US20170342139A1 - Methods for Treatment of Refractory Generalized Myasthenia Gravis - Google Patents

Methods for Treatment of Refractory Generalized Myasthenia Gravis Download PDF

Info

Publication number
US20170342139A1
US20170342139A1 US15/595,890 US201715595890A US2017342139A1 US 20170342139 A1 US20170342139 A1 US 20170342139A1 US 201715595890 A US201715595890 A US 201715595890A US 2017342139 A1 US2017342139 A1 US 2017342139A1
Authority
US
United States
Prior art keywords
eculizumab
patient
treatment
weeks
administering
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Abandoned
Application number
US15/595,890
Other languages
English (en)
Inventor
Camille Bedrosian
Fanny O'Brien
Jing Jing Wang
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.)
Alexion Pharmaceuticals Inc
Original Assignee
Alexion Pharmaceuticals Inc
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Alexion Pharmaceuticals Inc filed Critical Alexion Pharmaceuticals Inc
Priority to US15/595,890 priority Critical patent/US20170342139A1/en
Assigned to ALEXION PHARMACEUTICALS, INC. reassignment ALEXION PHARMACEUTICALS, INC. ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: BEDROSIAN, CAMILLE, WANG, JING JING, O'BRIEN, Fanny
Publication of US20170342139A1 publication Critical patent/US20170342139A1/en
Priority to US17/150,005 priority patent/US20210246196A1/en
Abandoned legal-status Critical Current

Links

Images

Classifications

    • 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
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61MDEVICES FOR INTRODUCING MEDIA INTO, OR ONTO, THE BODY; DEVICES FOR TRANSDUCING BODY MEDIA OR FOR TAKING MEDIA FROM THE BODY; DEVICES FOR PRODUCING OR ENDING SLEEP OR STUPOR
    • A61M1/00Suction or pumping devices for medical purposes; Devices for carrying-off, for treatment of, or for carrying-over, body-liquids; Drainage systems
    • A61M1/34Filtering material out of the blood by passing it through a membrane, i.e. hemofiltration or diafiltration
    • A61M1/3496Plasmapheresis; Leucopheresis; Lymphopheresis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P21/00Drugs for disorders of the muscular or neuromuscular system
    • A61P21/04Drugs for disorders of the muscular or neuromuscular system for myasthenia gravis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00
    • 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
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/55Medicinal preparations containing antigens or antibodies characterised by the host/recipient, e.g. newborn with maternal antibodies
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/20Immunoglobulins specific features characterized by taxonomic origin
    • C07K2317/24Immunoglobulins specific features characterized by taxonomic origin containing regions, domains or residues from different species, e.g. chimeric, humanized or veneered

Definitions

  • MG Myasthenia Gravis
  • NMJ neuromuscular junction
  • Abs auto-antibodies
  • proteins include the nicotine acetylcholine receptors (AChRs) or, less frequently, a muscle-specific tyrosine kinase (MuSK) involved in AChR clustering.
  • MG has a prevalence of 14-20 per 100,000 in the U.S., affecting roughly 60,000 Americans. It affects males and females in equal ratio, although the incidence in females peaks in the 3rd decade as compared to males in whom the peak age at onset is in the 6th or 7th decade. Mortality from MG is approximately 4%, mostly due to respiratory failure.
  • Myasthenia gravis is clinically characterized by weakness and fatigability of voluntary skeletal muscles.
  • MG may initially present with ocular muscle weakness affecting eye and eyelid movement, referred to as ocular MG (oMG).
  • oMG ocular MG
  • Ten percent of subjects have disease limited to ocular muscles.
  • Ninety percent of subjects have generalized MG, with muscle weakness involving neck, head, spine, bulbar, respiratory, or limb muscles.
  • Bulbar weakness refers to muscles controlled by nerves originating from the bulb-like part of the brainstem and manifests as difficulty in talking, chewing, swallowing, and control of the head.
  • MG may cause life-threatening respiratory failure, referred to as myasthenic crisis.
  • myasthenic crisis About 15% to 20% of subjects will experience a myasthenic crisis during the course of their disease, 75% within 2 years of diagnosis, requiring hospitalization and ventilatory support.
  • MG While there is no cure for MG, there are a variety of therapies that reduce muscle weakness and improve neuromuscular function.
  • Current available treatments for myasthenia gravis aim to modulate neuromuscular transmission, inhibit the production or effects of pathogenic antibodies, or inhibit inflammatory cytokines.
  • ISTs immunosuppressive therapies
  • AZA azathioprine
  • MMF mycophenolate mofetil
  • PE plasma exchange
  • IVIg intravenous immunoglobulin
  • This disclosure provides methods of treating refractory generalized myasthenia gravis in a patient in need thereof comprising administering a therapeutically effective amount of an anti-complement component 5 (C5) antibody or an antigen binding fragment thereof to the patient, wherein the patient is administered the anti-C5 antibody or antigen binding fragment thereof for at least 26 weeks.
  • C5 anti-complement component 5
  • this disclosure provides a method of treating refractory generalized myasthenia gravis in a patient in need thereof comprising administering a therapeutically effective amount of an anti-C5 antibody or an antigen binding fragment thereof to the patient, wherein the anti-C5 antibody, or an antigen binding fragment thereof is eculizumab or an eculizumab variant and wherein the patient is administered eculizumab or eculizumab variant for at least 26 weeks.
  • this disclosure provides a method comprising administering a therapeutically effective amount of eculizumab to a patient, wherein the patient is positive for auto-antibodies binding to nicotinic acetylcholine receptor (anti-AChR) and shows marked generalized weakness or bulbar signs and symptoms of myasthenia gravis while receiving therapy for myasthenia gravis including anticholinesterase inhibitor therapy and immunosuppressant therapy (IST) and requires chronic plasma exchange or chronic IVIg to maintain clinical stability; and wherein the patient is administered eculizumab for at least 26 weeks.
  • anti-AChR nicotinic acetylcholine receptor
  • this disclosure provides a method of treating refractory generalized myasthenia gravis in a patient in need thereof comprising administering eculizumab to the patient, wherein the patient is positive for auto-antibodies binding to nicotinic acetylcholine receptor (anti-AChR) and shows marked generalized weakness or bulbar signs and symptoms of myasthenia gravis while receiving therapy for myasthenia gravis including anticholinesterase inhibitor therapy and immunosuppressant therapy (IST) and had previously failed treatment with at least two immunosuppressive agents or failed treatment with at least one immunosuppressive agent and required chronic plasma exchange or IVIg, and had an MG-ADL total score ⁇ 6 at study entry; wherein eculizumab is administered using a phased dosing schedule with an induction phase comprising administering a 900 mg induction dose of eculizumab on day 1, administering 900 mg doses of eculizumab on days 7, 14, and 21, and administer
  • this disclosure provides a method of treating refractory generalized myasthenia gravis in a patient in need thereof comprising administering eculizumab to the patient, wherein the patient is positive for auto-antibodies binding to nicotinic acetylcholine receptor (anti-AChR) and shows marked generalized weakness or bulbar signs and symptoms of myasthenia gravis while receiving therapy for myasthenia gravis including anticholinesterase inhibitor therapy and immunosuppressant therapy (IST) or requires chronic plasma exchange or chronic IVIg to maintain clinical stability; wherein eculizumab is administered using a phased dosing schedule with an induction phase comprising administering a 900 mg induction dose of eculizumab on day 1, administering 900 mg doses of eculizumab on days 7, 14, and 21, and administering 1200 mg of eculizumab as a fifth induction dose on day 28, and wherein the patient is administered eculizumab
  • this disclosure provides a method wherein the 28 day induction phase of eculizumab treatment is followed by a maintenance phase comprising administering 1200 mg of eculizumab 14 days after the fifth induction dose and 1200 mg of eculizumab every 14 ⁇ 2 days thereafter.
  • this disclosure provides a method wherein the dosing regimen further comprises a third phase and wherein the third phase comprises performing plasmapheresis on the patient and administering eculizumab at a dose of between 300 and 1200 mg to the patient within 4 hours of completion of plasmapheresis.
  • the third phase comprises performing plasmapheresis on the patient and administering eculizumab at a dose of between 600 and 900 mg to the patient within 90 minutes of completion of plasmapheresis.
  • the third phase comprises performing plasmapheresis on the patient and administering eculizumab at a dose of 600 mg to the patient within 1 hour of completion of plasmapheresis.
  • the patient being treated by the methods provided herein experiences a clinically meaningful improvement (reduction) in Myasthenia Gravis Activities of Daily Living (MG-ADL) score after 26 weeks of treatment.
  • the clinically meaningful improvement the patient experiences is at least a 3 point reduction in the patient's MG-ADL score after 26 weeks of treatment.
  • the clinically meaningful improvement the patient experiences is at least a 4 point reduction in the patient's MG-ADL score after 26 weeks of treatment.
  • the patient being treated by the methods provided herein experiences a clinically meaningful improvement (reduction) in quantitative Myasthenia Gravis score (QMG) after 26 weeks of treatment.
  • the clinically meaningful improvement the patient experiences is at least a 4 point reduction in the patient's QMG score after 26 weeks of treatment.
  • the clinically meaningful improvement the patient experiences is a 5 point reduction in the patient's QMG score after 26 weeks of treatment.
  • the patient being treated by the methods provided herein experiences a clinically meaningful improvement (reduction) in Myasthenia Gravis Composite (MGC) score after 26 weeks of treatment.
  • the clinically meaningful improvement the patient experiences is at least a 6 point reduction in the patient's MGC score after 26 weeks of treatment.
  • the clinically meaningful improvement the patient experiences is at least a 10 point reduction in the patient's MGC score after 26 weeks of treatment.
  • the patient being treated by the methods provided herein experiences a clinically meaningful improvement (reduction) in quality of life as measured by the Myasthenia Gravis Quality of Life (MG-QOL-15) score after 26 weeks of treatment.
  • the clinically meaningful improvement the patient experiences is at least a 6 point reduction in the patient's MG-QOL-15 score after 26 weeks of treatment.
  • the clinically meaningful improvement the patient experiences is at least an 11 point reduction in the patient's MG-QOL-15 score after 26 weeks of treatment.
  • the patient being treated by the methods provided herein experiences a clinically meaningful improvement (reduction) in neuro-fatigue as measured by the Neuro-QOL Fatigue score after 26 weeks of treatment.
  • the clinically meaningful improvement the patient experiences is at least an 8 point reduction in the patient's Neuro-QOL score after 26 weeks of treatment.
  • the clinically meaningful improvement the patient experiences is at least a 16 point reduction in the patient's Neuro-QOL score after 26 weeks of treatment.
  • the patient being treated by the methods provided herein experiences a clinically meaningful improvement (increase) in health status as measured by the EQ-5D health status score after 26 weeks of treatment.
  • this disclosure provides a method of treating refractory generalized myasthenia gravis in a patient in need thereof comprising administering eculizumab to the patient, wherein the patient is positive for auto-antibodies binding to nicotinic acetylcholine receptor (anti-AChR) and shows marked generalized weakness or bulbar signs and symptoms of myasthenia gravis while receiving therapy for myasthenia gravis including anticholinesterase inhibitor therapy and immunosuppressant therapy (IST) or requires chronic plasma exchange or chronic IVIg to maintain clinical stability; wherein eculizumab is administered using a phased dosing schedule comprising administering a 900 mg induction dose of eculizumab on day 1, administering 900 mg doses of eculizumab on days 7, 14, and 21, and administering 1200 mg of eculizumab as a fifth dose on day 28, wherein the patient is administered eculizumab for at least 26 weeks; wherein
  • this disclosure provides a method of treating refractory generalized myasthenia gravis in a patient in need thereof comprising administering eculizumab to the patient, wherein the patient is positive for auto-antibodies binding to nicotinic acetylcholine receptor (anti-AChR) and shows marked generalized weakness or bulbar signs and symptoms of myasthenia gravis while receiving therapy for myasthenia gravis including anticholinesterase inhibitor therapy and immunosuppressant therapy (IST) and requires chronic plasma exchange or chronic IVIg to maintain clinical stability; wherein eculizumab is administered using a phased dosing schedule comprising administering a 900 mg induction dose on day 1, administering 900 mg doses of eculizumab on days 7, 14, and 21, and administering 1200 mg of eculizumab as a fifth dose on day 28, wherein the patient is administered eculizumab for at least 26 weeks; wherein the 28 induction phase of eculi
  • this disclosure provides a method of treating refractory generalized myasthenia gravis in a patient in need thereof comprising administering eculizumab to the patient, wherein the patient is positive for auto-antibodies binding to nicotinic acetylcholine receptor (anti-AChR) and shows marked generalized weakness or bulbar signs and symptoms of myasthenia gravis while receiving therapy for myasthenia gravis including anticholinesterase inhibitor therapy and immunosuppressant therapy (IST) or requires chronic plasma exchange or chronic IVIg to maintain clinical stability; wherein eculizumab is administered using a phased dosing schedule comprising administering a 900 mg induction dose on day 1, administering 900 mg doses of eculizumab on days 7, 14, and 21, and administering 1200 mg as a fifth dose on day 28, wherein the patient is administered eculizumab for at least 26 weeks; wherein the 28 day induction phase of eculizumab treatment is followed by
  • the patient has a clinically meaningful improvement (reduction) in four measurements of generalized myasthenia gravis severity selected from the group consisting of MG-ADL, QMG, MGC, MG-QOL, and Neuro-QOL.
  • the patient has a clinically meaningful improvement (reduction) in five measurements of generalized myasthenia gravis severity, wherein the five measurements of generalized myasthenia gravis severity are MG-ADL, QMG, MGC, MG-QOL, and Neuro-QOL.
