in bone marrow transplantation
US5876950A (en) 1995-01-26 1999-03-02 Bristol-Myers Squibb Company Monoclonal antibodies specific for different epitopes of human GP39 and methods for their use in diagnosis and therapy
AT405619B (en) 1995-04-25 1999-10-25 Voest Alpine Ind Anlagen ROLLING STAND
EP0813866A3 (en) 1996-06-17 1999-01-20 Eisai Co., Ltd. Therapeutic agent for joint diseases
KR19980066046A (en) 1997-01-18 1998-10-15 정용훈 High-CTLA4-Ig fusion protein
US5773796A (en) * 1997-02-13 1998-06-30 D&K Custom Machine Design, Inc. Heated roller assembly
IL133070A0 (en) 1997-06-11 2001-03-19 Us Navy Composition and method to prevent graft rejection and other counter-adaptive t lymphocyte mediated immune responses
EP0947524A1 (en) 1998-03-30 1999-10-06 Upither B.V. Novel peptides for the treatment of autoimmune diseases
JP2002510655A (en) 1998-04-03 2002-04-09 オシリス セラピューティクス,インコーポレイテッド Method of inhibiting mesenchymal stem cell T cell response as immunosuppressant and its use
JP2002516867A (en) 1998-06-05 2002-06-11 スーパージェン インコーポレイテッド Composition comprising methotrexate and pentostatin for treating rheumatoid arthritis
JP2000086519A (en) 1998-09-17 2000-03-28 Mitsui Chemicals Inc Medicine for reinforcing effect of antirheumatic wedicine
IT1304405B1 (en) 1998-10-21 2001-03-19 Consiglio Nazionale Ricerche PROCESS FOR ABSORPTION OF NITROGEN OXIDES FROM GASEOUS MIXTURES CONTAINING THE SAME.
IL126681A0 (en) 1998-10-21 1999-08-17 Opperbas Holding Bv Treatment of trauma-related conditions
DE60036635T2 (en) * 1999-11-24 2008-07-17 Innovative Development & Markenting As DEVICE FOR TRAINING THE PELVIS MUSCLES
EP1261376A1 (en) 2000-01-27 2002-12-04 Genetics Institute, LLC Antibodies against ctla4(cd152), conjugates comprising same, and uses thereof
WO2001090122A2 (en) 2000-05-23 2001-11-29 Genaissance Pharmaceuticals, Inc. Haplotypes of the ctla4 gene
CZ304451B6 (en) 2000-05-26 2014-05-14 Bristol-Myers Squibb Company CTLA4 mutant or nucleic acid molecule, vector and host vector system, host cell, process for preparing CTLA4 mutant protein and use thereof, pharmaceutical composition and regulation method
US20040022787A1 (en) * 2000-07-03 2004-02-05 Robert Cohen Methods for treating an autoimmune disease using a soluble CTLA4 molecule and a DMARD or NSAID
CN1388804A (en) 2000-07-04 2003-01-01 三井化学株式会社 Amino acid-N-carboxy anhydrides having substitent at nitrogen
US6715607B2 (en) 2001-06-20 2004-04-06 Pinnacle Design Technologies, Inc. Recording medium package having paperboard panels and plastic frame
DE10234925A1 (en) * 2002-07-31 2004-02-19 BSH Bosch und Siemens Hausgeräte GmbH Rotary selector switch for controlling different program sequences of electrical appliance e.g. washing machine, has ratchet body of rotatable operating element provided with magnetic device cooperating with magnetic field sensor
US9309316B2 (en) * 2005-12-20 2016-04-12 Bristol-Myers Squibb Company Stable subcutaneous protein formulations and uses thereof
US7915222B2 (en) * 2008-05-05 2011-03-29 Bristol-Myers Squibb Company Method of preventing the development of rheumatoid arthritis in subjects with undifferentiated arthritis

Patent Citations (54)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4399216A (en) * 1980-02-25 1983-08-16 The Trustees Of Columbia University Processes for inserting DNA into eucaryotic cells and for producing proteinaceous materials
US4603102A (en) * 1984-07-06 1986-07-29 Agfa-Gevaert Aktiengesellschaft Photographic silver halide recording material with cellulose dicarboxylic acid semiester particles in outer layer
