US20210130451A1 - Treatment for rheumatoid arthritis - Google Patents

Treatment for rheumatoid arthritis Download PDF

Info

Publication number
US20210130451A1
US20210130451A1 US17/144,685 US202117144685A US2021130451A1 US 20210130451 A1 US20210130451 A1 US 20210130451A1 US 202117144685 A US202117144685 A US 202117144685A US 2021130451 A1 US2021130451 A1 US 2021130451A1
Authority
US
United States
Prior art keywords
antibody
pharmaceutical composition
seq
composition according
sequence
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.)
Pending
Application number
US17/144,685
Inventor
Stefan HARTLE
Stephane Leclair
Amgad Shebl
Stefan Steidl
Bodo Brocks
Daniela Della Ducata
Kai Rosport
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.)
Morphosys AG
Original Assignee
Morphosys AG
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Morphosys AG filed Critical Morphosys AG
Priority to US17/144,685 priority Critical patent/US20210130451A1/en
Publication of US20210130451A1 publication Critical patent/US20210130451A1/en
Pending legal-status Critical Current

Links

Images

Classifications

    • 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
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/495Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with two or more nitrogen atoms as the only ring heteroatoms, e.g. piperazine or tetrazines
    • A61K31/505Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim
    • A61K31/519Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim ortho- or peri-condensed with heterocyclic rings
    • 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
    • A61K39/39533Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum against materials from animals
    • A61K39/3955Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum against materials from animals against proteinaceous materials, e.g. enzymes, hormones, lymphokines
    • 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
    • A61P29/00Non-central analgesic, antipyretic or antiinflammatory agents, e.g. antirheumatic agents; Non-steroidal antiinflammatory drugs [NSAID]
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/24Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against cytokines, lymphokines or interferons
    • C07K16/243Colony Stimulating Factors
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/28Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants
    • C07K16/2866Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants against receptors for cytokines, lymphokines, interferons
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/505Medicinal preparations containing antigens or antibodies comprising antibodies
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/54Medicinal preparations containing antigens or antibodies characterised by the route of administration
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/545Medicinal preparations containing antigens or antibodies characterised by the dose, timing or administration schedule
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/20Immunoglobulins specific features characterized by taxonomic origin
    • C07K2317/21Immunoglobulins specific features characterized by taxonomic origin from primates, e.g. man
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/70Immunoglobulins specific features characterized by effect upon binding to a cell or to an antigen
    • C07K2317/76Antagonist effect on antigen, e.g. neutralization or inhibition of binding