  • this disclosure provides a method of treating refractory generalized myasthenia gravis in a patient in need thereof comprising administering eculizumab to the patient, wherein the patient is positive for auto-antibodies binding to nicotinic acetylcholine receptor (anti-AChR) and shows marked generalized weakness or bulbar signs and symptoms of myasthenia gravis while therapy for myasthenia gravis including anticholinesterase inhibitor therapy and immunosuppressant therapy (IST) or requires chronic plasma exchange or chronic IVIg to maintain clinical stability; wherein eculizumab is administered using a phased dosing schedule comprising administering a 900 mg induction dose on day 1, administering 900 mg doses of eculizumab on days 7, 14, and 21, and administering 1200 mg as a fifth dose on day 28, wherein the patient is administered eculizumab for at least 26 weeks; wherein the 28 day induction phase of eculizumab treatment is followed by
  • the patient has a clinically meaningful improvement (reduction) in five measurements of generalized myasthenia gravis severity, wherein the five measurements of generalized myasthenia gravis severity are a reduction in MG-ADL of at least 4 points, a reduction in QMG of at least 5 points, a reduction in MGC of at least 10 points, a reduction in MG-QOL of at least 11 points, and a reduction in Neuro-QOL of at least 16 points.
  • the five measurements of generalized myasthenia gravis severity are a reduction in MG-ADL of at least 4 points, a reduction in QMG of at least 5 points, a reduction in MGC of at least 10 points, a reduction in MG-QOL of at least 11 points, and a reduction in Neuro-QOL of at least 16 points.
  • this disclosure provides a method of treating refractory generalized myasthenia gravis in a patient in need thereof comprising administering eculizumab by intravenous infusion.
  • eculizumab is administered subcutaneously.
  • the eculizumab comprises a heavy chain amino acid sequence according to SEQ ID NO: 10 and a light chain amino acid sequence according to SEQ ID NO: 11.
  • the eculizumab is an eculizumab variant comprising a heavy chain amino acid sequence according to SEQ ID NO: 14 and a light chain amino acid sequence according to SEQ ID NO: 11.
  • the eculizumab is an eculizumab variant comprising a heavy chain variable region amino acid sequence according to SEQ ID NO: 12 and a light chain amino acid sequence according to SEQ ID NO: 11.
  • this disclosure provides a method of treating refractory generalized myasthenia gravis in a patient in need thereof comprising administering an anti-C5 antibody, or antigen binding fragment thereof, wherein the antibody is an anti-C5 antibody or an antigen binding fragment thereof comprising a heavy chain variable region amino acid sequence according to SEQ ID NO: 27 and a light chain variable region amino acid sequence according to SEQ ID NO: 28.
  • the antibody is an anti-C5 antibody or an antigen binding fragment thereof comprising a heavy chain variable region amino acid sequence according to SEQ ID NO: 35 and a light chain variable region amino acid sequence according to SEQ ID NO: 36.
  • the antibody is an anti-C5 antibody or antigen binding fragment thereof comprising a heavy chain variable region amino acid sequence according to SEQ ID NO: 37 and a light chain variable region amino acid sequence according to SEQ ID NO: 38.
  • this disclosure provides a method of treating refractory generalized myasthenia gravis in a patient in need thereof comprising administering an anti-C5 antibody or antigen binding fragment thereof, wherein the patient has failed treatment over one year or more with two or more ISTs in sequence or in combination.
  • this disclosure provides a method of treating refractory generalized myasthenia gravis in a patient in need thereof comprising administering an anti-C5 antibody or antigen binding fragment thereof, wherein the patient has failed at least one IST and requires chronic plasma exchange or IVIg to control symptoms of myasthenia gravis.
  • this disclosure provides a method of treating refractory generalized myasthenia gravis in a patient in need thereof comprising administering a therapeutically effective amount of eculizumab is maintained at a concentration of between 50-100 pg/mL in the patient's serum.
  • this disclosure provides a method of treating refractory generalized myasthenia gravis in a patient in need thereof comprising administering a therapeutically effective amount of eculizumab, wherein the patient experiences a discontinuation in the administration of one or more IST following at least 26 weeks of treatment.
  • this disclosure provides a method of treating refractory generalized myasthenia gravis in a patient in need thereof comprising administering a therapeutically effective amount of eculizumab, wherein the patient experiences a reduction in IST dosing following at least 26 weeks of treatment.
  • FIG. 1 is a schematic of the overall design of the clinical trial disclosed herein.
  • FIGS. 2A and 2B are a schematic of the EUROQOL (EQ-5D) survey of health status questionnaire used in the clinical trial disclosed herein.
  • FIG. 3 is a schematic of the N. meningitidis vaccination schedule used in the clinical trial disclosed herein.
  • FIG. 4 is a schematic of the dosing schedule used in the clinical trial disclosed herein.
  • FIG. 5 is a schematic of the dosing, clinical evaluation and safety follow-up schedule, used in the clinical trial disclosed herein.
  • FIG. 6 is a schematic of the dosing schedule used in the clinical trial also including the extension period disclosed herein.
  • FIG. 7 is a graphical depiction of the changes from baseline in MG-ADL values obtained for placebo and eculizumab groups over the initial 26 weeks of the trial.
  • FIG. 8 is a graphical depiction of the changes from baseline in QMG values obtained for placebo and eculizumab groups over the initial 26 weeks of the trial.
  • FIG. 9 is a graphical depiction of the changes from baseline in MGC values obtained for placebo and eculizumab groups over the initial 26 weeks of the trial.
  • FIG. 10 is a graphical depiction of the changes from baseline in MG-QOL 15 values obtained for placebo and eculizumab groups over the initial 26 weeks of the trial.
  • FIG. 11 is a graphical depiction of the numbers of patients in both the placebo and eculizumab treated groups achieving between a 5 and 10 point reduction in QMG score over the initial 26 weeks of the trial.
  • FIG. 12 is a schematic of the REGAIN study design.
  • FIG. 13 is a graphical depiction of responder analyses (MG-ADL and QMG) illustrating the proportion of patients with improvement in total score and no rescue therapy at week 26 from baseline.
  • FIG. 14 is a graphical depiction of the proportion of patients with a ⁇ 3, ⁇ 5, or ⁇ 8-point reduction in MG-ADL total score and no rescue therapy over time from baseline to week 26.
  • FIG. 15 is a graphical depiction of the proportion of patients with ⁇ 5, ⁇ 7, or ⁇ 10-point reduction in QMG total score and no rescue over time from baseline to week 26.
  • FIG. 16 is a graphical depiction of dual responders (assessed by MG-ADL and QMG total scores) with no rescue therapy at week 26.
  • FIG. 17 is a graphical depiction of the proportion of patients with at least 3-point improvement in MG-ADL total score and ⁇ 5-point improvement in QMG total score and no rescue therapy assessed over time from baseline to week 26.
  • FIG. 18 is a graphical depiction of the percentage of patients who simultaneously met increasingly stringent criteria based on MG-ADL and QMG.
  • the bottom row describes a threshold for both scales above the MCID (minimal clinically meaningful difference: 3 for MG-ADL and 5 for QMG). Higher bars represent increasing thresholds.
  • the right-most panel displays odds ratios for meeting each threshold for eculizumab vs. placebo treated patients.
  • FIG. 19 is a graphical depiction of the change from baseline in MG-ADL total score (LS Mean and 95% CI) by treatment arm over time from ECU-MG-302 baseline to week 52 in study ECU-MG-302 using a repeated-measures model.
  • FIG. 20 is a graphical depiction of the change from baseline in MG-ADL total score (Mean and 95% CI) by treatment arm over time from ECU-MG-301 baseline to week 52 in study ECU-MG-302.
  • the disclosure provides methods of treating myasthenia gravis (MG) in subjects or patients in need thereof by administering an antibody that specifically binds complement component 5 (C5).
  • the antibody that specifically binds C5 reduces the rate at which C5 is cleaved, in vivo, into C5a and C5b.
  • the antibody that specifically binds C5 binds to one or both of the C5a and/or C5b fragments.
  • the antibody that specifically binds C5 blocks the complement cascade at C5, thereby reducing the release of proinflammatory mediators such as C5a and the formation of a C5b-9 Membrane Attack Complex (MAC).
  • MAC C5b-9 Membrane Attack Complex
  • the antibody that specifically binds C5 is eculizumab.
  • eculizumab is an antibody or a fragment thereof.
  • Eculizumab (h5G1.1-mAb) is a humanized monoclonal antibody (mAb) that was derived from the murine anti-human C5 antibody m5G1.1.
  • Eculizumab specifically binds the terminal complement protein C5, thereby inhibiting its cleavage to C5a and C5b during complement activation. This strategic blockade of the complement cascade at C5 prevents the release of proinflammatory mediators and the formation of the Membrane Attack Complex or cytolytic pore, while preserving the early components of complement activation that are essential for the opsonization of microorganisms and clearance of immune complexes.
  • the anti-C5 antibody is a monoclonal antibody having a hybrid IgG2/4 isotype.
  • the anti-C5 antibodies are effective in reducing the cell-lysing ability of complement present in human blood. This property of the antibodies can be determined by methods well known in the art such as, for example, by the chicken erythrocyte hemolysis method described in U.S. Pat. No. 6,355,245.
  • anti-C5 antibodies bind to C5 or fragments thereof, e.g., C5a or C5b.
  • the anti-C5 antibodies recognize and bind epitopes on either the alpha chain or the beta chain of purified human complement component C5 and are capable of blocking the conversion of C5 into C5a and C5b by C5 convertase. See Wurzner et al., Complement. Inflamm. 8(5-6): 328-40 (1991).
  • the anti-C5 antibodies recognize and bind epitopes within the alpha chain of purified human complement component C5.
  • the antibodies are capable of blocking the conversion of C5 into C5a and C5b by C5 convertase.
  • the antibodies can provide this blockade at substantially the same concentrations needed to block hemolytic activity.
  • the antibodies specifically bind to an amino-terminal region within the alpha chain, however, they do not specifically bind to free C5a.
  • the C5 antibody is able to substantially inhibit complement hemolytic activity and to substantially inhibit the conversion of C5 to produce C5a.
  • the C5 antibodies provide these functions when used at a molar ratio of antibody to antigen (C5) of 3:1 or less.
  • antibodies refers to immunoglobulins produced in vivo, as well as those produced in vitro by a hybridoma, and antigen binding fragments (e.g., Fab′ preparations) of such immunoglobulins, as well as to recombinantly expressed antibodies or antigen binding proteins, including immunoglobulins, chimeric immunoglobulins, “humanized” immunoglobulins, antigen binding fragments of such immunoglobulins, single chain antibodies, and other recombinant proteins containing antigen binding domains derived from immunoglobulins such as DVD-Ig and CODV-Ig. See U.S. Pat. Nos. 7,161,181 and 9,181,349. “Specificity” refers to the ability of a binding protein to selectively recognize and bind an antigen at a particular location or structure, known as an epitope, often found on the surface of the antigen.
  • the term “specifically binds,” means that a binding protein or fragment thereof forms a complex with an antigen that is relatively stable under physiologic conditions.
  • Specific binding can be characterized by a dissociation constant of at least about 1 ⁇ 10 ⁇ 6 M or smaller. In other embodiments, the dissociation constant is at least about 1 ⁇ 10 ⁇ 7 M, 1 ⁇ 10 ⁇ 8 M, 1 ⁇ 10 ⁇ 9 M, or 1 ⁇ 10 ⁇ 1 ° M.
  • Methods for determining whether two molecules specifically bind are well known in the art and include, for example, equilibrium dialysis, surface plasmon resonance, and the like.
  • anti-C5 antibodies described herein bind to complement component C5 (e.g., human C5) and inhibit the cleavage of C5 into fragments C5a and C5b.
  • complement component C5 e.g., human C5
  • Anti-C5 antibodies (or VH/VL domains derived therefrom) suitable for use in the invention can be generated using methods known in the art.
  • An exemplary anti-C5 antibody is eculizumab comprising heavy and light chains having the sequences shown in SEQ ID NOs: 10 and 11, respectively, or antigen binding fragments and variants thereof.
  • Eculizumab also known as SOLIRIS®
  • SOLIRIS® SOLIRIS®
  • Eculizumab is a humanized monoclonal antibody that is a terminal complement inhibitor.
  • the antibody comprises the heavy and light chain complementarity determining regions (CDRs) or variable regions of eculizumab. Accordingly, in one embodiment, the antibody comprises the CDR1, CDR2, and CDR3 domains of the VH region of eculizumab having the sequence set forth in SEQ ID NO: 7, and the CDR1, CDR2, and CDR3 domains of the VL region of eculizumab having the sequence set forth in SEQ ID NO: 8.
  • the antibody comprises heavy chain CDR1, CDR2, and CDR3 domains having the sequences set forth in SEQ ID NOs: 1, 2, and 3, respectively, and light chain CDR1, CDR2, and CDR3 domains having the sequences set forth in SEQ ID NOs: 4, 5, and 6, respectively.
  • the antibody comprises VH and VL regions having the amino acid sequences set forth in SEQ ID NO: 7 and SEQ ID NO: 8, respectively.
  • Empirical data indicate that serum eculizumab concentrations greater than 50 ⁇ g/mL and closer to at least 100 ⁇ g/mL are required to significantly reduce free C5 concentrations. Specifically, free C5 concentration was reduced significantly with increasing concentrations of eculizumab beginning at >50 ⁇ g/mL and was at near zero levels with eculizumab concentrations above 100 ⁇ g/ml.
  • the method comprises administering a therapeutically effective amount of eculizumab to the subject, wherein the therapeutically effective amount of eculizumab is maintained at a concentration of at least 50 ⁇ g/mL of eculizumab in serum of the subject.
  • the method comprises administering a therapeutically effective amount of eculizumab to the subject, wherein the therapeutically effective amount of eculizumab is maintained at a concentration of at least 60 ⁇ g/mL of eculizumab in serum of the subject. In one embodiment, the method comprises administering a therapeutically effective amount of eculizumab to the subject, wherein the therapeutically effective amount of eculizumab is maintained at a concentration of at least 70 ⁇ g/mL of eculizumab in serum of the subject.
  • the method comprises administering a therapeutically effective amount of eculizumab to the subject, wherein the therapeutically effective amount of eculizumab is maintained at a concentration of at least 80 ⁇ g/mL of eculizumab in serum of the subject. In another embodiment, the method comprises administering a therapeutically effective amount of eculizumab to the subject, wherein the therapeutically effective amount of eculizumab is maintained at a concentration of at least 90 ⁇ g/mL of eculizumab in serum of the subject.