US5194428A (en) * 1986-05-23 1993-03-16 Worcester Foundation For Experimental Biology Inhibition of influenza virus replication by oligonucleotide phosphorothioates
US5110802A (en) * 1987-07-14 1992-05-05 City Of Hope Oligonucleotide phosphonates and method of inhibiting a human immunodeficiency virus in vitro utilizing said oligonucleotide phosphonates
US6534055B1 (en) * 1988-11-23 2003-03-18 Genetics Institute, Inc. Methods for selectively stimulating proliferation of T cells
US5883223A (en) * 1988-11-23 1999-03-16 Gray; Gary S. CD9 antigen peptides and antibodies thereto
US20010053361A1 (en) * 1988-11-23 2001-12-20 The Regents Of The University Of Michigan Immunotherapy involving cd28 stimulation
US6685941B1 (en) * 1988-11-23 2004-02-03 The Regents Of The University Of Michigan Methods of treating autoimmune disease via CTLA-4Ig
US6830937B1 (en) * 1990-03-26 2004-12-14 Bristol-Myers Squibb Company Method for generating and identifying antibodies directed against a B7
US5580756A (en) * 1990-03-26 1996-12-03 Bristol-Myers Squibb Co. B7Ig fusion protein
US5521288A (en) * 1990-03-26 1996-05-28 Bristol-Myers Squibb Company CD28IG fusion protein
US6641809B1 (en) * 1990-03-26 2003-11-04 Bristol-Myers Squibb Company Method of regulating cellular processes mediated by B7 and CD28
US5354678A (en) * 1990-10-30 1994-10-11 Applied Immune Sciences, Inc. Production of recombinant adeno-associated virus vectors
US5770197A (en) * 1991-06-27 1998-06-23 Bristol-Myers Squibb Company Methods for regulating the immune response using B7 binding molecules and IL4-binding molecules
US6887471B1 (en) * 1991-06-27 2005-05-03 Bristol-Myers Squibb Company Method to inhibit T cell interactions with soluble B7
US6090914A (en) * 1991-06-27 2000-07-18 Bristol-Myers Squibb Company CTLA4/CD28Ig hybrid fusion proteins and uses thereof
US5434131A (en) * 1991-06-27 1995-07-18 Bristol Myers Squibb Co. Chimeric CTLA4 receptor and methods for its use
US5844095A (en) * 1991-06-27 1998-12-01 Bristol-Myers Squibb Company CTLA4 Ig fusion proteins
US5851795A (en) * 1991-06-27 1998-12-22 Bristol-Myers Squibb Company Soluble CTLA4 molecules and uses thereof
US5977318A (en) * 1991-06-27 1999-11-02 Bristol Myers Squibb Company CTLA4 receptor and uses thereof
US5885579A (en) * 1991-06-27 1999-03-23 Briston-Myers Squibb Company CTLA4 receptor and uses thereof
US5885796A (en) * 1991-06-27 1999-03-23 Bristol-Myers Squibb Company CTLA4 receptor and uses thereof
US5968510A (en) * 1991-06-27 1999-10-19 Bristol-Myers Squibb Company CTLA4 receptor and uses thereof
US5624823A (en) * 1991-11-22 1997-04-29 The General Hospital Corporation DNA encoding procine interleukin-10
US5958403A (en) * 1992-02-28 1999-09-28 Beth Israel Hospital Association Methods and compounds for prevention of graft rejection
US5747034A (en) * 1992-07-09 1998-05-05 Chiron Corporation Methods and materials for the induction of T cell anergy
US5397703A (en) * 1992-07-09 1995-03-14 Cetus Oncology Corporation Method for generation of antibodies to cell surface molecules
US5773253A (en) * 1993-01-22 1998-06-30 Bristol-Myers Squibb Company MYPPPY variants of CTL A4 and uses thereof
US6132992A (en) * 1993-02-01 2000-10-17 Bristol-Myers Squibb Co. Expression vectors encoding bispecific fusion proteins and methods of producing biologically active bispecific fusion proteins in a mammalian cell