Definitions

  • the present invention provides anti-GM-CSF antibodies for use in the treatment of rheumatoid arthritis, and methods for the treatment of rheumatoid arthritis using such antibodies.
  • Anti-GM-CSF antibodies, in particular MOR103, are administered to patients suffering from rheumatoid arthritis at dosages that are beneficial in a clinical setting.
  • RA Rheumatoid arthritis
  • RA RA primarily affects the joints and is characterized by chronic inflammation of the synovial tissue, which eventually leads to the destruction of cartilage, bone and ligaments and can cause joint deformity.
  • RA has a peak incidence between 40 and 60 years of age and affects primarily women.
  • the cause of RA is not known; however, certain histocompatibility antigens are associated with poorer outcomes.
  • Nonsteroidal anti-inflammatory drugs NSAIDs
  • DMARDs Disease-modifying antirheumatic drugs
  • cytokines such as tumor necrosis factor-alpha (TNF ⁇ ), interleukin-1, interleukin-6 and granulocyte macrophage colony stimulating factor (GM-CSF), which lead to the activation and proliferation of immune cells, are found to be increased in the inflamed joint.
  • TNF ⁇ tumor necrosis factor-alpha
  • GM-CSF granulocyte macrophage colony stimulating factor
  • CSFs granulocyte-macrophage colony-stimulation factor
  • MOR103 is a fully human anti-GM-CSF antibody (Mol Immunol (2008) 46, 135-44; WO 2006/122797). MOR 103 is also in a clinical Phase Ib trial for multiple sclerosis. The present invention describes the development of a clinically efficacious treatment regimen comprising MOR103 for RA.
  • the present invention provides an anti-GM-CSF antibody for use in the treatment of a patient suffering from rheumatoid arthritis, wherein said antibody is administered to said patient in a manner to achieve a therapeutically effective antibody level in the blood of said patient equal or higher compared to the intravenous administration of said antibody at a dose of at least 1.0 mg/kg when administered weekly over at least four weeks.
  • the present invention also provides a method to treat a patient suffering from rheumatoid arthritis, said method comprising administering to said patient an anti-GM-CSF antibody in a manner to achieve a therapeutically effective antibody level in the blood of said patient equal or higher compared to the intravenous administration of said antibody at a dose of at least 1.0 mg/kg when administered weekly over at least four weeks.
  • the anti-GM-CSF antibody is administered intravenously, optionally at a dosage of at least 1.0 mg/kg, or at a dose of about 1.0 mg/kg or about 1.5 mg/kg. In an embodiment, the anti-GM-CSD antibody is administered weekly, over at least four weeks.
  • the anti-GM-CSF antibody is administered subcutaneously, optionally at a dose of at least 2.0 mg/kg, or at a dose of about 2.0 mg/kg, about 3.0 mg/kg or about 4.0 mg/kg.
  • the anti-GM-CSF antibody is administered biweekly, monthly or bimonthly.
  • the antibody is administered at a fixed dose of about 75 mg, of about 100 mg, of about 150 mg, of about 200 mg, of about 300 mg or of about 400 mg. Administration of fixed doses may be every week, every second week, every third week, every fourth week or every sixth week.
  • the dosage of anti-GM-CSF antibody administered to said patient and frequency of said administration is sufficient to provide and maintain a serum concentration of said antibody at at least 2 ⁇ g/ml in said patient over the duration of said treatment.
  • the present invention provides an anti-GM-CSF antibody, wherein said anti-GM-CSF antibody is an antibody comprising an HCDR1 region of sequence GFTFSSYWMN (SEQ ID NO.: 2), an HCDR2 region of sequence GIENKYAGGATYYAASVKG (SEQ ID NO.: 3), an HCDR3 region of sequence GFGTDF (SEQ ID NO.: 4), an LCDR1 region of sequence SGDSIGKKYAY (SEQ ID NO.: 5), an LCDR2 region of sequence KKRPS (SEQ ID NO.: 6), and an LCDR3 region of sequence SAWGDKGM (SEQ ID NO.: 7) for use in the treatment of a patient suffering from rheumatoid arthritis, wherein said antibody is administered to said patient in a manner to achieve a therapeutically effective antibody level in the blood of said patient equal or higher compared to the intravenous administration of said antibody at a dose of at least 1.0 mg/kg when administered weekly over at least four weeks.
  • the present invention provides an anti-GM-CSF antibody, wherein said anti-GM-CSF antibody is an antibody comprising an HCDR1 region of sequence GFTFSSYWMN (SEQ ID NO.: 2), an HCDR2 region of sequence GIENKYAGGATYYAASVKG (SEQ ID NO.: 3), an HCDR3 region of sequence GFGTDF (SEQ ID NO.: 4), an LCDR1 region of sequence SGDSIGKKYAY (SEQ ID NO.: 5), an LCDR2 region of sequence KKRPS (SEQ ID NO.: 6), and an LCDR3 region of sequence SAWGDKGM (SEQ ID NO.: 7) for use in the treatment of a patient suffering from rheumatoid arthritis, wherein said antibody is administered intravenously at a dose of about 1.0 mg/kg or at a dose of about 1.5 mg/kg and wherein said antibody in administered weekly over at least four weeks.
  • said anti-GM-CSF antibody is an antibody comprising an HCDR1 region of
  • the present invention provides an anti-GM-CSF antibody for use in the treatment of a patient suffering from rheumatoid arthritis, wherein said antibody is administered to said patient in a manner to achieve a therapeutically effective antibody level in the blood of said patient equal or higher compared to the intravenous administration of said antibody at a dose of at least 1.0 mg/kg or at least 1.5 mg/kg when administered weekly over at least four weeks, and wherein said anti-GM-CSF antibody is administered in combination with a DMARD, such as methotrexate.
  • a DMARD such as methotrexate
  • the administration of said antibody to achieve such a therapeutically effective amount comprises the administration of said antibody intravenously at a dose at least 0.6, at least 0.7, at least 0.8, at least 0.9 or at least 1.0 mg/kg.
  • the antibody of the present invention is administered intravenously at a dose of about 1.0 mg/kg or a dose of about 1.5 mg/kg. Administration may be monthly, biweekly (every two weeks) or weekly.
  • the present invention provides an anti-GM-CSF antibody for use in the treatment of a patient suffering from rheumatoid arthritis, wherein said antibody is administered to said patient subcutaneously in a manner to achieve a therapeutically effective antibody level in the blood of said patient equal or higher compared to the intravenous administration of said antibody at a dose of at least 1.0 mg/kg or at least 1.5 mg/kg when administered weekly over at least four weeks, and wherein said anti-GM-CSF antibody is administered in combination with a DMARD, such as methotrexate.
  • a DMARD such as methotrexate
  • the administration of said antibody to achieve such a therapeutically effective amount comprises the administration of said antibody subcutaneously at a dose of at least 1.0, at least 1.5, at least 2.0, at least 2.5, at least 3.0, at least 3.5 or at least 4.0 mg/kg.
  • the antibody of the present invention is administered subcutaneously at a dose of about 2.0 mg/kg, a dose of about 3.0 mg/kg or a dose of about 4.0 mg/kg. Administration may be monthly, biweekly (every two weeks) or weekly.
  • the administration of said antibody to achieve such a therapeutically effective amount comprises the administration of said antibody subcutaneously at a fixed dose of about 40 mg, at a fixed dose of 75 mg, at a fixed dose of 100 mg, at a fixed dose of 140 mg, at a fixed dose of 150 mg, at a fixed dose of 180 mg, at a fixed dose of 200 mg, at a fixed dose of 280 mg, at a fixed dose of 300 mg or at a fixed dose of 400 mg.
  • Administration of fixed doses may be every week, every second week, every third week, every fourth week or every sixth week.
  • the present invention provides a method of treating a patient suffering from rheumatoid arthritis, said method comprising administering to said patient an anti-GM-CSF antibody subcutaneously at
  • the present invention provides an anti-GM-CSF antibody for inhibiting progression of structural joint damage in a rheumatoid arthritis patient comprising administering to said patient said antibody in a manner to achieve a therapeutically effective antibody level in the blood of said patient equal or higher compared to the intravenous administration of said antibody at a dose of at least 1.0 mg/kg when administered weekly over at least four weeks.
  • FIG. 1 shows the amino acid sequence and the DNA sequence of MOR04357.
  • FIG. 2 shows the mean changes of the DAS28 score after four weeks (left panel) and after eight weeks (right panel) of treatment compared. DAS28 score changes are compared to baseline levels, i.e. disease status prior to treatment.
  • FIG. 3 shows the average ACR20 score of all treatment arms after four weeks. An increase of the ACR20 scores corresponds to an improvement of the severity of disease.
  • FIG. 4 shows the average ACR20 score of all treatment arms after eight weeks. An increase of the ACR20 scores corresponds to an improvement of the severity of disease.
  • GM-CSF and “GMCSF” refer to the protein known as GM-CSF or Granulocyte-macrophage colony-stimulating factor, having the following synonyms: Colony-stimulating factor 2, CSF2, GMCSF, GM-CSF, Granulocyte-macrophage colony-stimulating factor, MGC131935, MGC138897, Molgramostin, Sargramostim.
  • Human GM-CSF has the amino acid sequence of (UniProt P04141):
  • MOR103 is an anti-GM-CSF antibody whose amino acid sequence and DNA sequence is provided in FIG. 1 .
  • MOR103 and MOR04357 and MOR4357 are used as synonyms to describe the antibody shown in FIG. 1 .
  • MOR04357 comprises an HCDR1 region of sequence GFTFSSYWMN (SEQ ID NO.: 2), an HCDR2 region of sequence GIENKYAGGATYYAASVKG (SEQ ID NO.: 3), an HCDR3 region of sequence GFGTDF (SEQ ID NO.: 4), an LCDR1 region of sequence SGDSIGKKYAY (SEQ ID NO.: 5), an LCDR2 region of sequence KKRPS (SEQ ID NO.: 6), and an LCDR3 region of sequence SAWGDKGM (SEQ ID NO.: 7).
  • MOR04357 comprises a variable heavy chain of the sequence QVQLVESGGGLVQPGGSLRLSCAASGFTFSSYWMNWVRQAPGKGLEWVSGIENKYAGGA TYYAASVKGRFTISRDNSKNTLYLQMNSLRAEDTAVYYCARGFGTDFWGQGTLVTVSS (SEQ ID NO.: 8) and a variable light chain of the sequence DIELTQPPSVSVAPGQTARISCSGDSIGKKYAYWYQQKPGQAPVLVIYKKRPSGIPERFSGS NSGNTATLTISGTQAEDEADYYCSAWGDKGMVFGGGTKLTVLGQ (SEQ ID NO.: 9).
  • the antibody used in the present invention is an antibody specific for GM-CSF. In other embodiments, the antibody used in the present invention is an antibody specific for a polypeptide encoding an amino acid sequence comprising SEQ ID NO.: 1.
  • binding affinity for antigen is of Kd value of 10 ⁇ 9 mol/l or lower (e.g. 10 ⁇ 10 mol/l), preferably with a Kd value of 10 ⁇ 10 mol/l or lower (e.g. 10 ⁇ 12 mol/l).
  • the binding affinity is determined with a standard binding assay, such as surface plasmon resonance technique (BIACORE®).
  • the antibody used in the present invention is MOR103.
  • the antibody used in the present invention is an antibody comprising an HCDR1 region of sequence GFTFSSYWMN (SEQ ID NO.: 2), an HCDR2 region of sequence GIENKYAGGATYYAASVKG (SEQ ID NO.: 3), an HCDR3 region of sequence GFGTDF (SEQ ID NO.: 4), an LCDR1 region of sequence SGDSIGKKYAY (SEQ ID NO.: 5), an LCDR2 region of sequence KKRPS (SEQ ID NO.: 6), and an LCDR3 region of sequence SAWGDKGM (SEQ ID NO.: 7).
  • the antibody used in the present invention is an antibody comprising a variable heavy chain of the sequence QVQLVESGGGLVQPGGSLRLSCAASGFTFSSYWMNWVVRQAPGKGLEWVSGIENKYAGGA TYYAASVKGRFTISRDNSKNTLYLQMNSLRAEDTAVYYCARGFGTDFWGQGTLVTVSS (SEQ ID NO.: 8) and a variable light chain of the sequence DIELTQPPSVSVAPGQTARISCSGDSIGKKYAYWYQQKPGQAPVLVIYKKRPSGIPERFSGS NSGNTATLTISGTQAEDEADYYCSAWGDKGMVFGGGTKLTVLGQ (SEQ ID NO.: 9).
  • the antibody used in the present invention is an antibody which cross-competes with an antibody comprising an HCDR1 region of sequence GFTFSSYWMN (SEQ ID NO.: 2), an HCDR2 region of sequence GIENKYAGGATYYAASVKG (SEQ ID NO.: 3), an HCDR3 region of sequence GFGTDF (SEQ ID NO.: 4), an LCDR1 region of sequence SGDSIGKKYAY (SEQ ID NO.: 5), an LCDR2 region of sequence KKRPS (SEQ ID NO.: 6), and an LCDR3 region of sequence SAWGDKGM (SEQ ID NO.: 7).
  • an antibody comprising an HCDR1 region of sequence GFTFSSYWMN (SEQ ID NO.: 2), an HCDR2 region of sequence GIENKYAGGATYYAASVKG (SEQ ID NO.: 3), an HCDR3 region of sequence GFGTDF (SEQ ID NO.: 4), an LCDR1 region of sequence SGDSIGKKYAY (SEQ
  • the antibody used in the present invention is an antibody which binds to the same epitope like an antibody specific for GM-CSF comprising an HCDR1 region of sequence GFTFSSYWMN (SEQ ID NO.: 2), an HCDR2 region of sequence GIENKYAGGATYYAASVKG (SEQ ID NO.: 3), an HCDR3 region of sequence GFGTDF (SEQ ID NO.: 4), an LCDR1 region of sequence SGDSIGKKYAY (SEQ ID NO.: 5), an LCDR2 region of sequence KKRPS (SEQ ID NO.: 6), and an LCDR3 region of sequence SAWGDKGM (SEQ ID NO.: 7).
  • an antibody specific for GM-CSF comprising an HCDR1 region of sequence GFTFSSYWMN (SEQ ID NO.: 2), an HCDR2 region of sequence GIENKYAGGATYYAASVKG (SEQ ID NO.: 3), an HCDR3 region of sequence GFGTDF (SEQ ID NO.:
  • antibody is used in the broadest sense and specifically covers monoclonal antibodies, polyclonal antibodies, multispecific antibodies (e.g. bispecific antibodies) formed from at least two intact antibodies, and antibody fragments so long as they exhibit the desired biological activity.
  • Antibody fragments herein comprise a portion of an intact antibody which retains the ability to bind antigen.
  • Examples of antibody fragments include Fab, Fab′, F(ab′)2, and Fv fragments; diabodies; linear antibodies; single-chain antibody molecules; and multispecific antibodies formed from antibody fragments.
  • the term “monoclonal antibody” as used herein refers to an antibody obtained from a population of substantially homogeneous antibodies, i.e., the individual antibodies comprising the population are identical and/or bind the same epitope, except for possible variants that may arise during production of the monoclonal antibody, such variants generally being present in minor amounts.
  • each monoclonal antibody is directed against a single determinant on the antigen.
  • the monoclonal antibodies are advantageous in that they are uncontaminated by other immunoglobulins.
  • the monoclonal antibodies herein specifically include “chimeric” antibodies (immunoglobulins) in which a portion of the heavy and/or light chain is identical with or homologous to corresponding sequences in antibodies derived from a particular species or belonging to a particular antibody class or subclass, while the remainder of the chain(s) is identical with or homologous to corresponding sequences in antibodies derived from another species or belonging to another antibody class or subclass, as well as fragments of such antibodies, so long as they exhibit the desired biological activity.
  • chimeric antibodies immunoglobulins in which a portion of the heavy and/or light chain is identical with or homologous to corresponding sequences in antibodies derived from a particular species or belonging to a particular antibody class or subclass, while the remainder of the chain(s) is identical with or homologous to corresponding sequences in antibodies derived from another species or belonging to another antibody class or subclass, as well as fragments of such antibodies, so long as they exhibit the desired biological activity.
  • “Humanized” forms of non-human (e.g., murine) antibodies are chimeric antibodies that contain minimal sequence derived from non-human immunoglobulin.
  • humanized antibodies are human immunoglobulins (recipient antibody) in which residues from a hypervariable region of the recipient are replaced by residues from a hypervariable region of a non-human species (donor antibody) such as mouse, rat, rabbit or nonhuman primate having the desired specificity, affinity, and capacity.
  • donor antibody such as mouse, rat, rabbit or nonhuman primate having the desired specificity, affinity, and capacity.
  • framework region (FR) residues of the human immunoglobulin are replaced by corresponding non-human residues.
  • humanized antibodies may comprise residues that are not found in the recipient antibody or in the donor antibody. These modifications are made to further refine antibody performance.
  • the humanized antibody will comprise substantially all of at least one, and typically two, variable domains, in which all or substantially all of the hypervariable regions correspond to those of a non-human immunoglobulin and all or substantially all of the FRs are those of a human immunoglobulin sequence, except for FR substitution(s) as noted above.
  • the humanized antibody optionally also will comprise at least a portion of an immunoglobulin constant region, typically that of a human immunoglobulin.
  • a “human antibody” herein is one comprising an amino acid sequence structure that corresponds with the amino acid sequence structure of an antibody obtainable from a human B-cell, and includes antigen-binding fragments of human antibodies.
  • Such antibodies can be identified or made by a variety of techniques, including, but not limited to: production by transgenic animals (e.g., mice) that are capable, upon immunization, of producing human antibodies in the absence of endogenous immunoglobulin; selection from phage display libraries expressing human antibodies or human antibody; generation via in vitro activated B; and isolation from human antibody producing hybridomas.