  • the method comprises administering a therapeutically effective amount of eculizumab to the subject, wherein the therapeutically effective amount of eculizumab is maintained at a concentration of at least 100 ⁇ g/mL of eculizumab in serum of the subject.
  • Another exemplary anti-C5 antibody is an eculizumab variant, known as antibody BNJ441, and engineered to have a longer half-life (T1 ⁇ 2) in humans comprising heavy and light chains having the sequences shown in SEQ ID NOs: 14 and 11, respectively, or antigen binding fragments and variants thereof.
  • BNJ441 also known as ALXN1210
  • ALXN1210 is described in International Publication No. WO 2015/134894 A1 and U.S. Pat. No. 9,079,949, the teachings or which are hereby incorporated by reference.
  • BNJ441 is a humanized monoclonal antibody that is structurally related to eculizumab (SOURIS®).
  • BNJ441 selectively binds to human complement protein C5, inhibiting its cleavage to C5a and C5b during complement activation. This inhibition prevents the release of the proinflammatory mediator C5a and the formation of the cytolytic pore-forming membrane attack complex C5b-9 while preserving the proximal or early components of complement activation (e.g., C3 and C3b) essential for the opsonization of microorganisms and clearance of immune complexes.
  • complement activation e.g., C3 and C3b
  • the antibody comprises the heavy and light chain CDRs or variable regions of BNJ441. Accordingly, in one embodiment, the antibody comprises the CDR1, CDR2, and CDR3 domains of the VH region of BNJ441 having the sequence set forth in SEQ ID NO: 12, and the CDR1, CDR2, and CDR3 domains of the VL region of BNJ441 having the sequence set forth in SEQ ID NO: 8. In another embodiment, the antibody comprises heavy chain CDR1, CDR2, and CDR3 domains having the sequences set forth in SEQ ID NOs: 19, 18, and 3, respectively, and light chain CDR1, CDR2, and CDR3 domains having the sequences set forth in SEQ ID NOs: 4, 5, and 6, respectively.
  • the antibody comprises VH and VL regions having the amino acid sequences set forth in SEQ ID NO: 12 and SEQ ID NO: 8, respectively.
  • the antibody may comprise the heavy chain constant region of BNJ441 having the amino acid sequence set forth in SEQ ID NO: 13.
  • eculizumab is administered in a multiphase dosing regimen.
  • the multiphase dosing regimen comprises a first phase and a second phase in various embodiments.
  • the first phase is an induction phase and comprises administration of eculizumab at between 900 mg once a week to the subject for between 1-10 weeks.
  • the induction phase is concluded by administering the first maintenance phase dose of 1200 mg one week after the last 900 mg dose.
  • the second phase is a maintenance phase and comprises administration of eculizumab at between 1000 and 1400 mg once every two weeks to the subject for 2 weeks, 4 weeks, 6 weeks, 8 weeks, 12, weeks, 26 weeks, or as long as myasthenia gravis persists.
  • the maintenance phase comprises administration of eculizumab at between 1000 and 1400 mg once every two weeks to the subject for 2 months, 4 months, 6 months, 8 months, 12 months, 2 years, three years, 4 years, 5 years, or for the remaining lifetime of the patient.
  • the maintenance phase comprises administration of eculizumab at about 1200 mg twice a month (biweekly) once the induction phase is complete.
  • the method comprises administering a therapeutically effective amount of eculizumab or an eculizumab variant to the subject, wherein the therapeutically effective amount of eculizumab or eculizumab variant is maintained at a concentration of between 50-100 ⁇ g/mL, between 60-100 ⁇ g/mL, between 70-100 ⁇ g/mL, between 80-100 ⁇ g/mL, or between 90-100 ⁇ g/mL of eculizumab in serum of the subject.
  • Another exemplary anti-C5 antibody is antibody BNJ421 comprising heavy and light chains having the sequences shown in SEQ ID NOs: 20 and 11, respectively, or antigen binding fragments and variants thereof.
  • BNJ421 also known as ALXN1211
  • ALXN1211 is described in International Publication No. WO 2015/134894 A1 and U.S. Pat. No. 9,079,949, the teachings or which are hereby incorporated by reference.
  • the antibody comprises the heavy and light chain CDRs or variable regions of BNJ421. Accordingly, in one embodiment, the antibody comprises the CDR1, CDR2, and CDR3 domains of the VH region of BNJ421 having the sequence set forth in SEQ ID NO: 12, and the CDR1, CDR2, and CDR3 domains of the VL region of BNJ421 having the sequence set forth in SEQ ID NO: 8. In another embodiment, the antibody comprises heavy chain CDR1, CDR2, and CDR3 domains having the sequences set forth in SEQ ID NOs: 19, 18, and 3, respectively, and light chain CDR1, CDR2, and CDR3 domains having the sequences set forth in SEQ ID NOs: 4, 5, and 6, respectively.
  • the antibody comprises VH and VL regions having the amino acid sequences set forth in SEQ ID NO: 12 and SEQ ID NO: 8, respectively.
  • the antibody may comprise the heavy chain constant region of BNJ421 having the amino acid sequence set forth in SEQ ID NO: 9.
  • the antibody may comprise the heavy and light chain CDRs or variable regions of the 7086 antibody. See U.S. Pat. Nos. 8,241,628 and 8,883,158.
  • the antibody, or a fragment thereof may comprise heavy chain CDR1, CDR2, and CDR3 domains having the sequences set forth in SEQ ID NOs: 21, 22, and 23, respectively, and light chain CDR1, CDR2, and CDR3 domains having the sequences set forth in SEQ ID NOs: 24, 25, and 26, respectively.
  • the antibody or fragment thereof may comprise the VH region of the 7086 antibody having the sequence set forth in SEQ ID NO: 27, and the VL region of the 7086 antibody having the sequence set forth in SEQ ID NO: 28.
  • the antibody may comprise the heavy and light chain CDRs or variable regions of the 8110 antibody.
  • the antibody, or fragment thereof may comprise heavy chain CDR1, CDR2, and CDR3 domains having the sequences set forth in SEQ ID NOs: 29, 30, and 31, respectively, and light chain CDR1, CDR2, and CDR3 domains having the sequences set forth in SEQ ID NOs: 32, 33, and 34, respectively.
  • the antibody may comprise the VH region of the 8110 antibody having the sequence set forth in SEQ ID NO: 35, and the VL region of the 8110 antibody having the sequence set forth in SEQ ID NO: 36.
  • Another exemplary anti-C5 antibody comprises a heavy chain variable region amino acid sequence according to SEQ ID NO: 37 and a light chain variable region amino acid sequence according to SEQ ID NO: 38.
  • eculizumab, an eculizumab variant such as BNJ441, or other anti-C5 antibody is administered to the subject once a month, once every two months, or once every three months depending on the dose.
  • the eculizumab, eculizumab variant such as BNJ441, or other anti-C5 antibody is administered once every two weeks, once a week, twice a week, or three times a week.
  • eculizumab, eculizumab variant such as BNJ441, or other anti-C5 antibody is administered once a week, once every two weeks, once every three weeks, once every four weeks, once every five weeks, once every six weeks, or once every eight weeks depending on the needs of the patient.
  • eculizumab, eculizumab variant such as BNJ441, or other anti-C5 antibody in administered intravenously (IV) or subcutaneously (SubQ).
  • compositions comprising an anti-C5 antibody or antigen binding fragment thereof with a pharmaceutically acceptable excipient for treating MG.
  • the composition comprises an antibody comprising the CDR1, CDR2, and CDR3 domains of the VH region of eculizumab having the sequence set forth in SEQ ID NO: 7, and the CDR1, CDR2, and CDR3 domains of the VL region of eculizumab having the sequence set forth in SEQ ID NO: 8.
  • the antibody comprises heavy chain CDR1, CDR2, and CDR3 domains having the sequences set forth in SEQ ID NOs: 1, 2, and 3, respectively, and light chain CDR1, CDR2, and CDR3 domains having the sequences set forth in SEQ ID NOs: 4, 5, and 6, respectively.
  • the antibody comprises VH and VL regions having the amino acid sequences set forth in SEQ ID NO: 7 and SEQ ID NO: 8, respectively.
  • the antibody comprises the heavy and light chain CDRs or variable regions of BNJ441.
  • the antibody comprises the CDR1, CDR2, and CDR3 domains of the VH region of BNJ441 having the sequence set forth in SEQ ID NO: 12, and the CDR1, CDR2, and CDR3 domains of the VL region of BNJ441 having the sequence set forth in SEQ ID NO: 8.
  • the antibody comprises heavy chain CDR1, CDR2, and CDR3 domains having the sequences set forth in SEQ ID NOs: 19, 18, and 3, respectively, and light chain CDR1, CDR2, and CDR3 domains having the sequences set forth in SEQ ID NOs: 4, 5, and 6, respectively.
  • the antibody comprises VH and VL regions having the amino acid sequences set forth in SEQ ID NO: 12 and SEQ ID NO: 8, respectively.
  • the antibody comprises the heavy and light chain CDRs or variable regions of BNJ421.
  • the antibody comprises the CDR1, CDR2, and CDR3 domains of the VH region of BNJ421 having the sequence set forth in SEQ ID NO: 12, and the CDR1, CDR2, and CDR3 domains of the VL region of BNJ421 having the sequence set forth in SEQ ID NO: 8.
  • the antibody comprises heavy chain CDR1, CDR2, and CDR3 domains having the sequences set forth in SEQ ID NOs: 19, 18, and 3, respectively, and light chain CDR1, CDR2, and CDR3 domains having the sequences set forth in SEQ ID NOs: 4, 5, and 6, respectively.
  • the antibody comprises VH and VL regions having the amino acid sequences set forth in SEQ ID NO: 12 and SEQ ID NO: 8, respectively.
  • the disclosure provides methods of treating subjects suffering from myasthenia gravis (MG) by administering an antibody that specifically binds C5.
  • the subject is a mammalian subject.
  • subjects and/or patients are interchangeable.
  • subjects and/or patients are mammals.
  • primates include humans.
  • the subjects or patients suffering from MG described herein are humans.
  • MG includes refractory generalized myasthenia gravis.
  • refractory generalized myasthenia gravis is characterized as including subjects or patients positive for auto-antibodies binding to nicotinic acetylcholine receptor (anti-AChR) who continue to show marked generalized weakness or bulbar signs and symptoms of myasthenia gravis while receiving current standard of care for myasthenia gravis such as cholinesterase inhibitor therapy and immunosuppressant therapy (IST) or who require chronic plasma exchange or chronic IVIg to maintain clinical stability.
  • anti-AChR nicotinic acetylcholine receptor
  • refractory generalized myasthenia gravis is characterized as including subjects or patients who continue to show marked generalized weakness or bulbar signs and symptoms of myasthenia gravis while receiving current standard of care for myasthenia gravis such as cholinesterase inhibitor therapy and immunosuppressant therapy (IST) or who require chronic plasma exchange or chronic IVIg to maintain clinical stability.
  • current standard of care for myasthenia gravis such as cholinesterase inhibitor therapy and immunosuppressant therapy (IST) or who require chronic plasma exchange or chronic IVIg to maintain clinical stability.
  • MG includes refractory generalized myasthenia gravis.
  • refractory generalized myasthenia gravis is characterized as including subjects or patients positive for auto-antibodies binding to nicotinic acetylcholine receptor (anti-AChR) who continue to show marked generalized weakness or bulbar signs and symptoms of myasthenia gravis while receiving cholinesterase inhibitor therapy and immunosuppressant therapy (IST) and who require chronic plasma exchange or chronic IVIg to maintain clinical stability.
  • anti-AChR nicotinic acetylcholine receptor
  • refractory generalized myasthenia gravis is characterized as including subjects or patients who continue to show marked generalized weakness or bulbar signs and symptoms of myasthenia gravis while receiving cholinesterase inhibitor therapy and immunosuppressant therapy (IST) and who require chronic plasma exchange or chronic IVIg to maintain clinical stability.
  • IST immunosuppressant therapy
  • the phrase “requires chronic plasma exchange” to maintain clinical stability refers to the use of plasma exchange therapy on a patient on a regular basis for the management of muscle weakness at least every 3 months over the last 12 months.
  • the phrase “requires chronic IVIg” to maintain clinical stability refers to the use of IVIg therapy on a patient on a regular basis for the management of muscle weakness at least every 3 months over the last 12 months.
  • treatment of MG includes the amelioration or improvement of one or more symptoms associated with MG.
  • Symptoms associated with MG include muscle weakness and fatigability. Muscles primarily affected by MG include muscles that control eye and eyelid movement, facial expressions, chewing, talking, swallowing, breathing, neck movements, and limb movements.
  • treatment of MG includes the improvement of a clinical marker for MG progression.
  • markers include MG activity of daily living profile (MG-ADL), quantitative Myasthenia Gravis (QMG) score for disease severity, Myasthenia Gravis composite (MGC), negative inspiratory force (NIF), forced vital capacity, MGFA post-intervention status, and other quality of life measurements.
  • MG-ADL is the primary score for measuring improvement of MG.
  • the MG-ADL is an 8-point questionnaire that focuses on relevant symptoms and functional performance of activities of daily living (ADL) in MG subjects (see Table 1).
  • the 8 items of the MG-ADL were derived from symptom-based components of the original 13-item QMG to assess disability secondary to ocular (2 items), bulbar (3 items), respiratory (1 item), and gross motor or limb (2 items) impairment related to effects from MG.
  • each response is graded 0 (normal) to 3 (most severe).
  • the range of total MG-ADL score is 0-24.
  • a clinically meaningful improvement in a patient's MG-ADL would be a 3 point or greater reduction in score after 26 weeks of treatment.
  • the current QMG scoring system consists of 13 items: ocular (2 items), facial (1 item), bulbar (2 items), gross motor (6 items), axial (1 item), and respiratory (1 item); each graded 0 to 3, with 3 being the most severe (see Table 2).
  • the range of total QMG score is 0 -39.
  • the QMG scoring system is considered to be an objective evaluation of therapy for MG and is based on quantitative testing of sentinel muscle groups.
  • the MGFA task force has recommended that the QMG score be used in prospective studies of therapy for MG.
  • a clinically meaningful improvement in a patient's QMG would be a 5 point or greater reduction in score after 26 weeks of treatment.