US5637481A (en) * 1993-02-01 1997-06-10 Bristol-Myers Squibb Company Expression vectors encoding bispecific fusion proteins and methods of producing biologically active bispecific fusion proteins in a mammalian cell
US5539082A (en) * 1993-04-26 1996-07-23 Nielsen; Peter E. Peptide nucleic acids
US5708037A (en) * 1993-12-01 1998-01-13 Sankyo Company, Limited Inhibitor of inflammatory cytokine formation comprising polyprenyl derivatives as the active ingredient
US6719972B1 (en) * 1994-06-03 2004-04-13 Repligen Corporation Methods of inhibiting T cell proliferation or IL-2 accumulation with CTLA4- specific antibodies
US6352694B1 (en) * 1994-06-03 2002-03-05 Genetics Institute, Inc. Methods for inducing a population of T cells to proliferate using agents which recognize TCR/CD3 and ligands which stimulate an accessory molecule on the surface of the T cells
US5824655A (en) * 1995-02-15 1998-10-20 The University Of Utah Anti-transforming growth factor-β gene therapy
US5993800A (en) * 1995-06-05 1999-11-30 Bristol-Myers Squibb Company Methods for prolonging the expression of a heterologous gene of interest using soluble CTLA4 molecules and an antiCD40 ligand
US6113898A (en) * 1995-06-07 2000-09-05 Idec Pharmaceuticals Corporation Human B7.1-specific primatized antibodies and transfectomas expressing said antibodies
US6444792B1 (en) * 1996-02-02 2002-09-03 Repligen Corporation CTLA4-Cγ4 fusion proteins
US6750334B1 (en) * 1996-02-02 2004-06-15 Repligen Corporation CTLA4-immunoglobulin fusion proteins having modified effector functions and uses therefor
US5916560A (en) * 1996-03-20 1999-06-29 Bristol-Myers Squibb Company Methods for inhibiting an immune response by blocking the GP39/CD40 and CTLA4/CD28/B7 pathways and compositions for use therewith
US20020031510A1 (en) * 1996-03-20 2002-03-14 Bristol-Myers Squibb Company Methods for inhibiting an immune response by blocking the GP39/CD40 and CTLA4/CD28/B7 pathways and compositions for use therewith
US20030219863A1 (en) * 1997-01-31 2003-11-27 Bristol-Myers Squibb Company Soluble CTLA4 mutant molecules and uses thereof
US20040014171A1 (en) * 1997-01-31 2004-01-22 Bristol-Myers Squibb Company Soluble CTLA4 mutant molecules and uses thereof
US6040292A (en) * 1999-06-04 2000-03-21 Celtrix Pharmaceuticals, Inc. Methods for treating diabetes
US20050214313A1 (en) * 2000-05-26 2005-09-29 Peach Robert J Soluble CTLA4 mutant molecules and uses thereof
US20020182211A1 (en) * 2000-05-26 2002-12-05 Peach Robert J. Soluble CTLA4 mutant molecules and uses thereof
US20020039577A1 (en) * 2000-06-09 2002-04-04 Townsend Robert M. Methods for regulating a lymphocyte-mediated immune response by blocking costimulatory signals and blocking LFA-1 mediated adhesion in lymphocytes
US20030083246A1 (en) * 2000-07-03 2003-05-01 Robert Cohen Methods for treating rheumatic diseases using a soluble CTLA4 molecule
US7455835B2 (en) * 2000-07-03 2008-11-25 Bristol-Myers Squibb Company Methods for treating immune system diseases using a soluble CTLA4 molecule
US20030007968A1 (en) * 2001-01-26 2003-01-09 Larsen Christian P. Methods of inducing organ transplant tolerance and correcting hemoglobinopathies
US20030022836A1 (en) * 2001-05-23 2003-01-30 Larsen Christian P. Methods for protecting allogeneic islet transplant using soluble CTLA4 mutant molecules
US20050019859A1 (en) * 2002-12-23 2005-01-27 Schilling Bernhard M. Mammalian ceII culture processes for protein production
US20050084933A1 (en) * 2002-12-23 2005-04-21 Schilling Bernhard M. Product quality enhancement in mammalian cell culture processes for protein production