  • the antibody used in the present invention is a monoclonal antibody.
  • the antibody used in the present invention is a chimeric, a humanized or a human antibody. In preferred embodiments, the antibody used in the present invention is a human antibody.
  • the antibody used in the present invention is administered in combination with an additional drug that treats RA.
  • the additional drug may be one or more medicaments, and include, for example, immunosuppressive agents, non-steroidal anti-inflammatory drugs (NSAIDs), disease modifying anti-rheumatic drugs (DMARDs) such as methotrexate (MTX), anti-B-cell surface marker antibodies, such as anti-CD20 antibodies (e.g. rituximab), TNF-alpha-inhibitors, corticosteroids, and co-stimulatory modifiers, or any combination thereof.
  • NSAIDs non-steroidal anti-inflammatory drugs
  • DMARDs disease modifying anti-rheumatic drugs
  • MTX methotrexate
  • anti-B-cell surface marker antibodies such as anti-CD20 antibodies (e.g. rituximab)
  • TNF-alpha-inhibitors e.g. rituximab
  • corticosteroids e.g. rituximab
  • additional drugs are generally used in the same dosages and with administration routes as used hereinbefore and hereinafter. If such additional drugs are used at all, preferably, they are used in lower amounts than if the first medicament were not present, especially in subsequent dosings beyond the initial dosing with the first medicament, so as to eliminate or reduce side effects caused thereby.
  • the combined administration of an additional drug includes co-administration (concurrent administration), using separate formulations or a single pharmaceutical formulation, and consecutive administration in either order, wherein preferably there is a time period while both (or all) active agents (medicaments) simultaneously exert their biological activities.
  • DMARD refers to “Disease-Modifying Anti-Rheumatic Drugs” and includes among others hydroxycloroquine, sulfasalazine, methotrexate, leflunomide, azathioprine, D-penicillamine, gold salts (oral), gold salts (intramuscular), minocycline, cyclosporine including cyclosporine A and topical cyclosporine, and TNF-inhibitors, including salts, variants, and derivatives thereof.
  • Exemplary DMARDs herein are non-biological, i.e. classic, DMARDs, including, azathioprine, chloroquine, hydroxychloroquine, leflunomide, methotrexate and sulfasalazine.
  • Methotrexate is an especially preferred DMARD of the present invention. Therefore, in certain embodiments, the antibody used in the present invention is administered in combination with a DMARD. In other embodiments, the antibody used in the present invention is administered in combination with methotrexate.
  • TNF-inhibitor refers to an agent that inhibits, to some extent, a biological function of TNF-alpha, generally through binding to TNF-alpha and/or its receptor and neutralizing its activity.
  • TNF inhibitors include etanercept (ENBREL®), infliximab (REMICADE®), adalimumab (HUMIRA®), certolizumab pegol (CIMZIA®), and golimumab (SIMPONI®).
  • Treatment of a patient or a subject refers to both therapeutic treatment and prophylactic or preventative measures.
  • the terms “effective amount” or “therapeutically effective” refer to an amount of the antibody that is effective for treating rheumatoid arthritis. Such effective amount can result in any one or more of reducing the signs or symptoms of RA (e.g. achieving ACR20), reducing disease activity (e.g. Disease Activity Score, DAS20), slowing the progression of structural joint damage or improving physical function. In one embodiment, such clinical response is comparable to that achieved with intravenously administered anti-GM-CSF antibody.
  • the antibody of the present invention may be administered in different suitable forms.
  • Potential forms of administration include systemic administration (subcutaneous, intravenous, intramuscular), oral administration, inhalation, transdermal administration, topical application (such as topical cream or ointment, etc.) or by other methods known in the art.
  • the doses (in mg/kg) specified in the present invention refer to milligrams of antibody per kilogram of body weight of the patient.
  • In vitro cell based assays showed that an anti-GM-CSF antibody (MOR103) is capable of inhibiting several GM-CSF mediated responses.
  • Evaluated responses include TF-1 cell proliferation, STAT5 phosphorylation, polymorphonuclear neutrophils (PMN) migration, PMN up-regulation of CD11b, monocyte up-regulation of MHC II, and eosinophil survival.
  • PMN polymorphonuclear neutrophils
  • Complete inhibitory effects were generally reached at concentrations of about 0.2 ⁇ g/ml anti-GM-CSF antibody.
  • GM-CSF concentrations up to 1 ng/ml were applied in such studies.
  • GM-CSF levels in the synovial fluid of RA patients were reported to be ⁇ 500 pg/ml. It is reasonable to consider that similar GM-CSF concentrations as used in these in vitro studies are present in affected tissues of RA patients
  • a specific anti-GM-CSF antibody (MOR103) has been administered to patients with active rheumatoid arthritis who received 4 intravenous weekly doses of 0.3, 1, and 1.5 mg/kg.
  • the anti-GM-CSF antibody showed significant clinical efficacy on DAS28, EULAR, ACR20, ACR50, ACR70 and tender joint counts following once a week dosing with 1 and 1.5 mg/kg as compared to placebo.
  • the antibody of the present invention is administered intravenously. In other embodiments, the antibody of the present invention is administered subcutaneously.
  • a concentration that leads to a certain level of the antibody in the blood when administered intravenously corresponds to about 50-76% of the blood concentration achieved when the same antibody concentration is administered subcutaneously (Meibohm, B.: Pharmacokinetics and Pharmacodynamics of Biotech Drugs, Wiley-VCH, 2006).
  • this ratio was determined to be 52%, i.e. a given concentration administered subcutaneously leads to a blood concentration which is equivalent to about 52% of the blood concentration when the same given concentration is administered intravenously. Therefore, the concentration of a subcutaneous formulation needs to be about twice as high to achieve the same drug blood level as compared to an intravenous formulation.
  • the blood level to be achieved in a patient is equal or higher compared to the blood concentration achieved with intravenous administration of the antibody of the present invention at a dose of at least 1.0 mg/kg when administered weekly over at least four weeks.
  • said blood concentration to be achieved is equal or higher compared to the blood concentration achieved with intravenous administration of the antibody of the present invention at a doses of at least 0.4, 0.5, 0.6, 0.7, 0.8 or 0.9 mg/kg when administered weekly over at least four weeks.
  • the blood level to be achieved in a patient is equal or higher compared to the blood concentration achieved with intravenous administration of the antibody of the present invention at a dose of at least 1.0 mg/kg when administered weekly over at least two weeks or at least three weeks.
  • the blood level to be achieved in a patient is equal or higher compared to the blood concentration achieved with intravenous administration of the antibody of the present invention at a dose of at least 1.0 mg/kg when administered biweekly over at least two weeks or at least four weeks.
  • the antibody of the present invention is administered intravenously. In other embodiments, the antibody of the present invention is administered intravenously at a dose at least 0.6, at least 0.7, at least 0.8, at least 0.9 or at least 1.0 mg/kg. In other embodiments, the antibody of the present invention is administered intravenously at a dose of about 1.0 mg/kg or a dose of about 1.5 mg/kg.
  • the antibody of the present invention is administered subcutaneously.
  • Various dosing regimen have been simulated using the subcutaneous delivery of MOR103 in order to produce plasma concentrations that are similar those obtained after 1 mg/kg iv, a dose that was efficacious in RA.
  • the majority of simulations produce trough concentration values greater than 2 ug/mL, a value that is believed to be the minimum blood concentration that is required to produce efficacy in the context of an anti-GM-CSF antibody.
  • the antibody of the present invention is administered subcutaneously at a dose at least 1.0, at least 1.5, at least 2.0, at least 2.5, at least 3.0, at least 3.5 or at least 4.0 mg/kg. In other embodiments, the antibody of the present invention is administered subcutaneously at a dose of about 2.0 mg/kg, a dose of about 3.0 mg/kg or a dose of about 4.0 mg/kg. In certain embodiments, the antibody of the present invention is subcutaneously administered biweekly, monthly or bimonthly.
  • the antibody of the present invention is administered subcutaneously at a fixed dose.
  • the antibody is administered at a certain, fixed, concentration, i.e. without taking into account a patient's body weight.
  • the antibody of the present invention is administered at a fixed dose of between 40 mg and 400 mg, optionally at a fixed dose of 75 mg, at a fixed dose of 100 mg, at a fixed dose of 140 mg, at a fixed dose of 150 mg, at a fixed dose of 180 mg, at a fixed dose of 200 mg, at a fixed dose of 280 mg, at a fixed dose of 300 mg or at a fixed dose of 400 mg.
  • Administration of fixed doses may be every week, every second week, every third week, every fourth week or every sixth week.
  • the antibody will be administered weekly at a fixed dose.
  • the antibody will be administered weekly, at a fixed subcutaneous dose of 40, 56, 70, 75 100, 140, 150, 180, 200, 210, or 280 mg.
  • the antibody will be administered biweekly, at a fixed subcutaneous dose of 70, 75, 100, 140, 150, 180, 200, 210, 280 or 300 mg.
  • the antibody will be administered monthly, at a fixed subcutaneous dose of 100, 140, 150, 180, 200, 210, 280, 300, 320, 350. 360 or 400 mg.
  • the antibody is administered in a dose sufficient to maintain trough concentration of antibody of at least 2 ug/mL.
  • the trough concentration of antibody may be maintained at 2.0 ug/mL, 2.5 ug/mL, 3.0 ug/mL, 3.5 ug/mL, 4.0 ug/mL, 4.5 ug/mL or 5.0 ug/mL, during the course of therapy.
  • the antibody will be administered weekly, at a fixed subcutaneous dose of 28 or 35 mg.
  • the present invention provides an anti-GM-CSF antibody for use in the treatment of a patient suffering from rheumatoid arthritis, wherein said antibody is administered to said patient in a manner to achieve a therapeutically effective antibody level in the blood of said patient equal or higher compared to the intravenous administration of said antibody at a dose of at least 1.0 mg/kg when administered weekly over at least four weeks.
  • the present invention provides a method to treat a patient suffering from rheumatoid arthritis, said method comprising administering to said patient an anti-GM-CSF antibody in a manner to achieve a therapeutically effective antibody level in the blood of said patient equal or higher compared to the intravenous administration of said antibody at a dose of at least 1.0 mg/kg when administered weekly over at least four weeks.
  • the present invention provides an anti-GM-CSF antibody for inhibiting progression of structural joint damage in a rheumatoid arthritis patient comprising administering to said patient said antibody in a manner to achieve a therapeutically effective antibody level in the blood of said patient equal or higher compared to the intravenous administration of said antibody at a dose of at least 1.0 mg/kg when administered weekly over at least four weeks.
  • drug and “medicament” refer to an active drug to treat rheumatoid arthritis or joint damage or symptoms or side effects associated with RA.
  • pharmaceutical formulation refers to a preparation which is in such form as to permit the biological activity of the active ingredient or ingredients, i.e. the antibody of the present invention, to be effective, and which contains no additional components which are unacceptably toxic to a subject to which the formulation would be administered. Such formulations are sterile.
  • the antibody herein is preferably recombinantly produced in a host cell transformed with nucleic acid sequences encoding its heavy and light chains (e.g. where the host cell has been transformed by one or more vectors with the nucleic acid therein).
  • the preferred host cell is a mammalian cell, most preferably a PER.C6 cell.
  • Therapeutic formulations of the antibody of the present invention are prepared for storage by mixing the antibody having the desired degree of purity with optional pharmaceutically acceptable carriers, excipients or stabilizers in the form of lyophilized formulations or aqueous solutions.
  • Acceptable carriers, excipients, or stabilizers are nontoxic to recipients at the dosages and concentrations employed, and include buffers such as phosphate, citrate, histidine and other organic acids; antioxidants including ascorbic acid and methionine; preservatives (such as octadecyldimethylbenzyl ammonium chloride; hexamethonium chloride; benzalkonium chloride, benzethonium chloride; phenol, butyl or benzyl alcohol; alkyl parabens such as methyl or propyl paraben; catechol; resorcinol; cyclohexanol; 3-pentanol; and m-cresol); low molecular weight (less than about 10 residues) poly
  • the present invention provides a pharmaceutical composition comprising an antibody of the present invention and a pharmaceutically acceptable carrier and/or excipient for use in any of the methods provided in the present invention.
  • the formulation for the antibody of the present invention consists of 30 mM histidine, pH 6.0, 200 mM sorbitol and 0.02% Tween-80.
  • the formulation for the antibody of the present invention consists of PBS, pH 6.2 (0.2 g/l KCl, 0.96 g/l KH 2 PO 4 , 0.66 g/l Na 2 HPO 4 ⁇ 7H 2 O, 8 g/l NaCl).
  • Example 1 Design and Concept of a Clinical Phase Lb/Phase Ila Trial
  • Primary outcome measures were the adverse event rate and the safety profile. Secondary outcome measures included DAS28 scores, ACR scores and EULAR28 response criteria.
  • the clinical trial comprised three treatment arms.
  • patient received either placebo or MOR103.
  • the MOR103 doses were 0.3 mg/kg body weight for treatment arm 1, 1.0 mg/kg body weight for treatment arm 2 and 1.5 mg/kg body weight for treatment arm 3.
  • MOR103 and placebo were administered intravenously, weekly with 4 doses in total.
  • Group 1 Drug: MOR103 MOR103, experimental MOR103 0.3 mg/kg or Biological: MOR103 0.3 mg/kg placebo iv ⁇ 4 doses or placebo
  • Group 2 Drug: MOR103 MOR103, experimental MOR103 1.0 mg/kg or Biological: MOR103 1.0 mg/kg placebo iv ⁇ 4 doses or placebo
  • Group 3 Drug: MOR103 MOR103, experimental MOR103 1.5 mg/kg or Biological: MOR103 1.5 mg/kg placebo iv ⁇ 4 doses or placebo
  • DAS28 score Prior to administration of MOR103 or the placebo the disease activity of all patients was measured according to accepted guidelines by calculating the DAS28 score, a 28-joint Disease Activity Score (see e.g. Ann Rheum Dis (2009) 68, 954-60). DAS28 score is a validated and commonly used tool to quantify the disease status of RA patients. The average DAS28 score was comparable for all treatment arms.
  • MOR103 Based on the available observed safety data, MOR103 showed a favorable safety profile among all doses tested. The key observations are as follows:
  • DAS28 scores 4 weeks and 8 weeks after the first administration of MOR103 (or placebo) the DAS28 scores of all patients was determined. A decrease in DAS28 scores correlates to diminished disease severity. Results are shown in FIG. 2 as the mean changes compared to baseline, i.e. disease status prior to treatment.
  • ACR criteria measure improvement in tender or swollen joint counts and improvement in certain other parameters.
  • the procedure to measure ACR scores is highly standardized.
  • the present clinical trial applied the respective applicable guidelines. Results are depicted in FIGS. 3 and 4 .
  • a higher score corresponds to an improvement in the severity of the disease.
  • the ACR scores show a strong clinical improvement of patients' condition upon treatment with either 1.0 mg/kg MOR103 or 1.5 mg/kg MOR103. The improvement after 4 weeks is highly significant for the 1.0 mg/kg group (p ⁇ 0.0001).
  • the ACR20 scores confirm the surprising finding that the efficacy of MOR103 can already be shown with a comparably low number or patients in each treatment arm and a comparably short treatment period.
  • MOR103 is administered to patients intravenously at a dose of 0.5 mg/kg (treatment arm 1) and 0.75 mg/kg (treatment arm 2). All other parameters are identical to Example 1.
  • Both treatment arms show a favorable safety profile and demonstrate clinical efficacy as measured by DAS28 scores and ACR20 scores.
  • MOR103 is administered to patients at 1.5 mg/kg, 2.0 mg/kg, 3.0 mg/kg and 4.0 mg/kg.
  • the drug is administered sub-cutaneously, either biweekly, monthly or bimonthly. All other parameters are identical to Example 1.
  • Example 8 Clinical Trial with a Sub-Cutaneous Formulation of MOR103 at a Fixed Dose
  • Example 7 is repeated with a fixed dose of MOR103.
  • MOR103 is administered to patients at fixed dose of 75 mg, of 100 mg, of 150 mg, of 200 mg, of 300 mg and of 400 mg.
  • the drug is administered sub-cutaneously every week, every second week, every fourth week or every sixth week. All other parameters are identical to the Examples described herein above.