  • MG ACTIVITY OF DAILY LIVING PROFILE Score Items Grade 0 Grade 1 Grade 2 Grade 3 (0, 1, 2, 3) 1. Talking Normal Intermittent Constant Difficult to slurring or slurring or understand nasal speech nasal, but can speech be understood 2. Chewing Normal Fatigue with Fatigue with Gastric Tube solid food soft food 3. Swallowing Normal Rare episode of Frequent Gastric Tube choking choking necessitating changes in diet 4. Breathing Normal Shortness of Shortness of Ventilator breath with breath at rest dependence exertion 5. Impairment of None Extra effort, Rest periods Cannot do one ability to brush but no rest needed of these teeth or comb hair periods needed functions 6.
  • the MGC is a validated assessment tool for measuring clinical status of subjects with MG (16).
  • the MGC assesses 10 important functional areas most frequently affected by MG and the scales are weighted for clinical significance that incorporates subject-reported outcomes. See Table 3.
  • MGC will be administered at Screening, Day 1, Weeks 1-4, 8, 12, 16, 20, and 26 or ET (Visits 1-6, 8, 10, 12, 14, and 17 or ET).
  • a clinically meaningful improvement in a patient's MGC would be a 3 point or greater reduction in score after 26 weeks of treatment.
  • the 15-item Myasthenia Gravis Qualify of Life 15 scale (MG-QOL 15) is a health-related quality of life evaluative instrument specific to subjects with MG. See Table 4. MG-QOL 15 was designed to provide information about subjects' perception of impairment and disability and the degree to which disease manifestations are tolerated and to be easy to administer and interpret. The range of total scores is from 0 to 60. Higher scores translate into a greater extent of a patient's dissatisfaction with MG related dysfunction. The MG-QOL 15 is completed by the subject. Higher scores indicate greater extent of and dissatisfaction with MG-related dysfunction. A clinically meaningful improvement in a patient's MG-QOL 15 would be a decrease in score after 26 weeks of treatment.
  • the Neuro-QOL Fatigue is a reliable and validated brief 19-item survey of fatigue completed by the subject. Higher scores indicate greater fatigue and greater impact of MG on activities (see Table 5). A clinically meaningful improvement in a patient's Neuro-QQL Fatigue score would be reflected in a decrease in score after 26 weeks of treatment.
  • the EUROQOL is a reliable and validated survey of health status in 5 areas: mobility, self-care, usual activities, pain/discomfort, and anxiety/depression, completed by the subject. Each area has 3 levels: level 1 (no problems), level 2 (some problems), and level 3 (extreme problems) (see FIGS. 2A and 2B ).
  • the EQ VAS records the subject's self-rated health on a vertical, 20 cm visual analogue scale where the endpoints are labeled “Best imaginable health state, marked as 100” and “Worst imaginable health state, marked as 0.”
  • the EQ-5D is administered at Day 1, Weeks 4, 8, 12, 16, 20, and 26 or ET (Visits 2, 6, 8, 10, 12, 14, and 17 or ET). A clinically meaningful improvement in a patient's EQ-5D would be reflected as an increase in score after 26 weeks of treatment.
  • FVC Forced Vital Capacity
  • NIF Negative Inspiratory Force
  • the MG clinical state is assessed using the MGFA Post-Intervention Status. Change in status categories of Improved, Unchanged, Worse, Exacerbation and Died of MG as well as the Minimal Manifestation (MM) can be assessed.
  • MM Minimal Manifestation
  • patients administered eculizumab show a reduced MG-ADL.
  • the subjects have an initial MG-ADL score of greater than 6 points.
  • the subjects have an initial MG-ADL score greater than 0, 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, or 23 points.
  • the MG-ADL score of the subject has been reduced to less than 6 points.
  • the MG-ADL score has been reduced at least 1 point, at least 2 points, at least 3 points, at least 4 points, at least 5 points, at least 6 points, at least 7 points, at least 8 points, at least 9 points, at least 10 points, at least 11 points, at least 12 points, at least 13 points, at least 14 points, at least 15 points, at least 16 points, at least 17 points, at least 18 points, at least 19 points, at least 20 points, at least 21 points, at least 22 points, at least 23 points, or at least 24 points after treatment with eculizumab.
  • the MG-ADL score of the patient is reduced by at least 1 point after a course of treatment with eculizumab.
  • the MG-ADL of the patient is reduced by 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, or 24 points after a course of treatment with eculizumab.
  • the course of treatment with eculizumab lasts for 26 weeks.
  • the course of treatment lasts for 26-52, 26-78, 26-104, 26-130, 26-156, 26-182, 26-208 weeks, or more.
  • the course of treatment lasts for greater than 26, 27, 28, 29, 30, 31, 32, 33, 34, 35, 36, 37, 38, 39, 40, 41, 42, 43, 44, 45, 46, 47, 48, 49, 50, 51, 52, 78, 104, 130, 156, or 182 weeks.
  • the course of treatment lasts for greater than 1, 2, 3, 4, 5, 10, 15, 20, 25, 30, 35, 40, 45, 50, 55, 60, 65, 70, 75, 80, or more years. In certain embodiments, the course of treatment lasts for the remainder of the subject's life.
  • one or more symptoms or scores associated with MG improves during the course of treatment and is maintained at the improved level throughout treatment.
  • MG-ADL can improve after 26 weeks of treatment with a therapeutic antibody that specifically binds C5 and then remain at the improved level for the duration of the treatment, which is 52 weeks of treatment with a therapeutic antibody that specifically binds C5.
  • a therapeutic antibody that binds C5 is eculizumab.
  • the first sign of improvement occurs by 26 weeks of treatment with a therapeutic antibody that specifically binds C5.
  • the first sign of improvement occurs between weeks 1-26, 26-52, 52-78, 78-104, 104-130, 130-156, 156-182, or 182-208 of treatment with a therapeutic antibody that specifically binds C5.
  • the first sign of improvement occurs at week 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 31, 32, 33, 34, 35, 36, 37, 38, 39, 40, 41, 42, 43, 44, 45, 46, 47, 48, 49, 50, 51, 52, 78, 104, 130, 156, or 182.
  • the first sign of improvement is maintained for a number of weeks during treatment with a binding protein that specifically binds C5, such as eculizumab or an eculizumab variant such as BNJ441.
  • this number of weeks is at least 26.
  • this number of weeks is 1-26, 26-52, 52-78, 78-104, 104-130, 130-156, 156-182, or 182-208.
  • this number of weeks is at least 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 31, 32, 33, 34, 35, 36, 37, 38, 39, 40, 41, 42, 43, 44, 45, 46, 47, 48, 49, 50, 51, 52, 78, 104, 130, 156, or 182.
  • eculizumab or other anti-C5 antibodies such as BNJ441, BNJ421, 7086, and 8110 can be administered to a subject suffering from MG at between 600 and 6000 mg.
  • the induction dose of eculizumab or other anti-C5 antibodies such as BNJ441, BNJ421, 7086, and 8110 is between 900 and 1500 mg, 900 and 1200 mg, 900 mg, or 1200 mg.
  • the maintenance dose of eculizumab or other anti-C5 antibodies such as BNJ441, BNJ 421, 7086, and 8110 is about 600, 700, 800, 900, 1000, 1100, 1200, 1300, 1400, 1500, 1600, 1700, 1800, 1900, 2000, 2500, 3000, 4000, 5000, or 6000 mg.
  • doses can be administered once a month, once every two weeks, once a week, twice a week, or daily. According to certain embodiments, the dose is administered once every two weeks or once a week. According to other embodiments, eculizumab is administered to a subject suffering from MG in a multiphase dosing regimen. According to certain embodiments, the multiphase dosing regimen has 2, 3, 4, 6, 7, 8, 9, 10, or more phases. In certain embodiments, each phase provides a higher dose than the phase before it.
  • the eculizumab multiphase dosing regimen has two phases.
  • the first phase is an induction phase.
  • This phase provides a dose of 600, 900, 1200, 1500, or 1800 mg per week. In certain embodiments, this phase lasts for 2, 3, 4, 5, 6, 7, 8, 9, or 10 weeks. In other embodiments, this phase lasts between 2 and 6 weeks. In other embodiments, the phase lasts for 5 weeks.
  • the dose given any week is higher than the previous week. In other embodiments, the dose remains the same for a number of weeks and is then increased. In some embodiments, the dose remains the same for the first 1, 2, 3, 4, 5, 6, 7, 8, or 9 weeks and is then increased. In other embodiments, the dose remains the same for the first 4 weeks.
  • the eculizumab dose is administered at between 600 and 1200 mg, 800 and 1500 mg, 900 and 1200 mg, 900 and 1100 mg, 900 and 1000 mg, 800 and 1000 mg, 800 and 1100 mg, or 800 and 1200 mg for a number of weeks and is then increased.
  • the eculizumab dose is administered at about 900 mg on day 1 and is followed by doses of 900 mg on day 7, 900 mg on day 14, 900 mg on day 21, and then is increased to 1200 mg for the fifth dose on day 28, and then 1200 mg is administered every 14 ⁇ 2 days thereafter.
  • the eculizumab induction phase dosing regimen comprises five administered doses on the following schedule:
  • the actual days between each dose may vary during the induction by 1 or 2 days to accommodate unexpected events in the patients' schedule.
  • the second phase of eculizumab dosing is the maintenance phase.
  • the maintenance phase of eculizumab dosing can last for between 6 weeks and the life of the subject. According to other embodiments, the maintenance phase lasts for 26-52, 26-78, 26-104, 26-130, 26-156, 26-182, 26-208 weeks, or more. In other embodiments, the maintenance phase lasts for greater than 26, 27, 28, 29, 30, 31, 32, 33, 34, 35, 36, 37, 38, 39, 40, 41, 42, 43, 44, 45, 46, 47, 48, 49, 50, 51, 52, 78, 104, 130, 156, or 182 weeks.
  • the maintenance phase lasts for greater than 1, 2, 3, 4, 5, 10, 15, 20, 25, 30, 35, 40, 45, 50, 55, 60, 65, 70, 75, 80 years, or more years. In certain embodiments, the maintenance phase lasts for the remainder of the subject's life.
  • the eculizumab multiphase dosing regimen includes a third phase.
  • This third phase is used when an MG patient must undergo a rescue procedure to maintain clinical stability and includes administering plasma exchange and/or dosing with IVIg.
  • a dose of eculizumab is administered to replace the drug lost in plasma exchange.
  • this post-rescue eculizumab dose is between 300 and 1200 mg, 400 and 1500 mg, 500 and 1000 mg, 400 and 800 mg, or 500 and 700 mg.
  • this post-rescue eculizumab dose is about 600 mg.
  • a 600 mg eculizumab dose is administered within 1 hour after completion of plasmapheresis. In another embodiment, in the third phase a 600 mg dose is administered within 2 hours after completion of plasmapheresis. In another embodiment, in the third phase a 600 mg dose is administered within 3 hours after completion of plasmapheresis. In another embodiment, in the third phase a 600 mg dose is administered within 4 hours after completion of plasmapheresis. In another embodiment, in the third phase a 600 mg dose is administered within 5 hours after completion of plasmapheresis. In another embodiment, in the third phase a 600 mg dose is administered within 6 hours after completion of plasmapheresis.
  • compositions comprising eculizumab, either alone or in combination with prophylactic agents, therapeutic agents, and/or pharmaceutically acceptable carriers are provided.
  • the pharmaceutical compositions comprising eculizumab provided herein are for use in, but not limited to, diagnosing, detecting, or monitoring a disorder, in preventing, treating, managing, or ameliorating a disorder or one or more symptoms thereof, and/or in research.
  • the formulation of pharmaceutical compositions, either alone or in combination with prophylactic agents, therapeutic agents, and/or pharmaceutically acceptable carriers, is known to one skilled in the art.
  • An exemplary, non-limiting range for a therapeutically or prophylactically effective amount of eculizumab or other anti-C5 antibodies such as BNJ441, BNJ 421, 7086, and 8110 provided herein is 600-5000 mg, for example, 900-2000 mg. It is to be noted that dosage values may vary with the type and severity of the condition to be alleviated. It is to be further understood that for any particular subject, specific dosage regimens may be adjusted over time according to the individual need and the professional judgment of the person administering or supervising the administration of the compositions, and that dosage ranges set forth herein are exemplary only and are not intended to limit the scope or practice of the claimed methods.
  • An anti-C5 antibody provided herein also can also be administered with one or more additional medicaments or therapeutic agents useful in the treatment of MG.
  • the additional agent can be a therapeutic agent art-recognized as being useful to treat myasthenia gravis or condition being treated by the antibody provided herein.
  • the combination can also include more than one additional agent, e.g., two or three additional agents.
  • the binding agent in various embodiments is administered with an agent that is a protein, a peptide, a carbohydrate, a drug, a small molecule, or a genetic material (e.g., DNA or RNA).
  • the agent is one or more cholinesterase inhibitors, one or more corticosteroids, and/or one or more immunosuppressive drugs (most commonly azathioprine [AZA], cyclosporin, and/or mycophenolate mofetil [MMF]).
  • Item 1 A method of treating refractory generalized myasthenia gravis in a patient in need thereof comprising administering a therapeutically effective amount of an anti-complement component 5 (C5) antibody or antigen binding fragment thereof to the patient, wherein the patient is administered the anti-C5 antibody or antigen binding fragment thereof for at least 26 weeks.
  • C5 anti-complement component 5
  • Item 2 A method of treating refractory generalized myasthenia gravis in a patient in need thereof comprising administering a therapeutically effective amount of an anti-C5 antibody or antigen binding fragment thereof to the patient, wherein the anti-C5 antibody or antigen binding fragment thereof is eculizumab or an eculizumab variant, and wherein the patient is administered eculizumab or eculizumab variant for at least 26 weeks.
  • Item 3 The method of either items 1 or 2, wherein the patient is positive for auto-antibodies binding to nicotinic acetylcholine receptor (anti-AChR) and shows marked generalized weakness or bulbar signs and symptoms of myasthenia gravis while receiving therapy for myasthenia gravis including anticholinesterase inhibitor therapy and immunosuppressant therapy (IST) and requires chronic plasma exchange or chronic IVIg to maintain clinical stability, and
  • anti-AChR nicotinic acetylcholine receptor
  • IST immunosuppressant therapy
  • the patient is administered eculizumab for at least 26 weeks.