US20050123539A1 (en) * 2003-08-04 2005-06-09 James Rusnak Methods for treating cardiovascular disease using a soluble CTLA4 molecule

Cited By (115)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20080031876A1 (en) * 1991-06-27 2008-02-07 Bristol-Myers Squibb Company CTLA4 molecules and IL4-binding molecules and uses thereof
US7311910B2 (en) 1991-06-27 2007-12-25 Bristol-Myers Squibb Company Methods for treating cancer and infectious disease by blocking CTLA4-B7 interactions
US7572772B2 (en) 1991-06-27 2009-08-11 Bristol-Myers Squibb Company Soluble CTLA4 mutant molecules
US7915395B2 (en) 1991-06-27 2011-03-29 Bristol-Myers Squibb Company Expression vectors encoding bispecific fusion proteins and methods of producing biologically active bispecific fusion proteins in a mammalian cell
US20050169919A1 (en) * 1991-06-27 2005-08-04 Bristol-Myers Squibb Company CTLA4 molecules and IL4-binding molecules and uses thereof
US20020031510A1 (en) * 1996-03-20 2002-03-14 Bristol-Myers Squibb Company Methods for inhibiting an immune response by blocking the GP39/CD40 and CTLA4/CD28/B7 pathways and compositions for use therewith
US20030219863A1 (en) * 1997-01-31 2003-11-27 Bristol-Myers Squibb Company Soluble CTLA4 mutant molecules and uses thereof
US20040014171A1 (en) * 1997-01-31 2004-01-22 Bristol-Myers Squibb Company Soluble CTLA4 mutant molecules and uses thereof
US9758565B2 (en) 2000-05-26 2017-09-12 Bristol-Myers Squibb Company Methods of treatment using CTLA4 mutant molecules
US7700556B2 (en) 2000-05-26 2010-04-20 Bristol-Myers Squibb Company Methods of treatment using CTLA4 mutant molecules
US8785398B2 (en) 2000-05-26 2014-07-22 Bristol-Myers Squibb Company Methods of treatment using CTLA4 mutant molecules
US20090041769A1 (en) * 2000-05-26 2009-02-12 Bristol-Myers Squibb Company Methods of treatment using CTLA4 mutant molecules
US7439230B2 (en) 2000-05-26 2008-10-21 Bristol-Myers Squibb Company Methods of treatment using CTLA4 mutant molecules
US20020182211A1 (en) * 2000-05-26 2002-12-05 Peach Robert J. Soluble CTLA4 mutant molecules and uses thereof
US7094874B2 (en) 2000-05-26 2006-08-22 Bristol-Myers Squibb Co. Soluble CTLA4 mutant molecules
US10370428B2 (en) 2000-05-26 2019-08-06 Bristol-Myers Squibb Company Methods of treatment using CTLA4 mutant molecules
US20020039577A1 (en) * 2000-06-09 2002-04-04 Townsend Robert M. Methods for regulating a lymphocyte-mediated immune response by blocking costimulatory signals and blocking LFA-1 mediated adhesion in lymphocytes
US20030083246A1 (en) * 2000-07-03 2003-05-01 Robert Cohen Methods for treating rheumatic diseases using a soluble CTLA4 molecule
US20030007968A1 (en) * 2001-01-26 2003-01-09 Larsen Christian P. Methods of inducing organ transplant tolerance and correcting hemoglobinopathies
US7304033B2 (en) 2001-05-23 2007-12-04 Bristol-Myers Squibb Company Methods for protecting allogeneic islet transplant using soluble CTLA4 mutant molecules
US20030022836A1 (en) * 2001-05-23 2003-01-30 Larsen Christian P. Methods for protecting allogeneic islet transplant using soluble CTLA4 mutant molecules
US7666411B2 (en) * 2002-06-14 2010-02-23 The United States Of America As Represented By The Department Of Health And Human Services Methods of treating and preventing colitis involving IL-13 and NK-T cells
US20060024306A1 (en) * 2002-06-14 2006-02-02 Warren Strober Methods of treating and preventing colitis involving il-13 and nk-t cells