Abstract

The present invention provides anti-GM-CSF antibodies for use in the treatment of rheumatoid arthritis. Anti-GM-CSF antibodies, in particular MOR103, are administered to patients suffering from rheumatoid arthritis at dosages that are beneficial in a clinical setting.

Description

    FIELD OF THE INVENTION
  • The present invention provides anti-GM-CSF antibodies for use in the treatment of rheumatoid arthritis, and methods for the treatment of rheumatoid arthritis using such antibodies. Anti-GM-CSF antibodies, in particular MOR103, are administered to patients suffering from rheumatoid arthritis at dosages that are beneficial in a clinical setting.
  • BACKGROUND OF THE INVENTION
  • Rheumatoid arthritis (RA) is a chronic systemic inflammatory disease that affects 0.5% to 1% of the adult population worldwide. RA primarily affects the joints and is characterized by chronic inflammation of the synovial tissue, which eventually leads to the destruction of cartilage, bone and ligaments and can cause joint deformity. RA has a peak incidence between 40 and 60 years of age and affects primarily women. The cause of RA is not known; however, certain histocompatibility antigens are associated with poorer outcomes. Nonsteroidal anti-inflammatory drugs (NSAIDs) provide only symptomatic relief. Disease-modifying antirheumatic drugs (DMARDs), the cornerstone of RA treatment throughout all stages of the disease, maintain or improve physical function and retard radiographic joint damage. Pro-inflammatory cytokines, such as tumor necrosis factor-alpha (TNFα), interleukin-1, interleukin-6 and granulocyte macrophage colony stimulating factor (GM-CSF), which lead to the activation and proliferation of immune cells, are found to be increased in the inflamed joint.
  • More recently, biological compounds, such as antibodies, that target tumor necrosis factor alpha (TNFα), B-cells, or T-cells have been used to treat RA, but still many patients fail to respond to these therapies. Colony-stimulating factors (CSFs) have been suggested for a potential point of intervention for inflammatory disorders, such as RA (reviewed e.g. in Nat Rev Immunol (2008) 8, 533-44) or Nat Rev Rheumatol (2009) 5, 554-9). One of such CSF is granulocyte-macrophage colony-stimulation factor (GM-CSF).
  • MOR103 is a fully human anti-GM-CSF antibody (Mol Immunol (2008) 46, 135-44; WO 2006/122797). MOR 103 is also in a clinical Phase Ib trial for multiple sclerosis. The present invention describes the development of a clinically efficacious treatment regimen comprising MOR103 for RA.
  • SUMMARY OF THE INVENTION
  • In one aspect, the present invention provides an anti-GM-CSF antibody for use in the treatment of a patient suffering from rheumatoid arthritis, wherein said antibody is administered to said patient in a manner to achieve a therapeutically effective antibody level in the blood of said patient equal or higher compared to the intravenous administration of said antibody at a dose of at least 1.0 mg/kg when administered weekly over at least four weeks.
  • In another aspect, the present invention also provides a method to treat a patient suffering from rheumatoid arthritis, said method comprising administering to said patient an anti-GM-CSF antibody in a manner to achieve a therapeutically effective antibody level in the blood of said patient equal or higher compared to the intravenous administration of said antibody at a dose of at least 1.0 mg/kg when administered weekly over at least four weeks.
  • In an embodiment, the anti-GM-CSF antibody is administered intravenously, optionally at a dosage of at least 1.0 mg/kg, or at a dose of about 1.0 mg/kg or about 1.5 mg/kg. In an embodiment, the anti-GM-CSD antibody is administered weekly, over at least four weeks.
  • In an embodiment, the anti-GM-CSF antibody is administered subcutaneously, optionally at a dose of at least 2.0 mg/kg, or at a dose of about 2.0 mg/kg, about 3.0 mg/kg or about 4.0 mg/kg. In an embodiment, the anti-GM-CSF antibody is administered biweekly, monthly or bimonthly. In another embodiment, the antibody is administered at a fixed dose of about 75 mg, of about 100 mg, of about 150 mg, of about 200 mg, of about 300 mg or of about 400 mg. Administration of fixed doses may be every week, every second week, every third week, every fourth week or every sixth week.
  • In an embodiment, the dosage of anti-GM-CSF antibody administered to said patient and frequency of said administration is sufficient to provide and maintain a serum concentration of said antibody at at least 2 μg/ml in said patient over the duration of said treatment.
  • In another aspect, the present invention provides an anti-GM-CSF antibody, wherein said anti-GM-CSF antibody is an antibody comprising an HCDR1 region of sequence GFTFSSYWMN (SEQ ID NO.: 2), an HCDR2 region of sequence GIENKYAGGATYYAASVKG (SEQ ID NO.: 3), an HCDR3 region of sequence GFGTDF (SEQ ID NO.: 4), an LCDR1 region of sequence SGDSIGKKYAY (SEQ ID NO.: 5), an LCDR2 region of sequence KKRPS (SEQ ID NO.: 6), and an LCDR3 region of sequence SAWGDKGM (SEQ ID NO.: 7) for use in the treatment of a patient suffering from rheumatoid arthritis, wherein said antibody is administered to said patient in a manner to achieve a therapeutically effective antibody level in the blood of said patient equal or higher compared to the intravenous administration of said antibody at a dose of at least 1.0 mg/kg when administered weekly over at least four weeks.
  • In another aspect, the present invention provides an anti-GM-CSF antibody, wherein said anti-GM-CSF antibody is an antibody comprising an HCDR1 region of sequence GFTFSSYWMN (SEQ ID NO.: 2), an HCDR2 region of sequence GIENKYAGGATYYAASVKG (SEQ ID NO.: 3), an HCDR3 region of sequence GFGTDF (SEQ ID NO.: 4), an LCDR1 region of sequence SGDSIGKKYAY (SEQ ID NO.: 5), an LCDR2 region of sequence KKRPS (SEQ ID NO.: 6), and an LCDR3 region of sequence SAWGDKGM (SEQ ID NO.: 7) for use in the treatment of a patient suffering from rheumatoid arthritis, wherein said antibody is administered intravenously at a dose of about 1.0 mg/kg or at a dose of about 1.5 mg/kg and wherein said antibody in administered weekly over at least four weeks.
  • In another aspect, the present invention provides an anti-GM-CSF antibody for use in the treatment of a patient suffering from rheumatoid arthritis, wherein said antibody is administered to said patient in a manner to achieve a therapeutically effective antibody level in the blood of said patient equal or higher compared to the intravenous administration of said antibody at a dose of at least 1.0 mg/kg or at least 1.5 mg/kg when administered weekly over at least four weeks, and wherein said anti-GM-CSF antibody is administered in combination with a DMARD, such as methotrexate.
  • In an embodiment, the administration of said antibody to achieve such a therapeutically effective amount comprises the administration of said antibody intravenously at a dose at least 0.6, at least 0.7, at least 0.8, at least 0.9 or at least 1.0 mg/kg. In other embodiments, the antibody of the present invention is administered intravenously at a dose of about 1.0 mg/kg or a dose of about 1.5 mg/kg. Administration may be monthly, biweekly (every two weeks) or weekly.
  • In another aspect, the present invention provides an anti-GM-CSF antibody for use in the treatment of a patient suffering from rheumatoid arthritis, wherein said antibody is administered to said patient subcutaneously in a manner to achieve a therapeutically effective antibody level in the blood of said patient equal or higher compared to the intravenous administration of said antibody at a dose of at least 1.0 mg/kg or at least 1.5 mg/kg when administered weekly over at least four weeks, and wherein said anti-GM-CSF antibody is administered in combination with a DMARD, such as methotrexate.
  • In an embodiment, the administration of said antibody to achieve such a therapeutically effective amount comprises the administration of said antibody subcutaneously at a dose of at least 1.0, at least 1.5, at least 2.0, at least 2.5, at least 3.0, at least 3.5 or at least 4.0 mg/kg. In other embodiments, the antibody of the present invention is administered subcutaneously at a dose of about 2.0 mg/kg, a dose of about 3.0 mg/kg or a dose of about 4.0 mg/kg. Administration may be monthly, biweekly (every two weeks) or weekly.
  • In an embodiment, the administration of said antibody to achieve such a therapeutically effective amount comprises the administration of said antibody subcutaneously at a fixed dose of about 40 mg, at a fixed dose of 75 mg, at a fixed dose of 100 mg, at a fixed dose of 140 mg, at a fixed dose of 150 mg, at a fixed dose of 180 mg, at a fixed dose of 200 mg, at a fixed dose of 280 mg, at a fixed dose of 300 mg or at a fixed dose of 400 mg. Administration of fixed doses may be every week, every second week, every third week, every fourth week or every sixth week.
  • In another aspect, the present invention provides a method of treating a patient suffering from rheumatoid arthritis, said method comprising administering to said patient an anti-GM-CSF antibody subcutaneously at
      • (i) a dose of at least 1.0 mg/kg, or
      • (ii) a fixed dose of between 40 mg and 400 mg.
        The anti-GM-CSF antibody may be administered to said patient in a manner to achieve to a serum concentration of said antibody at at least 2 μg/ml in said patient over the duration of said treatment. The antibody may be administered to said patient in a manner to achieve a therapeutically effective antibody level in the blood of said patient equal or higher compared to the intravenous administration of said antibody at a dose of at least 1.0 mg/kg when administered weekly over at least four weeks.
  • In another aspect, the present invention provides an anti-GM-CSF antibody for inhibiting progression of structural joint damage in a rheumatoid arthritis patient comprising administering to said patient said antibody in a manner to achieve a therapeutically effective antibody level in the blood of said patient equal or higher compared to the intravenous administration of said antibody at a dose of at least 1.0 mg/kg when administered weekly over at least four weeks.
  • BRIEF DESCRIPTION OF THE DRAWINGS
  • FIG. 1 shows the amino acid sequence and the DNA sequence of MOR04357.
  • FIG. 2 shows the mean changes of the DAS28 score after four weeks (left panel) and after eight weeks (right panel) of treatment compared. DAS28 score changes are compared to baseline levels, i.e. disease status prior to treatment.
  • FIG. 3 shows the average ACR20 score of all treatment arms after four weeks. An increase of the ACR20 scores corresponds to an improvement of the severity of disease.
  • FIG. 4 shows the average ACR20 score of all treatment arms after eight weeks. An increase of the ACR20 scores corresponds to an improvement of the severity of disease.
  • DESCRIPTION
  • The terms “GM-CSF” and “GMCSF” refer to the protein known as GM-CSF or Granulocyte-macrophage colony-stimulating factor, having the following synonyms: Colony-stimulating factor 2, CSF2, GMCSF, GM-CSF, Granulocyte-macrophage colony-stimulating factor, MGC131935, MGC138897, Molgramostin, Sargramostim. Human GM-CSF has the amino acid sequence of (UniProt P04141):
  • (SEQ ID NO.: 1)
    MWLQSLLLLGTVACSISAPARSPSPSTQPWEHVNAIQEARRLLNLSRDTA
    AEMNETVEVISEMFDLQEPTCLQTRLELYKQGLRGSLTKLKGPLTMMASH
    YKQHCPPTPETSCATQIITFESFKENLKDFLLVIPFDCWEPVQE
  • “MOR103” is an anti-GM-CSF antibody whose amino acid sequence and DNA sequence is provided in FIG. 1. “MOR103” and “MOR04357” and “MOR4357” are used as synonyms to describe the antibody shown in FIG. 1. MOR04357 comprises an HCDR1 region of sequence GFTFSSYWMN (SEQ ID NO.: 2), an HCDR2 region of sequence GIENKYAGGATYYAASVKG (SEQ ID NO.: 3), an HCDR3 region of sequence GFGTDF (SEQ ID NO.: 4), an LCDR1 region of sequence SGDSIGKKYAY (SEQ ID NO.: 5), an LCDR2 region of sequence KKRPS (SEQ ID NO.: 6), and an LCDR3 region of sequence SAWGDKGM (SEQ ID NO.: 7). MOR04357 comprises a variable heavy chain of the sequence QVQLVESGGGLVQPGGSLRLSCAASGFTFSSYWMNWVRQAPGKGLEWVSGIENKYAGGA TYYAASVKGRFTISRDNSKNTLYLQMNSLRAEDTAVYYCARGFGTDFWGQGTLVTVSS (SEQ ID NO.: 8) and a variable light chain of the sequence DIELTQPPSVSVAPGQTARISCSGDSIGKKYAYWYQQKPGQAPVLVIYKKRPSGIPERFSGS NSGNTATLTISGTQAEDEADYYCSAWGDKGMVFGGGTKLTVLGQ (SEQ ID NO.: 9).
  • In certain embodiments, the antibody used in the present invention is an antibody specific for GM-CSF. In other embodiments, the antibody used in the present invention is an antibody specific for a polypeptide encoding an amino acid sequence comprising SEQ ID NO.: 1.
  • As used herein, “specifically for” or “specifically binding to” refers to an antibody selectively or preferentially binding to GM-CSF. Preferably the binding affinity for antigen is of Kd value of 10−9 mol/l or lower (e.g. 10−10 mol/l), preferably with a Kd value of 10−10 mol/l or lower (e.g. 10−12 mol/l). The binding affinity is determined with a standard binding assay, such as surface plasmon resonance technique (BIACORE®).
  • In certain embodiments, the antibody used in the present invention is MOR103. In other embodiments, the antibody used in the present invention is an antibody comprising an HCDR1 region of sequence GFTFSSYWMN (SEQ ID NO.: 2), an HCDR2 region of sequence GIENKYAGGATYYAASVKG (SEQ ID NO.: 3), an HCDR3 region of sequence GFGTDF (SEQ ID NO.: 4), an LCDR1 region of sequence SGDSIGKKYAY (SEQ ID NO.: 5), an LCDR2 region of sequence KKRPS (SEQ ID NO.: 6), and an LCDR3 region of sequence SAWGDKGM (SEQ ID NO.: 7). In other embodiments, the antibody used in the present invention is an antibody comprising a variable heavy chain of the sequence QVQLVESGGGLVQPGGSLRLSCAASGFTFSSYWMNWVVRQAPGKGLEWVSGIENKYAGGA TYYAASVKGRFTISRDNSKNTLYLQMNSLRAEDTAVYYCARGFGTDFWGQGTLVTVSS (SEQ ID NO.: 8) and a variable light chain of the sequence DIELTQPPSVSVAPGQTARISCSGDSIGKKYAYWYQQKPGQAPVLVIYKKRPSGIPERFSGS NSGNTATLTISGTQAEDEADYYCSAWGDKGMVFGGGTKLTVLGQ (SEQ ID NO.: 9). In other embodiments, the antibody used in the present invention is an antibody which cross-competes with an antibody comprising an HCDR1 region of sequence GFTFSSYWMN (SEQ ID NO.: 2), an HCDR2 region of sequence GIENKYAGGATYYAASVKG (SEQ ID NO.: 3), an HCDR3 region of sequence GFGTDF (SEQ ID NO.: 4), an LCDR1 region of sequence SGDSIGKKYAY (SEQ ID NO.: 5), an LCDR2 region of sequence KKRPS (SEQ ID NO.: 6), and an LCDR3 region of sequence SAWGDKGM (SEQ ID NO.: 7). In other embodiments, the antibody used in the present invention is an antibody which binds to the same epitope like an antibody specific for GM-CSF comprising an HCDR1 region of sequence GFTFSSYWMN (SEQ ID NO.: 2), an HCDR2 region of sequence GIENKYAGGATYYAASVKG (SEQ ID NO.: 3), an HCDR3 region of sequence GFGTDF (SEQ ID NO.: 4), an LCDR1 region of sequence SGDSIGKKYAY (SEQ ID NO.: 5), an LCDR2 region of sequence KKRPS (SEQ ID NO.: 6), and an LCDR3 region of sequence SAWGDKGM (SEQ ID NO.: 7).