  • Item 4 A method of treating refractory generalized myasthenia gravis in a patient in need thereof comprising administering eculizumab to the patient:
  • anti-AChR nicotinic acetylcholine receptor
  • IST immunosuppressant therapy
  • eculizumab is administered using a phased dosing schedule with an induction phase comprising administering a 900 mg induction dose of eculizumab on day 1, administering 900 mg doses of eculizumab on days 7, 14, and 21, and administering 1200 mg of eculizumab as a fifth induction dose on day 28; and
  • the patient is administered eculizumab for at least 26 weeks.
  • Item 5 The method of any one of items 1-4, wherein the 28 day induction phase of eculizumab treatment is followed by a maintenance phase comprising administering 1200 mg of eculizumab 14 days after the fifth induction dose and administering 1200 mg of eculizumab every 14 ⁇ 2 days thereafter.
  • Item 6 The method of any one of items 1-5, wherein the dosing regimen further comprises a third phase.
  • Item 7 The method of any one of items 1-6, wherein the third phase comprises performing plasmapheresis on the patient and administering eculizumab at a dose of between 300 mg and 1200 mg to the patient within 4 hours of completion of plasmapheresis.
  • Item 8 The method of any one of items 1-7, wherein the third phase comprises performing plasmapheresis on the patient and administering eculizumab at a dose of between 600 mg and 900 mg to the patient within 90 minutes of completion of plasmapheresis.
  • Item 9 The method of any one of items 1-8, wherein the third phase comprises performing plasmapheresis on the patient and administering eculizumab at a dose of 600 mg to the patient within 1 hour of completion of plasmapheresis.
  • Item 10 The method of any one of items 1-9, wherein the patient experiences a clinically meaningful improvement (reduction) in Myasthenia Gravis Activities of Daily Living (MG-ADL) score after 26 weeks of treatment.
  • MG-ADL Myasthenia Gravis Activities of Daily Living
  • Item 11 The method of any one of items 1-10, wherein the clinically meaningful improvement the patient experiences is at least a 3 point reduction in the patient's MG-ADL score after 26 weeks of treatment.
  • Item 12 The method of any one of items 1-11, wherein the clinically meaningful improvement the patient experiences is at least a 4 point reduction in the patient's MG-ADL score after 26 weeks of treatment.
  • Item 13 The method of any one of items 1-12, wherein the patient experiences a clinically meaningful improvement (reduction) in quantitative Myasthenia Gravis score (QMG) after 26 weeks of treatment.
  • Item 14 The method of any one of items 1-13, wherein the clinically meaningful improvement the patient experiences is at least a 4 point reduction in the patient's QMG score after 26 weeks of treatment.
  • Item 15 The method of any one of items 1-14, wherein the clinically meaningful improvement the patient experiences is at least a 5 point reduction in the patient's QMG score after 26 weeks of treatment.
  • Item 16 The method of any one of items 1-15, wherein the patient experiences a clinically meaningful improvement (reduction) in Myasthenia Gravis Composite (MGC) score after 26 weeks of treatment.
  • MMC Myasthenia Gravis Composite
  • Item 17 The method of any one of items 1-16, wherein the clinically meaningful improvement the patient experiences is at least a 6 point reduction in the patient's MGC score after 26 weeks of treatment.
  • Item 18 The method of any one of items 1-17, wherein the clinically meaningful improvement the patient experiences is at least a 10 point reduction in the patient's MGC score after 26 weeks of treatment.
  • Item 19 The method of any one of items 1-18, wherein the patient experiences a clinically meaningful improvement in quality of life as measured by Myasthenia Gravis Quality of Life (MG-QOL-15) score after 26 weeks of treatment.
  • MG-QOL-15 Myasthenia Gravis Quality of Life
  • Item 20 The method of any one of items 1-19, wherein the clinically meaningful improvement the patient experiences is at least a 6 point reduction in the patient's MG-QOL-15 score after 26 weeks of treatment.
  • Item 21 The method of any one of items 1-20, wherein the clinically meaningful improvement the patient experiences is at least an 11 point reduction in the patient's MG-QOL-15 score after 26 weeks of treatment.
  • Item 22 The method of any one of items 1-21, wherein the patient experiences a clinically meaningful improvement (reduction) in neuro-fatigue as measured by Neuro-QOL Fatigue score after 26 weeks of treatment.
  • Items 23 The method of any one of items 1-22, wherein the clinically meaningful improvement the patient experiences is at least an 8 point reduction in the patient's Neuro-QOL score after 26 weeks of treatment.
  • Item 24 The method of any one of items 1-23, wherein the clinically meaningful improvement the patient experiences is at least a 16 point reduction in the patient's Neuro-QOL score after 26 weeks of treatment.
  • Item 25 The method of any one of items 1-24, wherein the patient experiences a clinically meaningful improvement (increase) in health status as measured by EQ-5D health status score after 26 weeks of treatment.
  • Item 26 A method of treating refractory generalized myasthenia gravis in a patient in need thereof comprising administering eculizumab to the patient:
  • anti-AChR nicotinic acetylcholine receptor
  • IST immunosuppressant therapy
  • eculizumab is administered using a phased dosing schedule comprising administering a 900 mg induction dose of eculizumab on day 1, administering 900 mg doses of eculizumab on days 7, 14, and 21, and administering 1200 mg as a fifth dose on day 28; wherein the patient is administered eculizumab for at least 26 weeks;
  • the 28 day induction phase of eculizumab treatment is followed by a maintenance phase comprising administering 1200 mg of eculizumab 14 days after the fifth induction dose and administering 1200 mg of eculizumab every 14 ⁇ 2 days thereafter; and wherein the patient has a clinically meaningful improvement (reduction) in at least two measurements of generalized myasthenia gravis severity selected from the group consisting of MG-ADL, QMG, MGC, MG-QOL, and Neuro-QOL.
  • Item 27 A method of treating refractory generalized myasthenia gravis in a patient in need thereof comprising administering eculizumab to the patient:
  • anti-AChR nicotinic acetylcholine receptor
  • IST immunosuppressant therapy
  • eculizumab is administered using a phased dosing schedule comprising administering a 900 mg induction dose of eculizumab on day 1, administering 900 mg doses of eculizumab on days 7, 14, and 21, and administering 1200 mg of eculizumab as a fifth induction dose on day 28;
  • the patient is administered eculizumab for at least 26 weeks;
  • the 28 day induction phase of eculizumab treatment is followed by a maintenance phase comprising administering 1200 mg of eculizumab 14 days after the fifth induction dose and administering 1200 mg of eculizumab every 14 ⁇ 2 days thereafter; and
  • the patient has a clinically meaningful improvement (reduction) in at least three measurements of generalized myasthenia gravis severity selected from the group consisting of MG-ADL, QMG, MGC, MG-QOL, and Neuro-QOL.
  • Item 28 The method of any one of items 1-27, wherein the patient has a clinically meaningful improvement (reduction) in at least four measurements of generalized myasthenia gravis severity selected from the group consisting of MG-ADL, QMG, MGC, MG-QOL, and Neuro-QOL.
  • Item 29 The method of any one of items 1-28, wherein the patient has a clinically meaningful improvement (reduction) in five measurements of generalized myasthenia gravis severity, wherein the five measurements of generalized myasthenia gravis severity are MG-ADL, QMG, MGC, MG-QOL, and Neuro-QOL.
  • Item 30 A method of treating refractory generalized myasthenia gravis in a patient in need thereof comprising administering eculizumab to the patient:
  • anti-AChR nicotinic acetylcholine receptor
  • IST immunosuppressant therapy
  • eculizumab is administered using a phased dosing schedule comprising administering a 900 mg induction dose of eculizumab on day 1, administering 900 mg doses of eculizumab on days 7, 14, and 21, and administering 1200 mg as a fifth induction dose on day 28;
  • the patient is administered eculizumab for at least 26 weeks; wherein the 28 day induction phase of eculizumab treatment is followed by a maintenance phase comprising administering 1200 mg of eculizumab 14 days after the fifth induction dose and administering 1200 mg of eculizumab every 14 ⁇ 2 days thereafter; and wherein the patient has a clinically meaningful improvement (reduction) in five measurements of generalized myasthenia gravis severity, wherein the five measurements of generalized myasthenia gravis severity are a reduction in MG-ADL of at least 3 points, a reduction in QMG of at least 4 points, a reduction in MGC of at least 6 points, a reduction in MG-QOL of at least 6 points, and a reduction in Neuro-QOL of at least 8 points.
  • the five measurements of generalized myasthenia gravis severity are a reduction in MG-ADL of at least 3 points, a reduction in QMG of at least 4 points, a reduction in MGC of at
  • Item 31 The method of any one of items 1-30, wherein the patient has a clinically meaningful improvement (reduction) in five measurements of generalized myasthenia gravis severity, wherein the five measurements of generalized myasthenia gravis severity are a reduction in MG-ADL of at least 4 points, a reduction in QMG of at least 5 points, a reduction in MGC of at least 10 points, a reduction in MG-QOL of at least 11 points, and a reduction in Neuro-QOL of at least 16 points.
  • the five measurements of generalized myasthenia gravis severity are a reduction in MG-ADL of at least 4 points, a reduction in QMG of at least 5 points, a reduction in MGC of at least 10 points, a reduction in MG-QOL of at least 11 points, and a reduction in Neuro-QOL of at least 16 points.
  • Item 32 The method of any one of items 1-31, wherein eculizumab is administered by intravenous infusion.
  • Item 33 The method of any one of items 1-32, wherein eculizumab is administered subcutaneously.
  • Item 34 The method of any one of items 1-33, wherein the eculizumab comprises a heavy chain amino acid sequence according to SEQ ID NO: 10 and a light chain amino acid sequence according to SEQ ID NO: 11.
  • Item 35 The method of any one of items 1-34, wherein the eculizumab is an eculizumab variant comprising a heavy chain amino acid sequence according to SEQ ID NO: 14 and a light chain amino acid sequence according to SEQ ID NO: 11.
  • Item 36 The method of any one of items 1-35, wherein the eculizumab is an eculizumab variant comprising a heavy chain variable region amino acid sequence according to SEQ ID NO: 12 and a light chain amino acid sequence according to SEQ ID NO: 11.
  • Item 37 The method of any one of items 1-36, wherein the anti-C5 antibody or antigen binding fragment thereof comprises a heavy chain variable region amino acid sequence according to SEQ ID NO: 27 and a light chain variable region amino acid sequence according to SEQ ID NO: 28.
  • Item 38 The method of any one of items 1-37, wherein the anti-C5 antibody or antigen binding fragment thereof comprises a heavy chain variable region amino acid sequence according to SEQ ID NO: 35 and a light chain variable region amino acid sequence according to SEQ ID NO: 36.
  • Item 39 The method of any one of items 1-38, wherein the patient has failed treatment over one year or more with two or more ISTs in sequence or in combination.
  • Item 40 The method of any one of items 1-39, wherein the patient has failed at least one IST and requires chronic plasma exchange or IVIg to control symptoms of myasthenia gravis.
  • Item 41 A method of treating refractory generalized myasthenia gravis in a patient in need thereof comprising administering eculizumab to the patient:
  • the patient is positive for auto-antibodies binding to nicotinic acetylcholine receptor (anti-AChR) and shows marked generalized weakness or bulbar signs and symptoms of myasthenia gravis while receiving therapy for myasthenia gravis including anticholinesterase inhibitor therapy and immunosuppressant therapy (IST) and wherein the patient had previously failed treatment with at least two immunosuppressive agents or failed treatment with at least one immunosuppressive agent and required chronic plasma exchange or IVIg, and had an MG-ADL total score ⁇ 6 at study entry;
  • anti-AChR nicotinic acetylcholine receptor
  • IST immunosuppressant therapy
  • eculizumab is administered using a phased dosing schedule with an induction phase comprising administering a 900 mg induction dose of eculizumab on day 1, administering 900 mg doses of eculizumab on days 7, 14, and 21, and administering 1200 mg of eculizumab as a fifth induction dose on day 28;
  • the patient is administered eculizumab for at least 26 weeks;
  • the 28 day induction phase of eculizumab treatment is followed by a maintenance phase comprising administering 1200 mg of eculizumab 14 days after the fifth induction dose and administering 1200 mg of eculizumab every 14 ⁇ 2 days thereafter; and
  • the patient has a clinically meaningful improvement (reduction) in at least two measurements of generalized myasthenia gravis severity selected from the group consisting of MG-ADL, QMG, and MGC.
  • Item 42 The method of any one of items 1-41, wherein the therapeutically effective amount of eculizumab or eculizumab variant administered to the patient is maintained at a concentration of between 50-100 ⁇ g/mL in the patient's serum.
  • Item 43 The method of any one of items 1-42, wherein the patient experiences a discontinuation in the administration of one or more IST following at least 26 weeks of treatment.
  • Item 44 The method of any one of items 1-43, wherein the patient experiences a reduction in IST dosing following at least 26 weeks of treatment.
  • Item 45 The method of any one of items 1-44, wherein the patient experiences a reduction in one or more IST dosing and a discontinuation in one or more IST following at least 26 of treatment.
  • the primary objective of this trial is to assess the efficacy of eculizumab as compared with placebo in the treatment of refractory gMG based on the improvement in the MG-specific Activities of Daily Living profile (MG-ADL).
  • MG-ADL MG-specific Activities of Daily Living profile
  • the secondary objectives of this trial include the following:
  • Described herein is a randomized, double-blind, parallel-group, placebo-controlled, multicenter ( ⁇ 100 sites in North America, South America, Europe, Asian Pacific) approximately two year trial to evaluate the safety and efficacy of eculizumab for the treatment in subjects with refractory gMG.
  • Approximately 92 eligible subjects are randomized on Day 1 on a 1:1 ratio to one of two treatment arms (1) eculizumab infusion or (2) placebo infusion.
  • Subjects may continue to receive stable dose/type of immunosuppressive therapy (IST), but no new ISTs and no increase in IST dosage are permitted during the trial.