US8173123B2 (en) 2002-06-14 2012-05-08 The United States Of America, As Represented By The Secretary, Department Of Health And Human Services Methods of treating colitis involving IL-13 and NK-T cells
US20050084933A1 (en) * 2002-12-23 2005-04-21 Schilling Bernhard M. Product quality enhancement in mammalian cell culture processes for protein production
US7541164B2 (en) 2002-12-23 2009-06-02 Bristol-Myers Squibb Company Mammalian cell culture processes for protein production
US7332303B2 (en) 2002-12-23 2008-02-19 Bristol-Myers Squibb Company Product quality enhancement in mammalian cell culture processes for protein production
US20050019859A1 (en) * 2002-12-23 2005-01-27 Schilling Bernhard M. Mammalian ceII culture processes for protein production
US20090068203A1 (en) * 2003-08-04 2009-03-12 Bristol-Myers Squibb Company Methods for treating cardiovascular disease using a soluble CTLA4 Molecule
US20050123539A1 (en) * 2003-08-04 2005-06-09 James Rusnak Methods for treating cardiovascular disease using a soluble CTLA4 molecule
US7307064B2 (en) 2003-08-04 2007-12-11 Bristol-Myers Squibb Company Methods for treating cardiovascular disease using a soluble CTLA4 molecule
US7671022B2 (en) 2003-08-04 2010-03-02 Bristol-Myers Squibb Company Methods for treating cardiovascular disease using a soluble CTLA4 molecule
US20070274960A1 (en) * 2003-10-08 2007-11-29 Vet-Stem Inc. Methods of Preparing and Using Novel Stem Cell Compositions and Kits Comprising the Same
US10668105B2 (en) 2003-10-08 2020-06-02 Vetstem Biopharma, Inc. Methods of preparing and using novel stem cell compositions and kits comprising the same
US9453202B2 (en) * 2003-10-08 2016-09-27 Vet-Stem, Inc. Methods of preparing and using novel stem cell compositions and kits comprising the same
US11129855B2 (en) 2003-10-08 2021-09-28 Vetstem Biopharma, Inc. Methods of preparing and using novel stem cell compositions and kits comprising the same
US20140065644A1 (en) * 2003-10-24 2014-03-06 Immunaid Pty. Ltd. Method of therapy
US20050238651A1 (en) * 2003-11-25 2005-10-27 Gurtner Gregory J Treatment of inflammatory bowel disease
US20050191276A1 (en) * 2003-11-25 2005-09-01 Gregory Gurtner Treatment of inflammatory bowel disease through induction of indoleamine 2.3-dioxygenase
US20050137262A1 (en) * 2003-12-22 2005-06-23 Hu Patrick C. Highly concentrated pourable aqueous solutions of potassium ibuprofen, their preparation and their uses
US10960025B2 (en) 2004-03-22 2021-03-30 Mesoblast International Sárl Mesenchymal stem cells and uses therefor
US11389484B2 (en) 2004-03-22 2022-07-19 Mesoblast International Sárl Mesenchymal stem cells and uses therefor
US10716814B2 (en) * 2004-03-22 2020-07-21 Mesoblast International Sàrl Mesenchymal stem cells and uses therefor
US20190175657A1 (en) * 2004-03-22 2019-06-13 Mesoblast International Sárl Mesenchymal stem cells and uses therefor
US10828334B1 (en) 2004-03-22 2020-11-10 Mesoblast International Sárl Mesenchymal stem cells and uses therefor
US10668101B2 (en) 2004-03-22 2020-06-02 Mesoblast International Sárl Mesenchymal stem cells and uses therefor
US10729727B2 (en) * 2004-03-22 2020-08-04 Mesoblast International Sárl Mesenchymal stem cells and uses therefor
US20080135164A1 (en) * 2004-04-01 2008-06-12 Swei Mu Wang Method for forming laminated synthetic leather
US10808021B2 (en) 2005-12-20 2020-10-20 Bristol-Myers Squibb Company Compositions and methods for producing a composition
US9309316B2 (en) 2005-12-20 2016-04-12 Bristol-Myers Squibb Company Stable subcutaneous protein formulations and uses thereof