  • The term “antibody” is used in the broadest sense and specifically covers monoclonal antibodies, polyclonal antibodies, multispecific antibodies (e.g. bispecific antibodies) formed from at least two intact antibodies, and antibody fragments so long as they exhibit the desired biological activity.
  • “Antibody fragments” herein comprise a portion of an intact antibody which retains the ability to bind antigen. Examples of antibody fragments include Fab, Fab′, F(ab′)2, and Fv fragments; diabodies; linear antibodies; single-chain antibody molecules; and multispecific antibodies formed from antibody fragments.
  • The term “monoclonal antibody” as used herein refers to an antibody obtained from a population of substantially homogeneous antibodies, i.e., the individual antibodies comprising the population are identical and/or bind the same epitope, except for possible variants that may arise during production of the monoclonal antibody, such variants generally being present in minor amounts. In contrast to polyclonal antibody preparations that typically include different antibodies directed against different determinants (epitopes), each monoclonal antibody is directed against a single determinant on the antigen. In addition to their specificity, the monoclonal antibodies are advantageous in that they are uncontaminated by other immunoglobulins.
  • The monoclonal antibodies herein specifically include “chimeric” antibodies (immunoglobulins) in which a portion of the heavy and/or light chain is identical with or homologous to corresponding sequences in antibodies derived from a particular species or belonging to a particular antibody class or subclass, while the remainder of the chain(s) is identical with or homologous to corresponding sequences in antibodies derived from another species or belonging to another antibody class or subclass, as well as fragments of such antibodies, so long as they exhibit the desired biological activity.
  • “Humanized” forms of non-human (e.g., murine) antibodies are chimeric antibodies that contain minimal sequence derived from non-human immunoglobulin. For the most part, humanized antibodies are human immunoglobulins (recipient antibody) in which residues from a hypervariable region of the recipient are replaced by residues from a hypervariable region of a non-human species (donor antibody) such as mouse, rat, rabbit or nonhuman primate having the desired specificity, affinity, and capacity. In some instances, framework region (FR) residues of the human immunoglobulin are replaced by corresponding non-human residues. Furthermore, humanized antibodies may comprise residues that are not found in the recipient antibody or in the donor antibody. These modifications are made to further refine antibody performance. In general, the humanized antibody will comprise substantially all of at least one, and typically two, variable domains, in which all or substantially all of the hypervariable regions correspond to those of a non-human immunoglobulin and all or substantially all of the FRs are those of a human immunoglobulin sequence, except for FR substitution(s) as noted above. The humanized antibody optionally also will comprise at least a portion of an immunoglobulin constant region, typically that of a human immunoglobulin.
  • A “human antibody” herein is one comprising an amino acid sequence structure that corresponds with the amino acid sequence structure of an antibody obtainable from a human B-cell, and includes antigen-binding fragments of human antibodies. Such antibodies can be identified or made by a variety of techniques, including, but not limited to: production by transgenic animals (e.g., mice) that are capable, upon immunization, of producing human antibodies in the absence of endogenous immunoglobulin; selection from phage display libraries expressing human antibodies or human antibody; generation via in vitro activated B; and isolation from human antibody producing hybridomas.
  • In certain embodiments, the antibody used in the present invention is a monoclonal antibody.
  • In other embodiments, the antibody used in the present invention is a chimeric, a humanized or a human antibody. In preferred embodiments, the antibody used in the present invention is a human antibody.
  • In certain embodiments, the antibody used in the present invention is administered in combination with an additional drug that treats RA.
  • The additional drug may be one or more medicaments, and include, for example, immunosuppressive agents, non-steroidal anti-inflammatory drugs (NSAIDs), disease modifying anti-rheumatic drugs (DMARDs) such as methotrexate (MTX), anti-B-cell surface marker antibodies, such as anti-CD20 antibodies (e.g. rituximab), TNF-alpha-inhibitors, corticosteroids, and co-stimulatory modifiers, or any combination thereof. Optionally, the second or additional drug is selected from the group consisting of non-biological DMARDs, NSAIDS, and corticosteroids.
  • These additional drugs are generally used in the same dosages and with administration routes as used hereinbefore and hereinafter. If such additional drugs are used at all, preferably, they are used in lower amounts than if the first medicament were not present, especially in subsequent dosings beyond the initial dosing with the first medicament, so as to eliminate or reduce side effects caused thereby. The combined administration of an additional drug includes co-administration (concurrent administration), using separate formulations or a single pharmaceutical formulation, and consecutive administration in either order, wherein preferably there is a time period while both (or all) active agents (medicaments) simultaneously exert their biological activities.
  • The term “DMARD” refers to “Disease-Modifying Anti-Rheumatic Drugs” and includes among others hydroxycloroquine, sulfasalazine, methotrexate, leflunomide, azathioprine, D-penicillamine, gold salts (oral), gold salts (intramuscular), minocycline, cyclosporine including cyclosporine A and topical cyclosporine, and TNF-inhibitors, including salts, variants, and derivatives thereof. Exemplary DMARDs herein are non-biological, i.e. classic, DMARDs, including, azathioprine, chloroquine, hydroxychloroquine, leflunomide, methotrexate and sulfasalazine.
  • Methotrexate is an especially preferred DMARD of the present invention. Therefore, in certain embodiments, the antibody used in the present invention is administered in combination with a DMARD. In other embodiments, the antibody used in the present invention is administered in combination with methotrexate.
  • A “TNF-inhibitor” as used herein refers to an agent that inhibits, to some extent, a biological function of TNF-alpha, generally through binding to TNF-alpha and/or its receptor and neutralizing its activity. Examples of TNF inhibitors include etanercept (ENBREL®), infliximab (REMICADE®), adalimumab (HUMIRA®), certolizumab pegol (CIMZIA®), and golimumab (SIMPONI®).
  • “Treatment” of a patient or a subject refers to both therapeutic treatment and prophylactic or preventative measures. The terms “effective amount” or “therapeutically effective” refer to an amount of the antibody that is effective for treating rheumatoid arthritis. Such effective amount can result in any one or more of reducing the signs or symptoms of RA (e.g. achieving ACR20), reducing disease activity (e.g. Disease Activity Score, DAS20), slowing the progression of structural joint damage or improving physical function. In one embodiment, such clinical response is comparable to that achieved with intravenously administered anti-GM-CSF antibody.
  • The antibody of the present invention may be administered in different suitable forms. Potential forms of administration include systemic administration (subcutaneous, intravenous, intramuscular), oral administration, inhalation, transdermal administration, topical application (such as topical cream or ointment, etc.) or by other methods known in the art. The doses (in mg/kg) specified in the present invention refer to milligrams of antibody per kilogram of body weight of the patient. In vitro cell based assays showed that an anti-GM-CSF antibody (MOR103) is capable of inhibiting several GM-CSF mediated responses. Evaluated responses include TF-1 cell proliferation, STAT5 phosphorylation, polymorphonuclear neutrophils (PMN) migration, PMN up-regulation of CD11b, monocyte up-regulation of MHC II, and eosinophil survival. Complete inhibitory effects were generally reached at concentrations of about 0.2 μg/ml anti-GM-CSF antibody. GM-CSF concentrations up to 1 ng/ml were applied in such studies. As a reference, GM-CSF levels in the synovial fluid of RA patients were reported to be <500 pg/ml. It is reasonable to consider that similar GM-CSF concentrations as used in these in vitro studies are present in affected tissues of RA patients
  • To effectively treat RA it may be important for an anti-GM-CSF antibody to penetrate the synovium. There is evidence to suggest that monoclonal antibodies can distribute into the synovium when dosed subcutaneously or intravenously. Based on a predicted penetration rate of 30%, continuous GM-CSF production and considering patient heterogeneity, the minimal or sub-optimal clinical effect level in RA patients is anticipated to be at a serum concentration of approximately 2 μg/ml antibody (thus, approximately 10-fold higher than the inhibitory concentration derived from in vitro studies).
  • A specific anti-GM-CSF antibody (MOR103) has been administered to patients with active rheumatoid arthritis who received 4 intravenous weekly doses of 0.3, 1, and 1.5 mg/kg. The anti-GM-CSF antibody showed significant clinical efficacy on DAS28, EULAR, ACR20, ACR50, ACR70 and tender joint counts following once a week dosing with 1 and 1.5 mg/kg as compared to placebo.
  • In certain embodiments, the antibody of the present invention is administered intravenously. In other embodiments, the antibody of the present invention is administered subcutaneously.
  • From other therapeutic antibodies it is known that a concentration that leads to a certain level of the antibody in the blood when administered intravenously corresponds to about 50-76% of the blood concentration achieved when the same antibody concentration is administered subcutaneously (Meibohm, B.: Pharmacokinetics and Pharmacodynamics of Biotech Drugs, Wiley-VCH, 2006). For MOR103 this ratio was determined to be 52%, i.e. a given concentration administered subcutaneously leads to a blood concentration which is equivalent to about 52% of the blood concentration when the same given concentration is administered intravenously. Therefore, the concentration of a subcutaneous formulation needs to be about twice as high to achieve the same drug blood level as compared to an intravenous formulation.
  • In certain embodiments the blood level to be achieved in a patient is equal or higher compared to the blood concentration achieved with intravenous administration of the antibody of the present invention at a dose of at least 1.0 mg/kg when administered weekly over at least four weeks.
  • In alternative embodiments said blood concentration to be achieved is equal or higher compared to the blood concentration achieved with intravenous administration of the antibody of the present invention at a doses of at least 0.4, 0.5, 0.6, 0.7, 0.8 or 0.9 mg/kg when administered weekly over at least four weeks. In alternative embodiments the blood level to be achieved in a patient is equal or higher compared to the blood concentration achieved with intravenous administration of the antibody of the present invention at a dose of at least 1.0 mg/kg when administered weekly over at least two weeks or at least three weeks. In alternative embodiments the blood level to be achieved in a patient is equal or higher compared to the blood concentration achieved with intravenous administration of the antibody of the present invention at a dose of at least 1.0 mg/kg when administered biweekly over at least two weeks or at least four weeks.
  • In certain embodiments, the antibody of the present invention is administered intravenously. In other embodiments, the antibody of the present invention is administered intravenously at a dose at least 0.6, at least 0.7, at least 0.8, at least 0.9 or at least 1.0 mg/kg. In other embodiments, the antibody of the present invention is administered intravenously at a dose of about 1.0 mg/kg or a dose of about 1.5 mg/kg.
  • In certain embodiments, the antibody of the present invention is administered subcutaneously. Various dosing regimen have been simulated using the subcutaneous delivery of MOR103 in order to produce plasma concentrations that are similar those obtained after 1 mg/kg iv, a dose that was efficacious in RA. The majority of simulations produce trough concentration values greater than 2 ug/mL, a value that is believed to be the minimum blood concentration that is required to produce efficacy in the context of an anti-GM-CSF antibody. These studies indicate that subcutaneous doses of 1, 2, 3 and 4 mg/kg can produce plasma concentration similar to 1 mg/kg, IV depending on the dosing frequency.
  • In other embodiments, the antibody of the present invention is administered subcutaneously at a dose at least 1.0, at least 1.5, at least 2.0, at least 2.5, at least 3.0, at least 3.5 or at least 4.0 mg/kg. In other embodiments, the antibody of the present invention is administered subcutaneously at a dose of about 2.0 mg/kg, a dose of about 3.0 mg/kg or a dose of about 4.0 mg/kg. In certain embodiments, the antibody of the present invention is subcutaneously administered biweekly, monthly or bimonthly.