  • IST immunosuppressive therapy
  • the overall trial duration for an individual subject is estimated to take up to 38 weeks including enrollment and Follow-up. Subjects may be provided the opportunity to participate in an extension trial (separate protocol) to receive eculizumab after completion of this trial.
  • a schedule of assessments for the screening, study and follow-up period is provided in Table 6.
  • the medical history review includes confirmation of MG diagnosis as defined in the inclusion criteria of this protocol, history of previous treatment/therapies for MG, e.g., thymectomy, IST including corticosteroids, IVIg and plasma exchange, history of MG exacerbation or crisis including the duration of each exacerbation/crisis, the medication taken at the time of each exacerbation/crisis and the treatment for each exacerbation/crisis.
  • subjects are vaccinated against N. meningitidis , if not already vaccinated within the time period of active coverage specified by the vaccine manufacturer or vaccinate according to current medical/country guidelines. Subjects must be vaccinated at least 14 days prior to receiving the first dose of study medication or be vaccinated and receive treatment with appropriate antibiotics until 14 days after the vaccination. See FIG. 3 .
  • the washout period for IVIg is 4 weeks prior to randomization.
  • the washout period for PE is also 4 weeks prior to randomization. If a subject experiences an MG Crisis during the Screening Period, the sponsor must be notified. Following discussion with the sponsor, a decision is made about whether the subject may continue in the trial, be withdrawn and possibly, re-screened at a later date.
  • AChR Ab Acetylcholine receptor antibody
  • B Baseline sample
  • C5 Complement protein 5
  • C-SSRS Columbia-Suicide Severity Rating Scale
  • ECG Electrocardiogram
  • ICU Intensive Care Unit
  • HAHA human anti-human antibody
  • IP investigational product
  • MG Myasthenia Gravis
  • MGC MG Composite Score
  • MG-ADL MG Activity of Daily Living (MG-ADL) Profile
  • MGFA Myasthenia Gravis Foundation of America
  • MGFA PIS MGFA Post-Intervention Status
  • NIF negative inspiratory force
  • P peak sample
  • PK/PD Pharmacokinetics/Pharmacodynamics
  • QMG Quantitative MG (QMG) Score for Disease Severity
  • QOL Quality of
  • Subjects receive IP, either eculizumab or placebo, according to the randomization and the regimen described in the Investigational Product and Administration, described below.
  • the treatment duration for each subject is 26-weeks.
  • Subjects must be informed of potential signs and symptoms of MG crisis and instructed to contact the Investigator as soon as possible at onset of symptom. Every effort is made for the subject reporting Clinical Deterioration to be evaluated as soon as possible and within 48 hours of notification of the Investigator of the symptom onset.
  • the Investigator or his/her designee performs the assessments as specified by this protocol. The Investigator determines whether or not the subject meets the definition of Clinical Deterioration as defined by this protocol and treats the subject accordingly.
  • Any ocular muscle weakness May have weakness of eye closure. All other muscle strength is normal. II Mild weakness affecting other than ocular muscles. May also have ocular muscle weakness of any severity. IIa Predominantly affecting limb or axial muscles or both. May also have lesser involvement of oropharyngeal muscles. IIb Predominantly affecting oropharyngeal or respiratory muscles or both. May also have lesser or equal involvement of limb or axial muscles or both. III Moderate weakness affecting other than ocular muscles. May also have ocular muscle weakness of any severity. IIIa Predominantly affecting limb or axial muscles or both. May also have lesser involvement of oropharyngeal muscles.
  • IIIb Predominantly affecting oropharyngeal or respiratory muscles or both. May also have lesser or equal involvement of limb or axial muscles or both.
  • IVa Predominantly affecting limb and/or axial muscles. May also have lesser involvement of oropharyngeal muscles.
  • IVb Predominantly affecting oropharyngeal or respiratory muscles or both. May also have lesser or equal involvement of limb or axial muscles or both.
  • V Defined by intubation, with or without mechanical ventilation, except when employed during routine postoperative management The use of a feeding tube without intubation places the patient in class IVb.
  • subjects may be provided an opportunity to enter an extension trial (separate protocol) to receive open-label eculizumab.
  • the visit interval between this trial and the extension trial is 2 weeks from the last of IP administration (Visit 17) so there is no interruption in IP dosing.
  • Subjects entering the extension trial undergo a blinded eculizumab induction phase similar to the induction in this trial in order to maintain the blinded treatment assignment of this trial. If a subject withdraws from this trial at any time after receiving any amount of IP or does not wish to enter the extension trial after completion of this trial, the subject is required to complete the Follow-up Visit for safety measures.
  • a subject withdraws or is discontinued from this trial at any time after receiving any amount of IP or does not wish to enter the extension trial after completion of this trial the subject will be required to complete the Follow-up Visit for safety measures 8 weeks after the last IP dose administration. If a subject is discontinued due to an AE, the event will be followed until it is resolved or in the opinion of the PI is medically stable.
  • Clinical Deterioration is defined as follows:
  • the Clinical Evaluators are study staff that have been trained and certified in administering the MG-ADL, QMG and MGC.
  • the Clinical Evaluator may be a neurologist, physical therapist or other study team member delegated by the PI. Clinical Evaluator training and certification for this protocol will take place either at the Investigator's meeting or via the sponsor's designated on-line training portal. 1.2.3.
  • MG assessments Responsibilities for MG assessments are listed in Table 9. Throughout the trial, MG assessments should be performed at approximately the same time of day by a properly trained evaluator, preferably the same evaluator.
  • Visit intervals during Induction Phase are weekly (every 7 ⁇ 2 days after the last visit).
  • Visit intervals during the Maintenance Phase are every 2 weeks (every 14 days ⁇ 2 days since the last visit).
  • Subjects who fail to return for a scheduled visit must be contacted by the site study staffs to determine the reason for missing the appointment.
  • Subjects are strongly encouraged to return to the investigational site for evaluation if Clinical Deterioration or an AE is suspected to have occurred.
  • the investigational site obtains relevant medical records as documentation from the local physician's examination, and enters relevant data in the eCRF as appropriate.
  • IG(a) IVIg therapy acute (for exacerbations or preoperatively)
  • IVIg therapy acute (for exacerbations or preoperatively)
  • OT Other forms of therapy
  • IP will be administered after completion of other tests and procedures, excluding the peak blood sampling for PK/PD and free C5 assay.
  • Visit 8 At Visit 8 (Week 8), Visit 10 (Week 12), Visit 12 (Week 16), Visit 14 (Week 20), and until the EOS, Visit 17 (Week 26) or at ET, the following procedures are also completed, in addition to the 5 preceding procedures listed for the maintenance phase:
  • Unscheduled visits outside the specified visits are permitted at the discretion of the Investigator. Procedures, tests, and assessments are performed at the discretion of the Investigator. If an Unscheduled Visit is performed, any tests, procedures, or assessments performed at the Unscheduled Visits must be recorded on the eCRFs.
  • MM-0 The patient has received no MG treatment for at least 1 year.
  • MM-1 The patient continues to receive some form of immuno- suppression, but no cholinesterase inhibitors or other symptomatic therapy.
  • MM-2 The patient has received on low-dose cholinesterase inhibitors ( ⁇ 120 mg pyridostigmine/day) for at least 1 year.
  • MM-3 The patient has received cholinesterase inhibitors or other symptomatic therapy and some form of immuno- suppression during the past year.
  • Change in Status Improved (I) A substantial decrease in pretreatment clinical manifestations or a sustained substantial reduction in MG medications as defined in the protocol.
  • Subjects are allowed to withdraw consent at any time. Every effort should be made to ensure subjects are willing to comply with trial participation prior to conducting the screening procedures and the subjects should be fully informed of the restrictions related to the change of concomitant medications during the trial. Investigators may choose to discontinue a subject's treatment because of AEs, as well as conditions or illnesses that preclude compliance with the protocol from the standpoint of the subject's safety or well-being. The study staff should notify the Sponsor and their site monitor of all trial withdrawals as soon as possible.
  • eculizumab should not be administered to pregnant women.
  • all subjects of childbearing potential must continue to use adequate contraception for up to 5 months following discontinuation of eculizumab treatment. If a subject becomes pregnant, the IP must be immediately discontinued and the Sponsor must be notified. Each pregnancy will be followed to term and the Sponsor notified regarding the outcome.
  • Alexion Pharmaceuticals, Inc. or a regulatory authority may discontinue the trial at any time for any reason including, for example, clinical or administrative reasons.
  • the end of trial is defined as the last visit completed by the last patient.
  • Eculizumab 600 mg, 900 mg or 1200 mg
  • matching placebo is administered intravenously over approximately 35 minutes according to the regimen shown in Table 11.
  • Eculizumab or placebo 3 vials of IP (equivalent to 900 mg of eculizumab) weekly for 4 weeks (every 7 days ⁇ 2 days) followed by 4 vials of IP (equivalent to 1200 mg of eculizumab) one week later for the fifth dose (Visit 6/Week 4).
  • Eculizumab or placebo 4 vials of IP (equivalent to 1200 mg of eculizumab) every 2 weeks (14 days ⁇ 2 days)
  • PE is administered due to a Clinical Deterioration (as defined by this protocol)
  • supplemental IP 2 vials, equivalent to 600 mg of eculizumab or matching placebo
  • subjects will receive the regularly scheduled number of vials (3 vials on Visits 2-4; 4 vials on all other visits) within 60 minutes after each PE session.
  • immunosuppressive agents are allowed during the trial: corticosteroid, AZA, MMF, MTX, tacrolimus, cyclosporine, or cyclophosphamide.
  • the immunosuppressive agent(s) and its appropriate dose level to be used for an individual subject is at the discretion of the treating physician.
  • PE or IVIg will be allowed for subjects who experience a Clinical Deterioration as defined by this protocol.
  • the rescue therapy used for a particular subject is at the discretion of the treating physician. Every effort should be made to notify the Sponsor within 24 hours should a subject require a rescue therapy.
  • PE is administered as a rescue therapy
  • supplemental IP (2 vials) are administered within 60 minutes after the end of each PE session.
  • Routine (per protocol schedule) IP administration is continued per the specified dose-administration schedule for the subject. If the subject is scheduled to receive the protocol-scheduled dose on the day of a PE session, then the scheduled dose is administered within 60 minutes after the end of the PE session.
  • IP into subjects is under the supervision of the PI/Sub-Investigator or their designee, to ensure that the subject receives the appropriate dose at the appropriate time-points during the trial.
  • Subjects are randomized on Day 1 after the Investigator has verified that they are eligible. Subjects are randomized in a 1:1 ratio of eculizumab infusion to placebo infusion. The randomization will be across centers using an IXRS. The randomization stratification will be based on MGFA clinical classification assessed at the Screening Visit according to the following 4 groupings:
  • All trial subjects, investigational site personnel, sponsor staff, sponsor designees, and all staff directly associated with the conduct of the trial are blinded to the subject treatment assignments.
  • the double blind is maintained by using identical IP kits and labels for eculizumab and placebo.
  • the placebo has an identical appearance to that of eculizumab.
  • the random code is maintained by Almac Clinical Services. There is no antidote to reverse the effects of eculizumab.
  • unblinding would not be helpful in the planning of patient treatment for a given event. Unblinding should only be considered for the safety of the subject. If unblinding is deemed necessary by the Investigator, the Investigator can unblind the patient's treatment allocation using IXRS. The Investigator must note the date, time and reason for unblinding.
  • the Investigator should inform the Medical Monitor that the patient was unblinded, however they are not required to reveal to the Medical Monitor the patients' treatment allocation.
  • the blind When an AE is an unexpected related serious AE, the blind will be broken by the Sponsor only for that specific subject. The blind will be maintained for persons responsible for the ongoing conduct of the study (such as the management, monitors, investigators, etc.) and those responsible for data analysis and interpretation of results at the conclusion of the study, such as biometrics personnel. Unblinded information will only be accessible to those who need to be involved in the safety reporting to Health authorities, Ethics Committees and/or IRBs.
  • Each vial of IP contains eculizumab 300 mg or matching placebo for IV administration.
  • the active IP, eculizumab is manufactured and supplied by Alexion in single 30 mL vials as a solution concentration of 10 mg/ml.
  • the comparator product is manufactured by Alexion Pharmaceuticals, Inc., as a matching sterile, clear, colorless solution with the same buffer components but without active ingredient, in an identical 30 ml vial. See Table 12.
  • IP vials will be individually packaged into a kit. Both vials and kits will be labeled according to the protocol and local regulatory requirements. Each kit will have a label describing the contents and a place for the pharmacist to record the subject number and initials.
  • IP is released to the site upon receipt of all required essential documents based upon federal, state, and local regulations. See Table 12.
  • IP Upon arrival at the center, the IP is promptly removed from the shipping cooler and stored in refrigerated conditions at 2 to 8° C. The pharmacist should immediately record the reception of the IP and notify the distributor if vials are damaged and/or if temperature excursions have occurred during transportation. IP must be stored in a secure, limited-access storage area and temperature must be monitored daily.
  • Diluted solutions of IP may be stored at 2 to 8° C. (36-46° F.) for up to 24 hours prior to administration. If the IP is prepared more than 4 hours in advance of a subject's visit, the diluted material should be stored at 2 to 8° C. The solution should be allowed to warm to room temperature prior to administration. The material must not be heated (e.g., by using a microwave or other heat source) other than by ambient air temperature.
  • IP Infusions of IP are prepared using aseptic technique. Each vial of IP contains 300 mg of active ingredient in 30 mL of product solution or matching placebo.
  • IP IP from the vials.
  • Transfer the recommended dose to an infusion bag Dilute the IP to a final concentration of 5 mg/mL by addition to the infusion bag of the appropriate amount (equal volume) of 0.9% Sodium Chloride Injection, USP; 0.45% Sodium Chloride Injection, USP; 5% Dextrose in Water Injection, USP; or Ringer's Injection, USP.
  • the final volume of a 5 mg/mL diluted IP solution is 120 mL for 600 mg doses (2 vials), 180 mL for 900 mg doses (3 vials) and 240 mL for 1200 mg doses (4 vials) as shown in Table 13.