US10851150B2 (en) 2005-12-20 2020-12-01 Bristol-Myers Squibb Company Carbohydrate content of CTLA4 molecules
WO2007076354A2 (en) 2005-12-20 2007-07-05 Bristol-Myers Squibb Company Stable protein formulations
US10941189B2 (en) 2005-12-20 2021-03-09 Bristol-Myers Squibb Company Carbohydrate content of CTLA4 molecules
US10265401B2 (en) 2005-12-20 2019-04-23 Bristol-Myers Squibb Company Stable subcutaneous protein formulations and uses thereof
US20100166774A1 (en) * 2005-12-20 2010-07-01 Bristol-Myers Squibb Company Stable Protein Formulations
US8476239B2 (en) 2005-12-20 2013-07-02 Bristol-Myers Squibb Company Stable protein formulations
WO2007076032A2 (en) 2005-12-20 2007-07-05 Bristol-Myers Squibb Company Compositions and methods for producing a composition
US8969531B2 (en) 2006-01-24 2015-03-03 Bristol-Myers Squibb Company Anti-CD80 antibody
US8378082B2 (en) 2006-01-24 2013-02-19 Catherine Aversa Fleener Anti-CD86 antibody
US7510844B2 (en) 2006-01-24 2009-03-31 Bristol-Myers Squibb Company CD86 and CD80 receptor competition assays
US20070172895A1 (en) * 2006-01-24 2007-07-26 Bristol-Myers Squibb Company CD86 and CD80 receptor competition assays
US7528111B2 (en) 2006-05-12 2009-05-05 Bristol-Myers Squibb Company Method of vaccinating subjects receiving immune modulating therapy
US20080019999A1 (en) * 2006-05-12 2008-01-24 Bristol-Myers Squibb Company Method of vaccinating subjects receiving immune modulating therapy
US8268587B2 (en) 2007-11-01 2012-09-18 Perseid Therapeutics Llc Immunosuppressive polypeptides and nucleic acids
US20110166324A1 (en) * 2007-11-01 2011-07-07 Perseid Therapeutics Llc Immunosuppressive polypeptides and nucleic acids
US8496935B2 (en) 2007-11-01 2013-07-30 Perseid Therapeutics Llc Immunosuppressive polypeptides and nucleic acids
US8491899B2 (en) 2007-11-01 2013-07-23 Perseid Therapeutics Llc Immunosuppressive polypeptides and nucleic acids
US8445230B2 (en) 2007-11-01 2013-05-21 Perseid Therapeutics Llc Immunosuppressive polypeptides and nucleic acids
US20100260759A1 (en) * 2007-11-01 2010-10-14 Perseid Therapeutics Llc Immunosuppressive polypeptides and nucleic acids
US8071095B2 (en) 2007-11-01 2011-12-06 Perseid Therapeutics, LLC Immunosuppressive polypeptides and nucleic acids
US20110177071A1 (en) * 2007-11-01 2011-07-21 Perseid Therapeutics Llc Immunosuppressive polypeptides and nucleic acids
US7794718B2 (en) 2007-11-01 2010-09-14 Perseid Therapeutics, LLC Immunosuppressive polypeptides and nucleic acids
US8318176B2 (en) 2007-11-01 2012-11-27 Perseid Therapeutics Llc Immunosuppressive polypeptides and nucleic acids
US20090258031A1 (en) * 2007-11-01 2009-10-15 Maxygen, Inc. Immunosuppressive polypeptides and nucleic acids
US20110159542A1 (en) * 2007-11-01 2011-06-30 Perseid Therapeutics Llc Immunosuppressive polypeptides and nucleic acids
US20110130546A1 (en) * 2007-11-01 2011-06-02 Perseid Therapeutics Llc Immunosuppressive polypeptides and nucleic acids
US20110129875A1 (en) * 2007-11-01 2011-06-02 Perseid Therapeutics Llc Immunosupprressive polypeptides and nucleic acids
US8283447B2 (en) 2007-11-01 2012-10-09 Perseid Therapeutics Llc Immunosuppressive polypeptides and nucleic acids
US9090549B2 (en) * 2008-03-26 2015-07-28 Centre National De La Recherche Scientifique (C.N.R.S.) 1,4-naphthoquinones derivatives and therapeutic use thereof
US20110059972A1 (en) * 2008-03-26 2011-03-10 Centre National De La Recherche Scientifique 1,4-naphthoquinones derivatives and therapeutic use thereof