  • In other embodiments, the antibody of the present invention is administered subcutaneously at a fixed dose. In such “fixed dose” treatment the antibody is administered at a certain, fixed, concentration, i.e. without taking into account a patient's body weight. In certain embodiments, the antibody of the present invention is administered at a fixed dose of between 40 mg and 400 mg, optionally at a fixed dose of 75 mg, at a fixed dose of 100 mg, at a fixed dose of 140 mg, at a fixed dose of 150 mg, at a fixed dose of 180 mg, at a fixed dose of 200 mg, at a fixed dose of 280 mg, at a fixed dose of 300 mg or at a fixed dose of 400 mg. Administration of fixed doses may be every week, every second week, every third week, every fourth week or every sixth week. Typically, the antibody will be administered weekly at a fixed dose.
  • In an embodiment, the antibody will be administered weekly, at a fixed subcutaneous dose of 40, 56, 70, 75 100, 140, 150, 180, 200, 210, or 280 mg.
  • In an embodiment, the antibody will be administered biweekly, at a fixed subcutaneous dose of 70, 75, 100, 140, 150, 180, 200, 210, 280 or 300 mg.
  • In an embodiment, the antibody will be administered monthly, at a fixed subcutaneous dose of 100, 140, 150, 180, 200, 210, 280, 300, 320, 350. 360 or 400 mg.
  • In an embodiment, the antibody is administered in a dose sufficient to maintain trough concentration of antibody of at least 2 ug/mL. The trough concentration of antibody may be maintained at 2.0 ug/mL, 2.5 ug/mL, 3.0 ug/mL, 3.5 ug/mL, 4.0 ug/mL, 4.5 ug/mL or 5.0 ug/mL, during the course of therapy.
  • In alternative embodiments, the antibody will be administered weekly, at a fixed subcutaneous dose of 28 or 35 mg.
  • In certain embodiments, the present invention provides an anti-GM-CSF antibody for use in the treatment of a patient suffering from rheumatoid arthritis, wherein said antibody is administered to said patient in a manner to achieve a therapeutically effective antibody level in the blood of said patient equal or higher compared to the intravenous administration of said antibody at a dose of at least 1.0 mg/kg when administered weekly over at least four weeks.
  • In certain embodiments, the present invention provides a method to treat a patient suffering from rheumatoid arthritis, said method comprising administering to said patient an anti-GM-CSF antibody in a manner to achieve a therapeutically effective antibody level in the blood of said patient equal or higher compared to the intravenous administration of said antibody at a dose of at least 1.0 mg/kg when administered weekly over at least four weeks.
  • In certain embodiments, the present invention provides an anti-GM-CSF antibody for inhibiting progression of structural joint damage in a rheumatoid arthritis patient comprising administering to said patient said antibody in a manner to achieve a therapeutically effective antibody level in the blood of said patient equal or higher compared to the intravenous administration of said antibody at a dose of at least 1.0 mg/kg when administered weekly over at least four weeks.
  • The terms “drug” and “medicament” refer to an active drug to treat rheumatoid arthritis or joint damage or symptoms or side effects associated with RA. The term “pharmaceutical formulation” refers to a preparation which is in such form as to permit the biological activity of the active ingredient or ingredients, i.e. the antibody of the present invention, to be effective, and which contains no additional components which are unacceptably toxic to a subject to which the formulation would be administered. Such formulations are sterile.
  • The antibody herein is preferably recombinantly produced in a host cell transformed with nucleic acid sequences encoding its heavy and light chains (e.g. where the host cell has been transformed by one or more vectors with the nucleic acid therein). The preferred host cell is a mammalian cell, most preferably a PER.C6 cell.
  • Therapeutic formulations of the antibody of the present invention are prepared for storage by mixing the antibody having the desired degree of purity with optional pharmaceutically acceptable carriers, excipients or stabilizers in the form of lyophilized formulations or aqueous solutions. Acceptable carriers, excipients, or stabilizers are nontoxic to recipients at the dosages and concentrations employed, and include buffers such as phosphate, citrate, histidine and other organic acids; antioxidants including ascorbic acid and methionine; preservatives (such as octadecyldimethylbenzyl ammonium chloride; hexamethonium chloride; benzalkonium chloride, benzethonium chloride; phenol, butyl or benzyl alcohol; alkyl parabens such as methyl or propyl paraben; catechol; resorcinol; cyclohexanol; 3-pentanol; and m-cresol); low molecular weight (less than about 10 residues) polypeptides; proteins, such as serum albumin, gelatin, or immunoglobulins; hydrophilic polymers such as polyvinylpyrrolidone; amino acids such as glycine, glutamine, asparagine, histidine, arginine, or lysine; monosaccharides, disaccharides, and other carbohydrates including glucose, mannose, or dextrins; chelating agents such as EDTA; sugars such as sucrose, mannitol, trehalose or sorbitol; salt-forming counter-ions such as sodium; metal complexes {e.g. Zn-protein complexes); and/or non-ionic surfactants such as TWEEN™ (such as Tween-80), PLURONICS™ or polyethylene glycol (PEG).
  • In certain embodiments, the present invention provides a pharmaceutical composition comprising an antibody of the present invention and a pharmaceutically acceptable carrier and/or excipient for use in any of the methods provided in the present invention. In certain embodiments, the formulation for the antibody of the present invention consists of 30 mM histidine, pH 6.0, 200 mM sorbitol and 0.02% Tween-80. In other embodiments, the formulation for the antibody of the present invention consists of PBS, pH 6.2 (0.2 g/l KCl, 0.96 g/l KH2PO4, 0.66 g/l Na2HPO4×7H2O, 8 g/l NaCl).
  • EXAMPLES Example 1: Design and Concept of a Clinical Phase Lb/Phase Ila Trial
  • A multi-center, randomized, double-blinded, placebo-controlled study to evaluate the safety, preliminary clinical activity and immunogenicity of multiple doses of MOR103 administered intravenously to patients with active rheumatoid arthritis was conducted.
  • Primary outcome measures were the adverse event rate and the safety profile. Secondary outcome measures included DAS28 scores, ACR scores and EULAR28 response criteria.
  • The clinical trial comprised three treatment arms. In each treatment arm patient received either placebo or MOR103. The MOR103 doses were 0.3 mg/kg body weight for treatment arm 1, 1.0 mg/kg body weight for treatment arm 2 and 1.5 mg/kg body weight for treatment arm 3. MOR103 and placebo were administered intravenously, weekly with 4 doses in total.
  • Summary of the treatment arms:
  • Arms Assigned Interventions
    Experimental: Group 1: Drug: MOR103
    MOR103, experimental MOR103 0.3 mg/kg or
    Biological: MOR103 0.3 mg/kg placebo iv × 4 doses
    or placebo
    Experimental: Group 2: Drug: MOR103
    MOR103, experimental MOR103 1.0 mg/kg or
    Biological: MOR103 1.0 mg/kg placebo iv × 4 doses
    or placebo
    Experimental: Group 3: Drug: MOR103
    MOR103, experimental MOR103 1.5 mg/kg or
    Biological: MOR103 1.5 mg/kg placebo iv × 4 doses
    or placebo
  • Eligible for participation in the study were patients of 18 years and older and of either sex (male and female). Healthy volunteers were not accepted.
  • Inclusion criteria were as follows:
      • Rheumatoid arthritis (RA) per revised 1987 ACR criteria
      • Active RA: ≥3 swollen and 3 tender joints with at least 1 swollen joint in the hand, excluding the PIP joints
      • CRP>5.0 mg/L (RF and anti-CCP seronegative); CRP>2 mg/l (RF and/or anti-CCP seropositive)
      • DAS28≤5.1
      • Stable regimen of concomitant RA therapy (NSAIDs, steroids, non-biological DMARDs).
      • Negative PPD tuberculin skin test
  • Exclusion criteria were as follows:
      • Previous therapy with B or T cell depleting agents other than Rituximab (e.g. Campath). Prior treatment with Rituximab, TNF-inhibitors, other biologics (e.g. anti-IL-1 therapy) and systemic immunosuppressive agents is allowed with a washout period.
      • Any history of ongoing, significant or recurring infections
      • Any active inflammatory diseases other than RA
      • Treatment with a systemic investigational drug within 6 months prior to screening
      • Women of childbearing potential, unless receiving stable doses of methotrexate or leflunomide
      • Significant cardiac or pulmonary disease (including methotrexate-associated lung toxicity)
      • Hepatic or renal insufficiency
    Example 2: Patient Recruitment and Patient Population
  • Clinical sites for patient recruitment, screening and treatment were located in Bulgaria, Germany, the Netherlands, Poland and the Ukraine.
  • 96 patients were included in the trial. 27 patients received placebo, 24 patients received MOR103 at a dose of 0.3 mg/kg, 22 patients received MOR103 at a dose of 1.0 mg/kg and 23 patients received MOR103 at a dose of 1.5 mg/kg. The average age and the average Body Mass Index (BMI) was about the same for all treatment groups. Key characteristics are summarized in the following Table:
  • MOR 103 Active Treatment Groups
    0.3 1.0 1.5 Total
    Placebo mg/kg mg/kg mg/kg active
    Characteristic N = 27 N = 24 N = 22 N = 23 N = 69
    Age 53.8 57.4 49 53 53.3
    BMI 26.3 26.3 26.1 25.7 26.0
    Gender-Female 19 (70%) 21 (88%) 17 (77%) 18 (78%) 56 (81%)
    White 27 24 22 23 69
  • 90% of all patients of the study were previously treated with DMARDs. The most commonly used DMARD was methotrexate (75% of all patients). The rate of previous treatment with DMARDs was comparable in all treatment arms.
  • Prior to administration of MOR103 or the placebo the disease activity of all patients was measured according to accepted guidelines by calculating the DAS28 score, a 28-joint Disease Activity Score (see e.g. Ann Rheum Dis (2009) 68, 954-60). DAS28 score is a validated and commonly used tool to quantify the disease status of RA patients. The average DAS28 score was comparable for all treatment arms.
  • Example 3: Safety Profile
  • Based on the available observed safety data, MOR103 showed a favorable safety profile among all doses tested. The key observations are as follows:
      • No deaths were observed during the conduct of the trial
      • No infusion related reactions were observed
      • Two serious adverse events (SAEs) were observed:
        • One patient in the placebo group developed paronychia
        • One patient in the 0.3 mg/kg treatment arm developed pleurisy
      • More treatment-emergent adverse effects (TEAEs) were observed in the placebo group (25.9%) than in the active groups (14.5%)
      • Most TEAEs were mild
      • No severe TEAEs were observed in the active groups
  • In summary, it can be concluded that treatment with MOR103 at all doses tested is safe. Two serious adverse events were observed, both none in the treatment arms that showed clinical efficacy (see below). Sub-cutaneous administration of MOR103 at a dose that leads to an antibody drug level in the blood of patients equivalent to the intravenous application of the present study is expected to show a similar safety profile.
  • Example 4: Efficacy—DAS28
  • 4 weeks and 8 weeks after the first administration of MOR103 (or placebo) the DAS28 scores of all patients was determined. A decrease in DAS28 scores correlates to diminished disease severity. Results are shown in FIG. 2 as the mean changes compared to baseline, i.e. disease status prior to treatment.
  • The placebo group only shows marginal changes. Patients treated with MOR103 at 0.3 mg/kg showed a slight decrease in DAS28 scores, indicating slightly less severity of the disease. In contrast, patients treated with MOR103 at 1.0 mg/kg or with 1.5 mg/kg showed a significant decrease in DAS28 scores, indicating the high efficacy of MOR103 at these doses.
  • Example 5: Efficacy—ACR20
  • As another measure of efficacy the ACR20 criteria were used. ACR criteria measure improvement in tender or swollen joint counts and improvement in certain other parameters. The procedure to measure ACR scores is highly standardized. The present clinical trial applied the respective applicable guidelines. Results are depicted in FIGS. 3 and 4. A higher score corresponds to an improvement in the severity of the disease.
  • In line with the results of the DAS28 scores (see Example 4), also the ACR scores show a strong clinical improvement of patients' condition upon treatment with either 1.0 mg/kg MOR103 or 1.5 mg/kg MOR103. The improvement after 4 weeks is highly significant for the 1.0 mg/kg group (p<0.0001). Taken together, the ACR20 scores confirm the surprising finding that the efficacy of MOR103 can already be shown with a comparably low number or patients in each treatment arm and a comparably short treatment period.
  • Example 6: Clinical Trial with Additional Doses of MOR103
  • The clinical trial set out herein above is repeated with additional doses of MOR103. MOR103 is administered to patients intravenously at a dose of 0.5 mg/kg (treatment arm 1) and 0.75 mg/kg (treatment arm 2). All other parameters are identical to Example 1.
  • Both treatment arms show a favorable safety profile and demonstrate clinical efficacy as measured by DAS28 scores and ACR20 scores.
  • Example 7: Clinical Trial with a Sub-Cutaneous Formulation of MOR103
  • The clinical trial set out herein above is repeated with a sub-cutaneous formulation of MOR103. In order to achieve similar levels of MOR103 in the blood of patients as observed for intravenous treatment, the sub-cutaneous dose of MOR103 is increased.
  • In different treatment arms MOR103 is administered to patients at 1.5 mg/kg, 2.0 mg/kg, 3.0 mg/kg and 4.0 mg/kg. The drug is administered sub-cutaneously, either biweekly, monthly or bimonthly. All other parameters are identical to Example 1.
  • All treatment arms show a favorable safety profile and demonstrate clinical efficacy as measured by DAS28 scores and ACR20 scores.
  • Example 8: Clinical Trial with a Sub-Cutaneous Formulation of MOR103 at a Fixed Dose
  • Example 7 is repeated with a fixed dose of MOR103. In different treatment arms MOR103 is administered to patients at fixed dose of 75 mg, of 100 mg, of 150 mg, of 200 mg, of 300 mg and of 400 mg. The drug is administered sub-cutaneously every week, every second week, every fourth week or every sixth week. All other parameters are identical to the Examples described herein above.
  • All treatment arms show a favorable safety profile and demonstrate clinical efficacy as measured by DAS28 scores and ACR20 scores.