  • Do not Administer as an Iv Push or Bolus Injection IP is only administered via IV infusion and must be diluted to a final concentration of 5 mg/mL prior to administration. Prior to administration, if the diluted solution is refrigerated, it is allowed to warm to room temperature by exposure to ambient air. The diluted solution must not be heated in a microwave or with any heat source other than ambient air temperature. Parenteral drug products are inspected visually for particulate matter and discoloration prior to administration.
  • the diluted IP is intravenously administered over 35 minutes (range 25 to 45 minutes). It is not necessary to protect the infusion bags from light while IP is being administered to the subject.
  • the diluted IP may be administered via gravity feed, a syringe-type pump, or an infusion pump. The subjects are monitored for 1 hour following infusion.
  • the infusion may be slowed or stopped at the discretion of the Investigator, depending upon the nature and severity of the event.
  • the overall time of infusion should not exceed 2 hours.
  • the AE must be captured in the subject's source document and CRF.
  • the 26-week Study Period defines the time period for assessment of the study endpoints (specified in Table 6, the schedule of assessments). Efficacy will be assessed comparing eculizumab outcomes to placebo outcomes. Statistical analyses of the efficacy endpoints are summarized below and described in more detail in the statistical analysis plan. For all scales noted below except the EQ Visual Analog Scale (VAS) and Myasthenia Gravis Foundation of America (MGFA) Post-Intervention Status (PIS) the higher the score the greater the impairment.
  • VAS EQ Visual Analog Scale
  • MGFA Myasthenia Gravis Foundation of America
  • MG-ADL MG Activities of Daily Living Profile
  • the MG-ADL is an 8-point questionnaire that focuses on relevant symptoms and functional performance of activities of daily living (ADL) in MG subjects (see Table 1).
  • the 8 items of the MG-ADL were derived from symptom-based components of the original 13-item QMG to assess disability secondary to ocular (2 items), bulbar (3 items), respiratory (1 item), and gross motor or limb (2 items) impairment related to effects from MG.
  • each response is graded 0 (normal) to 3 (most severe).
  • the range of total MG-ADL score is 0-24.
  • a clinically meaningful improvement in a patient's MG-ADL would be a 3 point or greater reduction in score after 26 weeks of treatment.
  • the recall period for MG-ADL is the preceding 7 days.
  • MG-ADL will be performed at Screening, Day 1, Weeks1-4, 8, 10, 12, 16, 20, and 26 or ET (Visits 2-6, 8, 10, 12, 14, and 17, or ET) by a properly trained evaluator, preferably the same evaluator throughout the study.
  • the current QMG scoring system consists of 13 items: ocular (2 items), facial (1 item), bulbar (2 items), gross motor (6 items), axial (1 item) and respiratory (1 item); each graded 0 to 3, with 3 being the most severe (see Table 2).
  • the range of total QMG score is 0-39.
  • the QMG scoring system is considered to be an objective evaluation of therapy for MG and is based on quantitative testing of sentinel muscle groups.
  • the MGFA task force has recommended that the QMG score be used in prospective studies of therapy for MG(15).
  • a clinically meaningful improvement in a patient's QMG would be a 4 point or greater reduction in score after 26 weeks of treatment.
  • the QMG will be administered at Screening, Day 1, Weeks 1-4, 8, 12, 16, 20, and 26 or ET (Visits 1-6, 8, 10, 12, 14, and 17 or ET).
  • the MGC is a validated assessment tool for measuring clinical status of subjects with MG (16).
  • the MGC assesses 10 important functional areas most frequently affected by MG and the scales are weighted for clinical significance that incorporate subject-reported outcomes (see Table 3).
  • a clinically meaningful improvement in a patient's MGC would be a 3 point or greater reduction in score after 26 weeks of treatment.
  • MGC will be administered at Screening, Day 1, Weeks 1-4, 8, 12, 16, 20, and 26 or ET (Visits 1-6, 8, 10, 12, 14, and 17 or ET).
  • the 15-item Myasthenia Gravis Qualify of Life scale (MG-QOL 15) (see FIG. 1 ) is a health-related quality of life evaluative instrument specific to subjects with MG.
  • MG-QOL15 was designed to provide information about subjects' perception of impairment and disability and the degree to which disease manifestations are tolerated and to be easy to administer and interpret (17).
  • the MG-QOL 15 is completed by the subject. Higher scores indicate greater extent of and dissatisfaction with MG-related dysfunction. A clinically meaningful improvement in a patient's MG-QOL 15 would be an increase in score after 26 weeks of treatment.
  • the MG-QOL 15 is administered at Screening, Day 1, Weeks 4, 8, 12, 16, 20, and 26 or ET (Visits 1-2, 6, 8, 10, 12, 14, and 17 or ET).
  • the Neuro-QOL Fatigue is a reliable and validated brief 19-item survey of fatigue, completed by the subject (18). Higher scores indicate greater fatigue and greater impact of MG on activities (see Table 5). A clinically meaningful improvement in a patient's Neuro-QQL Fatigue score would be reflected in a decrease in score after 26 weeks of treatment.
  • the Neuro-QOL Fatigue is administered at Day 1, Weeks 4, 8, 12, 16, 20, and 26 or ET (Visits 2, 6, 8, 10, 12, 14, and 17 or ET).
  • the EUROQOL (EQ-5D) is a reliable and validated survey of health status in 5 areas: mobility, self-care, usual activities, pain/discomfort, and anxiety/depression, completed by the subject (19). Each area has 3 levels: level 1 (no problems), level 2 (some problems), and level 3 (extreme problems) (see FIGS. 2A and 2B ).
  • the EQ VAS records the subject's self-rated health on a vertical, 20 cm visual analogue scale where the endpoints are labeled “Best imaginable health state, marked as 100” and “Worst imaginable health state, marked as 0.”
  • a clinically meaningful improvement in a patient's EQ-5D would be reflected as an increase in score after 26 weeks of treatment.
  • the EQ-5D is administered at Day 1, Weeks 4, 8, 12, 16, 20, and 26 or ET (Visits 2, 6, 8, 10, 12, 14, and 17 or ET).
  • FVC Forced Vital Capacity
  • NIF Negative Inspiratory Force
  • the MG clinical state is assessed using the MGFA Post-Intervention Status. See Table 10. Change in status categories of Improved, Unchanged, Worse, Exacerbation and Died of MG as well as the Minimal manifestation (MM) is assessed and recorded at Weeks 4, 12 and 26 or ET (Visits 6, 10 and 17 or ET) by the PI or the same neurologist skilled in the evaluation of MG subjects throughout the trial.
  • the sub-scores of MM i.e., MM-0, MM-1, and MM-3, will not be used in this protocol.
  • the study design is a randomized, double blind, placebo-controlled design. Subjects will be randomly assigned 1:1 to eculizumab or placebo.
  • the randomization stratification variable will be based on MG clinical classification by the Myasthenia Gravis Foundation of America (MGFA) according to the following 4 groupings (Class IIa and IIIa, Class IVa, Class IIb and IIIb and Class IVb).
  • Analyses are produced for the double-blind Study Period in order to compare the eculizumab group with placebo group.
  • the analyses include efficacy, safety, and PK/PD analyses.
  • the full analysis set is the population on which primary, secondary, and tertiary efficacy analyses is performed and consists of all subjects who are randomized to IP and who have received at least 1 dose of IP (eculizumab or placebo treatment) and have at least one efficacy assessment post IP infusion. Subjects are compared for efficacy according to the treatment they were randomized to receive, irrespective of the treatment they actually received.
  • the Per-Protocol (PP) Set is a subset of the Full Analysis Set (FAS) population, excluding subjects with major protocol deviations.
  • FAS Full Analysis Set
  • the possible categories of major protocol deviations are defined in the statistical analysis plan.
  • the per-protocol population will include all subjects who:
  • the PP population will be fully described in the statistical analysis plan, and subjects identified prior to database lock. Efficacy analyses will also be performed on the PP data set.
  • Baseline is defined as the last available assessment prior to treatment for all subjects, regardless of treatment group.
  • the primary efficacy endpoint is change from baseline in the MG-ADL total score at Week 26 of the Study Period.
  • the primary efficacy analysis is conducted on the available 26 week data from the Study Period for all subjects. The trial is considered to have met its primary efficacy objective if a statistically significant difference (p ⁇ 0.05) between the eculizumab treatment group and placebo group is observed for change from baseline in the MG-ADL total score at Week 26. Confidence intervals and p-values will be presented.
  • treatment groups are compared using a worst-rank score analysis (i.e., analysis of covariance [ANCOVA] analysis with ranks) with effects for treatment.
  • the baseline MG-ADL total score and the randomization stratification variables are also to be covariates in the model.
  • the actual changes from baseline are ranked from highest (best improvement in MG-ADL score) to lowest (least improvement/most worsening in MG-ADL score) across all subjects who did not need rescue therapy. Then, any subject who needed rescue therapy would be given lower ranks. These lower ranks are based on the time to rescue therapy from the start of investigational product (Day 1).
  • the subject with the shortest time to rescue therapy would get the absolute lowest rank in the analysis and the subject with the longest time to rescue therapy would get a rank that is one lower than the lowest ranked subject without rescue therapy.
  • Last observation carried-forward is used for missing changes from baseline at Week 26 for patients with missing Week 26 who did not require rescue therapy.
  • a sensitivity analysis for the actual change from baseline in the MG-ADL total score at Week 26 is also performed.
  • Treatment groups are compared using ANCOVA analysis using the actual change from baseline in the MG-ADL total score at Week 26 with effects for treatment.
  • the baseline MG-ADL total score and the randomization stratification variable are also covariates in the model.
  • Last observation carried-forward is used for missing changes from baseline at Week 26.
  • a sensitivity analysis for the actual change from baseline in the MG-ADL total score at Week 26 is also performed.
  • treatment groups are compared using repeated measures model with effects for treatment and visits.
  • the baseline MG-ADL total score, the randomization stratification variable, and an indicator for the IST treatment status of the subject are also covariates in the model.
  • Subjects have an IST treatment status variable defined based on the IST treatments the subject receives.
  • summaries of changes from baseline in the MG-ADL total score at Week 26 are produced by treatment group for subjects who have failed ISTs.
  • the secondary efficacy analyses use the available 26-week data from the Study Period. Hypothesis testing comparing eculizumab treatment with placebo treatment for the secondary efficacy analyses are performed using a closed testing procedure with the following rank order:
  • the secondary endpoints that involve changes from baseline are analyzed using a worst-case ranked analysis of covariance (ANCOVA) like that described for the primary efficacy endpoints as the primary analysis for the particular secondary endpoint.
  • ANCOVA worst-case ranked analysis of covariance
  • the ranked ANCOVA will have effects for treatment, the baseline for the particular endpoint, and the randomization stratification variable.
  • a sensitivity analysis for the change from baseline in QMG at Week 26 is analyzed using repeated measures model with effects for treatment, visits, and baseline QMG score in order to compare treatment groups.
  • the randomization stratification variable is also a covariate in the model.
  • a sensitivity analysis for the actual change from baseline in QMG score at Week 26 will also be performed.
  • Treatment groups are compared using ANCOVA analysis using the actual change from baseline in the QMG score at Week 26 with effects for treatment.
  • the baseline QMG score and the randomization stratification variable are also covariates in the model. Last observation carried-forward will be used for missing changes from baseline at Week 26.
  • a sensitivity analysis for the change from baseline in MGC at Week 26 is analyzed using repeated measures model with effects for treatment, visits, and baseline MGC score in order to compare treatment groups.
  • the randomization stratification variable is also a covariate in the model.
  • a sensitivity analysis for the change from baseline in MG-QOL-15 at Week 26 is analyzed using repeated measures model with effects for treatment, visits, and baseline MG-QOL-15 score in order to compare treatment groups.
  • the randomization stratification variable is also a covariate in the model.
  • the proportion of subjects with at least a 3 point reduction in the MG-ADL total score from baseline to Week 26 with no rescue therapy are analyzed by the Cochran-Mantel-Haenszel test stratified by randomization stratification variable in order to compare eculizumab versus placebo.
  • the proportion of subjects with at least a 5 point reduction in the QMG total score from baseline to Week 26 with no rescue therapy are analyzed by the Cochran-Mantel-Haenszel test stratified by randomization stratification variable in order to compare eculizumab versus placebo.
  • Additional sensitivity analyses are performed in order that assess the impact of IST treatment status on the various secondary endpoints.
  • a sensitivity analysis for the change from baseline in the secondary endpoints (i.e., QMG, MGC, and MG-QOL-15) at Week 26 are analyzed using repeated measures model with effects for treatment, visits, and baseline score in order to compare treatment groups.
  • the randomization stratification variable and an indicator for the IST treatment status of the subject are also covariates in the model. Subjects will have an IST treatment status variable defined based on the IST treatments the subject receives.
  • the tertiary efficacy analyses for the Study Period include the following:
  • treatment groups are compared using Cox PH regression with robust variance estimation.
  • the randomization stratification variable is also a covariate in the model. Inference is based on the Wald test of the log hazard ratio.
  • the tertiary endpoints that involve changes from baseline are analyzed using a worst-case ranked ANCOVA like that described for the primary efficacy endpoints as the primary analysis for the particular tertiary endpoint.
  • the ranked ANCOVA has effects for treatment, the baseline for the particular endpoint, and the randomization stratification variable.
  • a sensitivity analysis for the change from baseline in NIF at Week 26 for subjects with abnormal NIF at baseline is analyzed using repeated measures model with effects for treatment, visits, and baseline NIF in order to compare treatment groups.
  • the randomization stratification variables are also covariates in the model.
  • a sensitivity analysis for the change from baseline in FVC is analyzed using repeated measures model with effects for treatment, visits, and baseline FVC in order to compare treatment groups.
  • the randomization stratification variable is also a covariate in the model.
  • a sensitivity analysis for the change from baseline in the MG-ADL individual items and sub-categories at Week 26 in subjects that are abnormal at baseline are analyzed using repeated measures model with effects for treatment, visits, and baseline MG-ADL individual item and sub-categories, as applicable for the analysis, in order to compare treatment groups.
  • the randomization stratification variable is also a covariate in the model.
  • a sensitivity analysis for the change from baseline in the MG-ADL individual items and sub-categories at Week 26 are performed using repeated measures model with effects for treatment, visits, and baseline MG-ADL individual item or sub-categories score, as applicable for the analysis, in order to compare treatment groups.