AU2014213571B2 (en) * 2008-05-05 2015-12-03 Bristol-Myers Squibb Company Method of preventing the development of rheumatoid arthritis in subjects with un-differentiated arthritis
EP2891665A1 (en) 2008-05-05 2015-07-08 Bristol-Myers Squibb Company Method of preventing the development of rheumatoid arthritis in subjects with undifferentiated arthritis
US9012408B2 (en) 2008-05-05 2015-04-21 Bristol-Myers Squibb Company Method of preventing the development of rheumatoid arthritis in subjects with undifferentiated arthritis
US7915222B2 (en) 2008-05-05 2011-03-29 Bristol-Myers Squibb Company Method of preventing the development of rheumatoid arthritis in subjects with undifferentiated arthritis
WO2009137424A1 (en) * 2008-05-05 2009-11-12 Bristol-Myers Squibb Company Method of preventing the development of rheumatoid arthritis in subjects with undifferentiated arthritis
US20090280119A1 (en) * 2008-05-05 2009-11-12 Bristol-Myers Squibb Company Method of preventing the development of rheumatoid arthritis in subjects with undifferentiated arthritis
TWI454278B (en) * 2008-05-05 2014-10-01 必治妥美雅史谷比公司 Use of ctla4 molecule for the treatment of undifferentiated arthritis
CN102037010A (en) * 2008-05-05 2011-04-27 百时美施贵宝公司 Method of preventing the development of rheumatoid arthritis in subjects with undifferentiated arthritis
AU2009244448B2 (en) * 2008-05-05 2014-07-31 Bristol-Myers Squibb Company Method of preventing the development of rheumatoid arthritis in subjects with undifferentiated arthritis
US8435952B2 (en) 2008-05-05 2013-05-07 George Vratsanos Method for retarding progression to definite rheumatoid arthritis in subjects with undifferentiated arthritis
US20110212071A1 (en) * 2008-10-30 2011-09-01 Yeda Research And Development Co. Ltd. Anti third party central memory t cells, methods of producing same and use of same in transplantation and disease treatment
US9738872B2 (en) 2008-10-30 2017-08-22 Yeda Research And Development Co. Ltd. Anti third party central memory T cells, methods of producing same and use of same in transplantation and disease treatment
US9421228B2 (en) 2008-10-30 2016-08-23 Yeda Research And Development Co. Ltd. Use of anti third party central memory T cells for anti-leukemia/lymphoma treatment
US10714208B2 (en) 2009-05-27 2020-07-14 Biotempus Pty Ltd Computer systems for treating diseases
WO2012032525A2 (en) 2010-09-08 2012-03-15 Yeda Research And Development Co. Ltd. An immunosuppressive drug combination for a stable and long term engraftment
US9112551B2 (en) * 2010-11-15 2015-08-18 Telefonaktiebolaget L M Ericsson (Publ) Antenna architecture for maintaining beam shape in a reconfigurable antenna
US20130235806A1 (en) * 2010-11-15 2013-09-12 Telefonaktiebolaget L M Ericsson (Publ) Antenna architecture for maintaining beam shape in a reconfigurable antenna
US11324777B2 (en) 2011-09-08 2022-05-10 Yeda Research And Development Co. Ltd. Anti third party central memory T cells, methods of producing same and use of same in transplantation and disease treatment
WO2014151230A2 (en) 2013-03-15 2014-09-25 Bristol-Myers Squibb Company Method of treating granulomatosis with polyangiitis
US10195153B2 (en) 2013-08-12 2019-02-05 Pharmaceutical Manufacturing Research Services, Inc. Extruded immediate release abuse deterrent pill
US10639281B2 (en) 2013-08-12 2020-05-05 Pharmaceutical Manufacturing Research Services, Inc. Extruded immediate release abuse deterrent pill