Claims (19)

1.-20. (canceled)
21. A pharmaceutical composition comprising:
(a) an anti-Granulocyte-Macrophage Colony Stimulating Factor (anti-GM-CSF) antibody;
(b) histidine;
(c) sorbitol; and
(d) Tween-80,
wherein said anti-GM-CSF antibody comprises the HCDR1 region of sequence GFTFSSYWMN (SEQ ID NO.: 2), the HCDR2 region of sequence GIENKYAGGATYYAASVKG (SEQ ID NO.: 3), the HCDR3 region of sequence GFGTDF (SEQ ID NO.: 4), the LCDR1 region of sequence SGDSIGKKYAY (SEQ ID NO.: 5), the LCDR2 region of sequence KKRPS (SEQ ID NO.: 6), and the LCDR3 region of sequence SAWGDKGM (SEQ ID NO.: 7).
22. The pharmaceutical composition according to claim 21, wherein said anti-GM-CSF antibody comprises the variable heavy chain peptide of sequence:
(SEQ ID NO: 8) QVQLVESGGGLVQPGGSLRLSCAASGFTFSSYWMNWVRQAPGKGLEWVSG IENKYAGGATYYAASVKGRFTISRDNSKNTLYLQMNSLRAEDTAVYYCAR GFGTDFWGQGTLVTVSS;
and the variable light chain peptide of sequence:
(SEQ ID NO: 9) DIELTQPPSVSVAPGQTARISCSGDSIGKKYAYWYQQKPGQAPVLVIYKK RPSGIPERFSG SNSGNTATLTISGTQAEDEADYYCSAWGDKGMVFGGGT KLTVLGQ.
23. The pharmaceutical composition according to claim 21, comprising:
(b) 30 mM histidine;
(c) 200 mM sorbitol; and
(d) 0.02% Tween-80.
24. The pharmaceutical composition according to claim 23, comprising 40 mg to 400 mg of the anti-GM-CSF antibody.
25. The pharmaceutical composition according to claim 23, comprising 75 mg, 100 mg, 150 mg, 180 mg, 200 mg, 300 mg, or 400 mg of the anti-GM-CSF antibody.
26. The pharmaceutical composition according to claim 23, comprising 75 mg of the anti-GM-CSF antibody.
27. The pharmaceutical composition according to claim 23, comprising 100 mg of the anti-GM-CSF antibody.
28. The pharmaceutical composition according to claim 23, comprising 150 mg of the anti-GM-CSF antibody.
29. The pharmaceutical composition according to claim 23, comprising 180 mg of the anti-GM-CSF antibody.
30. The pharmaceutical composition according to claim 23, wherein the composition has a pH of 6.0.
31. The pharmaceutical composition according to claim 22, comprising:
(b) 30 mM histidine;
(c) 200 mM sorbitol; and
(d) 0.02% Tween-80.
32. The pharmaceutical composition according to claim 31, comprising 40 mg to 400 mg of the anti-GM-CSF antibody.
33. The pharmaceutical composition according to claim 31, comprising 75 mg, 100 mg, 150 mg, 180 mg, 200 mg, 300 mg, or 400 mg of the anti-GM-CSF antibody.
34. The pharmaceutical composition according to claim 31, comprising 75 mg of the anti-GM-CSF antibody.
35. The pharmaceutical composition according to claim 31, comprising 100 mg of the anti-GM-CSF antibody.
36. The pharmaceutical composition according to claim 31, comprising 150 mg of the anti-GM-CSF antibody.
37. The pharmaceutical composition according to claim 31, comprising 180 mg of the anti-GM-CSF antibody.
38. The pharmaceutical composition according to claim 31, wherein the composition has a pH of 6.0.
US17/144,685 2012-09-20 2021-01-08 Treatment for rheumatoid arthritis Pending US20210130451A1 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
US17/144,685 US20210130451A1 (en) 2012-09-20 2021-01-08 Treatment for rheumatoid arthritis

Applications Claiming Priority (7)

Application Number Priority Date Filing Date Title
EP12185235 2012-09-20
EP12185235.4 2012-09-20
US201261703871P 2012-09-21 2012-09-21
PCT/EP2013/069501 WO2014044768A1 (en) 2012-09-20 2013-09-19 Treatment for rheumatoid arthritis
US201514429996A 2015-03-20 2015-03-20
US15/880,726 US10913792B2 (en) 2012-09-20 2018-01-26 Treatment for rheumatoid arthritis
US17/144,685 US20210130451A1 (en) 2012-09-20 2021-01-08 Treatment for rheumatoid arthritis