US10172797B2 (en) 2013-12-17 2019-01-08 Pharmaceutical Manufacturing Research Services, Inc. Extruded extended release abuse deterrent pill
US9492444B2 (en) 2013-12-17 2016-11-15 Pharmaceutical Manufacturing Research Services, Inc. Extruded extended release abuse deterrent pill
US10792254B2 (en) 2013-12-17 2020-10-06 Pharmaceutical Manufacturing Research Services, Inc. Extruded extended release abuse deterrent pill
US9707184B2 (en) 2014-07-17 2017-07-18 Pharmaceutical Manufacturing Research Services, Inc. Immediate release abuse deterrent liquid fill dosage form
US10959958B2 (en) 2014-10-20 2021-03-30 Pharmaceutical Manufacturing Research Services, Inc. Extended release abuse deterrent liquid fill dosage form
US20160380009A1 (en) * 2015-06-24 2016-12-29 Shenzhen China Star Optoelectronics Technology Co. Ltd Thin film transistor array substrate and manufacturing method thereof
US10933124B2 (en) 2015-07-16 2021-03-02 Yeda Research And Development Co. Ltd. Methods of transplantation and disease treatment
US11179448B2 (en) 2015-07-16 2021-11-23 Yeda Research And Development Co. Ltd. Genetically modified anti-third party central memory T cells and use of same in immunotherapy
US11844827B2 (en) 2015-07-16 2023-12-19 Yeda Research And Development Co. Ltd. Genetically modified anti-third party central memory T cells and use of same in immunotherapy
EP3192805A1 (en) 2016-01-15 2017-07-19 Humanitas Mirasole S.p.A. Inhibitors of t cell activation or stimulation and uses thereof
WO2017121502A1 (en) 2016-01-15 2017-07-20 Humanitas Mirasole Spa Therapeutic use of inhibitors of t cell activation or stimulation
WO2018081624A1 (en) 2016-10-27 2018-05-03 The United States Government As Represented By The Department Of Veterans Affairs Microrna in t cell activation
US10751368B2 (en) 2017-01-18 2020-08-25 Yeda Research And Development Co. Ltd. Methods of transplantation and disease treatment
US11555178B2 (en) 2017-01-18 2023-01-17 Yeda Research And Development Co. Ltd. Genetically modified veto cells and use of same in immunotherapy

Also Published As

Publication number Publication date
US20100166756A1 (en) 2010-07-01
US8703718B2 (en) 2014-04-22
US8227420B2 (en) 2012-07-24
US10052360B2 (en) 2018-08-21
US9296808B2 (en) 2016-03-29
US8722632B2 (en) 2014-05-13
US8148332B2 (en) 2012-04-03
US20140286948A1 (en) 2014-09-25
US20110311529A1 (en) 2011-12-22
US20120258094A1 (en) 2012-10-11
US20160158318A1 (en) 2016-06-09
US8497247B2 (en) 2013-07-30
US20130230518A1 (en) 2013-09-05
US20130251713A1 (en) 2013-09-26

Similar Documents

Publication Publication Date Title
US10052360B2 (en) Methods for treating dermatomyositis or polymyositis by administering a soluble CTLA4 molecule
EP1935427B1 (en) Uses of soluble CTLA4 mutant molecules
US7671022B2 (en) Methods for treating cardiovascular disease using a soluble CTLA4 molecule
US8785398B2 (en) Methods of treatment using CTLA4 mutant molecules
AU2001273174A1 (en) Methods for treating rheumatic diseases using a soluble CTLA4 molecule
AU2003243152A1 (en) Methods for treating an autoimmune disease using a soluble ctla4 molecule and a dmard or nsaid

Legal Events

Date Code Title Description
AS Assignment

Owner name: BRISTOL-MYERS SQUIBB COMPANY, NEW JERSEY

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:COHEN, ROBERT;CARR, SUZETTE;HAGERTY, DAVID;AND OTHERS;REEL/FRAME:013924/0477;SIGNING DATES FROM 20030725 TO 20030827

STCB Information on status: application discontinuation

Free format text: ABANDONED -- FAILURE TO RESPOND TO AN OFFICE ACTION