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
US15/880,726 Division US10913792B2 (en) 2012-09-20 2018-01-26 Treatment for rheumatoid arthritis

Publications (1)

Publication Number Publication Date
US20210130451A1 true US20210130451A1 (en) 2021-05-06

Family

ID=46963526

Family Applications (3)

Application Number Title Priority Date Filing Date
US14/429,996 Abandoned US20150246969A1 (en) 2012-09-20 2013-09-19 Treatment for rheumatoid arthritis
US15/880,726 Active US10913792B2 (en) 2012-09-20 2018-01-26 Treatment for rheumatoid arthritis
US17/144,685 Pending US20210130451A1 (en) 2012-09-20 2021-01-08 Treatment for rheumatoid arthritis

Family Applications Before (2)

Application Number Title Priority Date Filing Date
US14/429,996 Abandoned US20150246969A1 (en) 2012-09-20 2013-09-19 Treatment for rheumatoid arthritis
US15/880,726 Active US10913792B2 (en) 2012-09-20 2018-01-26 Treatment for rheumatoid arthritis

Country Status (23)

Country Link
US (3) US20150246969A1 (en)
EP (3) EP3916013A1 (en)
JP (3) JP2015533806A (en)
KR (3) KR20230041086A (en)
CN (2) CN109999195A (en)
AU (2) AU2013320261A1 (en)
BR (1) BR112015006189B1 (en)
CA (1) CA2884124A1 (en)
CL (2) CL2015000696A1 (en)
CR (1) CR20150153A (en)
DO (1) DOP2015000070A (en)
EA (1) EA031489B1 (en)
HK (1) HK1208231A1 (en)
IL (1) IL237554B (en)
IN (1) IN2015KN00657A (en)
MA (1) MA37946B1 (en)
MX (2) MX2015003644A (en)
MY (1) MY175388A (en)
PE (1) PE20151079A1 (en)
PH (1) PH12015500591A1 (en)
SG (2) SG11201501595YA (en)
UA (1) UA117228C2 (en)
WO (1) WO2014044768A1 (en)

Families Citing this family (13)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
SG11201501595YA (en) * 2012-09-20 2015-05-28 Morphosys Ag Treatment for rheumatoid arthritis
AR093297A1 (en) * 2012-10-31 2015-05-27 Amgen Res (Munich) Gmbh LIQUID FORMULATION THAT INCLUDES A GM-CSF NEUTRALIZING COMPOUND
WO2014068029A1 (en) * 2012-10-31 2014-05-08 Takeda Gmbh Lyophilized formulation comprising gm-csf neutralizing compound
ES2860480T3 (en) * 2013-08-30 2021-10-05 Takeda Gmbh Neutralizing antibodies to GM-CSF for use in the treatment of rheumatoid arthritis or as analgesics
US11299528B2 (en) * 2014-03-11 2022-04-12 D&D Pharmatech Inc. Long acting TRAIL receptor agonists for treatment of autoimmune diseases
US11173208B2 (en) * 2014-05-07 2021-11-16 Takeda Gmbh Liquid formulation comprising GM-CSF neutralizing compound
GB201519331D0 (en) * 2015-11-02 2015-12-16 Glaxosmithkline Ip Dev Ltd Treatment paradigm
AU2016371021B2 (en) 2015-12-17 2020-04-09 The Johns Hopkins University Ameliorating systemic sclerosis with death receptor agonists
EA201892260A1 (en) 2016-04-07 2019-03-29 Дзе Джонс Хопкинс Юниверсити COMPOSITIONS AND METHODS FOR THE TREATMENT OF PANCREATITIS AND PAIN WITH THE APPLICATION OF THE AGONISTS OF THE DEATH RECEPTOR
PT3328887T (en) 2016-09-19 2021-10-19 I Mab Biopharma Hangzhou Co Ltd Anti-gm-csf antibodies and uses thereof
US11655293B2 (en) 2018-02-22 2023-05-23 Universitat Zurich Ligands to GM-CSF or GM-CSF-receptor for use in leukemia in a patient having undergone allo-HCT
EP3623382A1 (en) 2018-09-14 2020-03-18 Universität Zürich Ligands to gm-csf or gm-csf-receptor for use in leukemia in a patient having undergone allo-hct
WO2021204649A1 (en) 2020-04-06 2021-10-14 Glaxosmithkline Intellectual Property Development Limited Gm-csf antagonists for use in the treatment of severe pulmonary covid-19, cytokine release syndrome and/or acute respiratory distress syndrome

Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20090053213A1 (en) * 2005-05-18 2009-02-26 Morphosys Ag Anti-Gm-Csf Antibodies and Uses Therefor
US10913792B2 (en) * 2012-09-20 2021-02-09 Morphosys Ag Treatment for rheumatoid arthritis

Family Cites Families (21)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6267958B1 (en) 1995-07-27 2001-07-31 Genentech, Inc. Protein formulation
AU4344697A (en) 1996-10-04 1998-04-24 Amgen, Inc. Pharmaceutical compositions containing an mpl ligand
US7455836B2 (en) 2000-05-08 2008-11-25 The University Of Melbourne Method of treatment and agents useful for same
US7790679B2 (en) 2005-08-05 2010-09-07 Amgen Inc. Pharmaceutical formulations
US9399061B2 (en) * 2006-04-10 2016-07-26 Abbvie Biotechnology Ltd Methods for determining efficacy of TNF-α inhibitors for treatment of rheumatoid arthritis
US8093360B2 (en) * 2006-09-28 2012-01-10 Elusys Therapeutics, Inc. Antibodies that bind B. anthracis exotoxin, formulations thereof, and methods of use
BRPI0719109A2 (en) * 2006-11-21 2013-12-10 Kalobios Pharmaceuticals Inc METHODS TO TREAT CHRONIC INFLAMMATORY DISEASES USING A GM-CSF ANTAGONIST
CA2682170A1 (en) * 2007-03-30 2008-10-09 Medimmune, Llc Antibodies with decreased deamidation profiles
AU2008232506A1 (en) * 2007-04-02 2008-10-09 Genentech, Inc. Biological markers predictive of rheumatoid arthritis response to B-cell antagonists
SI2142174T1 (en) 2007-04-05 2011-04-29 Sandoz Ag Stable aqueous g-csf formulations
US20090004189A1 (en) * 2007-06-18 2009-01-01 Genentech, Inc. Biological markers predictive of rheumatoid arthritis response to b-cell antagonists
TW200918553A (en) * 2007-09-18 2009-05-01 Amgen Inc Human GM-CSF antigen binding proteins
CA2705539A1 (en) 2007-11-13 2009-05-22 Evec Inc. Monoclonal antibodies that bind to hgm-csf and medical compositions comprising same
EP2196476A1 (en) * 2008-12-10 2010-06-16 Novartis Ag Antibody formulation
CN108014335A (en) * 2008-12-22 2018-05-11 墨尔本大学 The purposes of the antagonist of granulocyte macrophage colony stimulating factor
CN106397591A (en) 2008-12-22 2017-02-15 墨尔本大学 Osteoarthritis treatment
JP2010241718A (en) 2009-04-03 2010-10-28 Kyowa Hakko Kirin Co Ltd Stable aqueous solution preparation of antibody
KR20140064943A (en) * 2009-05-05 2014-05-28 모르포시스 아게 Treatment for multiple sclerosis
PT2341061E (en) 2009-12-31 2013-11-05 Arven Ilac Sanayi Ve Ticaret As A novel process for preparing g-csf (granulocyte colony stimulating factor)
MX347504B (en) * 2010-03-01 2017-04-28 Cytodyn Inc Concentrated protein formulations and uses thereof.
KR20140061379A (en) 2011-07-06 2014-05-21 모르포시스 아게 Therapeutic combinations of anti-cd20 and anti-gm-csf antibodies and uses thereof

Patent Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20090053213A1 (en) * 2005-05-18 2009-02-26 Morphosys Ag Anti-Gm-Csf Antibodies and Uses Therefor
US10913792B2 (en) * 2012-09-20 2021-02-09 Morphosys Ag Treatment for rheumatoid arthritis

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
Wang et al. "Antibody Structure, Instability, and Formulation". JOURNAL OF PHARMACEUTICAL SCIENCES, VOL. 96, NO. 1, JANUARY 2007. (Year: 2007) *

Also Published As

Publication number Publication date
EA201590359A1 (en) 2015-08-31
EP2897977A1 (en) 2015-07-29
JP2018138551A (en) 2018-09-06
EP3345923A1 (en) 2018-07-11
KR20150056846A (en) 2015-05-27
SG10201803778PA (en) 2018-06-28
BR112015006189B1 (en) 2022-04-05
MX2015003644A (en) 2015-09-25
MX2021006111A (en) 2021-07-07
WO2014044768A1 (en) 2014-03-27
US20180230208A1 (en) 2018-08-16
PH12015500591B1 (en) 2015-05-11
BR112015006189A2 (en) 2019-11-26
CN109999195A (en) 2019-07-12
CR20150153A (en) 2015-09-14
AU2016250388A1 (en) 2016-11-17
MY175388A (en) 2020-06-23
SG11201501595YA (en) 2015-05-28
US20150246969A1 (en) 2015-09-03
KR20230041086A (en) 2023-03-23
IL237554B (en) 2019-12-31
AU2016250388B2 (en) 2018-07-19
CL2018000933A1 (en) 2018-07-20
CL2015000696A1 (en) 2015-10-23
CN104995210A (en) 2015-10-21
PE20151079A1 (en) 2015-08-07
UA117228C2 (en) 2018-07-10
PH12015500591A1 (en) 2015-05-11
US10913792B2 (en) 2021-02-09
JP2015533806A (en) 2015-11-26
MA37946B1 (en) 2018-09-28
KR20220028177A (en) 2022-03-08
EP3916013A1 (en) 2021-12-01
EA031489B1 (en) 2019-01-31
JP2022169709A (en) 2022-11-09
IL237554A0 (en) 2015-04-30
MA37946A1 (en) 2018-01-31
IN2015KN00657A (en) 2015-07-17
AU2013320261A1 (en) 2015-04-09
CA2884124A1 (en) 2014-03-27
HK1208231A1 (en) 2016-02-26
DOP2015000070A (en) 2015-08-16

Similar Documents

Publication Publication Date Title
US20210130451A1 (en) Treatment for rheumatoid arthritis
US20200231666A1 (en) Treatment paradigm
JP6554155B2 (en) Methods of treating generalized pustular psoriasis (GPP) using an IL-17 antagonist
US20170369583A1 (en) Methods and compositions for treating lupus
JP2024001125A (en) Methods of treating new-onset plaque type psoriasis using il-17 antagonists
US20230203149A1 (en) Treatment of atopic dermatitis
JP7132256B2 (en) Treatment of rheumatoid arthritis
US20110275791A1 (en) A B Cell Depleting Agent for the Treatment of Atherosclerosis
NZ744721A (en) Treatment for rheumatoid arthritis
NZ744721B2 (en) Treatment for rheumatoid arthritis
EP4139353A1 (en) Pan-elr+ cxc chemokine antibodies for the treatment of respiratory disease
JP2022523530A (en) Anti-IL-α antibody for the treatment of hidradenitis suppurativa

Legal Events

Date Code Title Description
STPP Information on status: patent application and granting procedure in general

Free format text: APPLICATION DISPATCHED FROM PREEXAM, NOT YET DOCKETED

STPP Information on status: patent application and granting procedure in general

Free format text: DOCKETED NEW CASE - READY FOR EXAMINATION

STPP Information on status: patent application and granting procedure in general

Free format text: NON FINAL ACTION MAILED