  • the randomization stratification variable is also a covariate in the model. Finally, similar sensitivity analyses and/or summaries are produced (depending on the number of subjects) in the subset of subjects who were normal at baseline and became abnormal after baseline in the MG-ADL individual items and sub-categories.
  • a summary of subjects going from normal to abnormal for NIF and FVC are presented.
  • a summary of subjects going from normal to abnormal for a particular MG-ADL individual items and sub-categories are produced.
  • the eculizumab treated group is compared to the placebo group and all hypothesis testing is two-sided and performed at the 0.05 level of significance, unless otherwise specified.
  • Estimates of treatment effect on efficacy parameters are accompanied by two-sided 95% confidence intervals for the effect size.
  • the extension trial lasts for 4 years (FPFV to LPLV).
  • the first visit occurs within 2 weeks of Visit 17 (Week 26) in the trial described above.
  • Visit 17 Week 26
  • “Home infusion” at selected visits is performed with permission of the primary investigator in accordance with regulations. Assessments, Treatment, Concomitant/Prohibited medications were performed as in the study described above.
  • the inclusion criteria for the extension trial was completion of the previous trial. Exclusion criteria were withdrawing from the previous trial and pregnancy or intention to get pregnant. IST treatment could be changed at the treating physician's discretion but rituximab was prohibited.
  • Efficacy was measured by MG-ADL, QMG, MGC, NIF, FVC, QOL, G-QOL15, Neuro-QOL Fatigue, EQ-5D and MGFA Post-Intervention Status.
  • the REGAIN study is a randomized, double-blind, placebo-controlled, multicenter trial evaluating the safety and efficacy of eculizumab in patients with refractory gMG.
  • the study enrolled and treated 125 adult patients across North America, South America, Europe, and Asia. Patients had a confirmed MG diagnosis with positive serologic test for anti-AChR antibodies. All patients had previously failed treatment with at least two immunosuppressive agents or failed treatment with at least one immunosuppressive agent and required chronic plasma exchange or IVIg, and had an MG-ADL total score ⁇ 6 at study entry.
  • the breakdown of the MGFA classification at screening was as follows: Class IIa 25 total patients; Class IIb 22 total patients; Class IIIa 36 total patients; Class IIIb 30 total patients; Class IVa 6 total patients; and Class IVb 6 total patients.
  • the patients were assigned to the placebo group as follows: Class IIa 15 (23.8%) total patients; Class IIb 14 (22.2%) total patients; Class IIIa 16 (25.4%) total patients; Class IIIb 13 (20.6%) total patients; Class IVa 2 (3.2%) total patients; and Class IVb 3 (4.8%) total patients.
  • the protocol defines clinical deterioration as a subject who has one of the following:
  • Rescue therapy is defined in the protocol as follows: Use of PE or IVIg will be allowed for subjects who experience a Clinical Deterioration as defined by this protocol. The rescue therapy used for a particular subject is at the discretion of the treating physician
  • PE is administered as a rescue therapy
  • supplemental IP (2 vials) are administered within 60 minutes after the end of each PE session.
  • Routine (per protocol schedule) IP administration is continued per the specified dose-administration schedule for the subject. If the subject is scheduled to receive the protocol-scheduled dose on the day of a PE session, then the scheduled dose is administered within 60 minutes after the end of the PE session.
  • One primary endpoint in the MG-ADL score at week 26 The score ranges from 0-24 and contains 3 bulbar items, 1 respiratory item, 2 gross motor or limb items, and 2 ocular items.
  • a clinically meaningful improvement in MG-ADL is defined as a 3 points or greater reduction. See Table 1.
  • Refractory gMG is an ultra-rare segment of MG—a debilitating, complement-mediated neuromuscular disease—in which patients have largely exhausted conventional therapy and continue to suffer profound muscle weakness throughout the body, resulting in slurred speech, impaired swallowing and choking, double vision, upper and lower extremity weakness, disabling fatigue, shortness of breath due to respiratory muscle weakness, and episodes of respiratory failure.
  • MG-ADL Myasthenia Gravis-Activities of Daily Living Profile
  • the current QMG scoring system consists of 13 items: ocular (2 items), facial (1 item), bulbar (2 items), gross motor (6 items), axial (1 item) and respiratory (1 item); each graded 0 to 3, with 3 being the most severe (see Table 2).
  • the range of total QMG score is 0-39.
  • the QMG scoring system is considered to be an objective evaluation of therapy for MG and is based on quantitative testing of sentinel muscle groups.
  • the MGFA task force has recommended that the QMG score be used in prospective studies of therapy for MG.
  • a clinically meaningful improvement in a patient's QMG would be a 5 point or greater reduction in score after 26 weeks of treatment.
  • the QMG score for the full data set was ⁇ 5 in the eculizumab treated group and therefore resulted in a clinically significant improvement for all patients not needing rescue or dropping out of the study. See Table 22 below and FIG. 11 for the results.
  • the MGC score was evaluated for all study participants over time.
  • the MGC is a validated assessment tool for measuring clinical status of subjects with MG (16).
  • the MGC assesses 10 important functional areas most frequently affected by MG and the scales are weighted for clinical significance that incorporate subject-reported outcomes (see Table 3).
  • MGC will be administered at Screening, Day 1, Weeks 1-4, 8, 12, 16, 20, and 26 or ET (Visits 1-6, 8, 10, 12, 14, and 17 or ET). Total scores range from 0-50.
  • a clinically meaningful improvement in a patient's MGC would be a 3 point or greater reduction in score after 26 weeks of treatment.
  • the MGC score for the full data set was ( ⁇ 10) in the eculizumab treated group and therefore resulted in a clinically significant improvement for all patients not needing rescue or dropping out of the study. See Table 23 above for the results.
  • the 15-item Myasthenia Gravis Qualify of Life scale (MG-QOL 15) is a health-related quality of life evaluative instrument specific to subjects with MG. See Table 4. MG-QOL15 was designed to provide information about subjects' perception of impairment and disability and the degree to which disease manifestations are tolerated and to be easy to administer and interpret. The MG-QOL 15 is completed by the subject. Total scores range from 0 to 60 and higher scores indicate greater extent of and dissatisfaction with MG-related dysfunction. A clinically meaningful improvement in a patient's MGQOL would be a decrease in score after 26 weeks of treatment.
  • the MG-QOL15 median score for the full data set was ( ⁇ 11.5) in the eculizumab treated group and therefore resulted in a clinically significant improvement for all patients not needing rescue or dropping out of the study. See Table 24 below for the results.
  • the Neuro-QOL Fatigue is a reliable and validated brief 19-item survey of fatigue completed by the subject. Higher scores indicate greater fatigue and greater impact of MG on activities (see Table 5). A clinically meaningful improvement in a patient's Neuro-QQL Fatigue score would be reflected in a decrease in score after 26 weeks of treatment.
  • the eculizumab treated group realized a clinically meaningful improvement (reduction) in their Neuro-QQL Fatigue score after 26 weeks of treatment.
  • QMG Quantitative Myasthenia Gravis
  • the second and third prospectively defined secondary efficacy endpoints of responder status in MG-ADL and QMG achieved p-values of ⁇ 0.05: the proportion of patients with at least a 3-point reduction in MG-ADL total score and no rescue therapy from baseline to week 26 with eculizumab treatment, compared to placebo, achieved a p-value of 0.0229, and the proportion of patients with at least a 5-point reduction in QMG total score and no rescue therapy from baseline to week 26 with eculizumab treatment compared to placebo achieved a p-value of 0.0018.
  • Pre-specified sensitivity analyses were prospectively defined to validate results for the primary and first secondary endpoints.
  • Three of the four prospectively defined MG-ADL sensitivity analyses achieved p-values ⁇ 0.05, including the sensitivity analysis around the primary endpoint for change from baseline in MG-ADL using repeated measures, which showed a mean change with eculizumab treatment at week 26 of ⁇ 4.2 versus a mean change with placebo at week 26 of ⁇ 2.3 and achieved a p-value of 0.0058.
  • Table 27 below contains sequences of anti-complement protein C5 specific humanized antibodies that can be used in treating refractory MG.
  • the antibody was an anti-C5 antibody such as eculizumab having three heavy chain complementarity determining regions (CDRs) as set forth in Table 27 using the Kabat definitions of CDRs as heavy chain CDR1 in SEQ ID NO: 1, heavy chain CDR2 in SEQ ID NO: 2, and heavy chain CDR3 in SEQ ID NO: 3.
  • the eculizumab light chain CDRs are set forth below as as light chain CDR1 in SEQ ID NO: 4, light chain CDR2 in SEQ ID NO: 5 and light chain CDR3 in SEQ ID NO: 6.
  • the heavy chain variable region of eculizumab is set forth in SEQ ID NO: 7 and the light chain variable region of eculizumab is set forth in SEQ ID NO: 8.
  • the complete heavy chain of eculizumab is set forth below as SEQ ID NO: 10 and the light chain is set forth below as SEQ ID NO: 11
  • the antibody may be an eculizumab variant known as BNJ441 and having selected mutations in the CDR regions combined with mutations in the Fc region to increase the T1 ⁇ 2 of the antibody in the patient.
  • the BNJ441 antibody has heavy chain variable region as set forth in SEQ ID NO: 12 and the light chain variable region of BNJ441 is set forth in SEQ ID NO: 8.
  • the complete heavy chain of BNJ441 is set forth below as SEQ ID NO: 14 and the light chain is set forth below as SEQ ID NO: 11
  • the antibody may be an anti-C5 antibody unrelated to eculizumab such as the 7086 antibody and having three heavy chain complementarity determining regions (CDRs) as set forth in Table Table 27 using the Kabat definitions of CDRs as heavy chain CDR1 in SEQ ID NO: 21, heavy chain CDR2 in SEQ ID NO: 22, and heavy chain CDR3 in SEQ ID NO: 23.
  • the 7086 antibody light chain CDRs are set forth below as light chain CDR1 in SEQ ID NO: 24, light chain CDR2 in SEQ ID NO: 25 and light chain CDR3 in SEQ ID NO: 26.
  • the heavy chain variable region of 7086 is set forth in SEQ ID NO: 27 and the light chain variable region of 7086 is set forth in SEQ ID NO: 28.
  • the antibody may be an anti-C5 antibody unrelated to eculizumab such as the 8110 antibody and having three heavy chain complementarity determining regions (CDRs) as set forth in Table 27 using the Kabat definitions of CDRs as heavy chain CDR1 in SEQ ID NO: 29, heavy chain CDR2 in SEQ ID NO: 30, and heavy chain CDR3 in SEQ ID NO: 31.
  • the 7086 antibody light chain CDRs are set forth below as light chain CDR1 in SEQ ID NO: 32, light chain CDR2 in SEQ ID NO: 33 and light chain CDR3 in SEQ ID NO: 34.
  • the heavy chain variable region of 8110 is set forth in SEQ ID NO: 35 and the light chain variable region of 8110 is set forth in SEQ ID NO: 36.
  • the antibody may be an anti-C5 antibody or antigen binding fragment thereof comprising a heavy chain variable region amino acid sequence according to SEQ ID NO: 37 and a light chain variable region amino acid sequence according to SEQ ID NO: 38.
  • the objective of the example was to assess the time course of response in patients who demonstrated a clinically meaningful response to eculizumab and the proportion of patients who had clinically meaningful relevant responses on both the MG-ADL and the QMG.
  • the Myasthenia Gravis Activities of Daily Living is a physician-directed, patient-reported measure of symptom severity related to MG-specific ADLs (Muppidi, Ann. N.Y. Acad. Sci. 1274: 114-19 (2012)), and the Quantitative Myasthenia Gravis (QMG) tool is a clinician-reported measure of muscle strength (Barohn et al., Ann. N.Y Acad. Sci. 841: 769-72 (1998)).
  • Pre-specified responder analyses included the proportion of patients who responded with ⁇ 3-point improvement in MG-ADL total score with no rescue; and the proportion of patients with ⁇ 5-point improvement in QMG total score with no rescue.
  • response was defined as an improvement of ⁇ 3 points from baseline in the MG-ADL total score and improvement of ⁇ 5 points from baseline in the QMG total score, with no rescue therapy.
  • ⁇ 3-point improvement for MG-ADL and ⁇ 5-point improvement for QMG thresholds of ⁇ 4, 5, 6, 7, and 8 for MG-ADL and ⁇ 6, 7, 8, 9, and 10 for QMG were also examined.
  • P values from a Cochran—Mantel—Haenszel (CMH) test were provided for the more stringent criteria to aid interpretation.
  • FIG. 13 More patients receiving eculizumab than those who received placebo experienced clinically meaningful responses as defined above, and also clinically meaningful relevant responses based on the more stringent thresholds for both MG-ADL and QMG total scores ( FIG. 13 ).
  • MG-ADL responder analyses conducted at each assessment date over the 26-week study are shown in FIG. 14 (the proportion of patients with a ⁇ 3, 5, or 8 point change in the MG-ADL).
  • FIG. 15 the proportion of patients with a greater than 5, 7, or 10 point change in the QMG.
  • the response rate was substantially higher in eculizumab-treated patients (40.3%) than in placebo-treated patients (12.7%), showing a clinically significant response by both patient- and physician-assessed outcome measures in patients treated with eculizumab.
  • the superiority of response to eculizumab over placebo becomes more pronounced, with odds ratios exceeding 10.
  • ECU-MG-302. Patients who completed REGAIN were allowed to continue into an open-label extension study known as ECU-MG-302. Each patient enrolled in the extension trial underwent an initial 4-week blinded induction before receiving open-label eculizumab maintenance treatment (1200 mg every 2 weeks). MG-ADL, QMG, MGC, and MG-QOL15 scores and safety were assessed.
  • IST background immunosuppressant therapy
  • AChI acetylcholinesterase inhibitors
  • the MG-ADL total score in eculizumab/eculizumab patients was unchanged from open-label baseline through week 52.
  • the placebo/eculizumab patients did not demonstrate rapid improvement in MG-ADL total score from open-label baseline with a change from ECU-MG-302 baseline in MG-ADL total score observed as early as week 1 ( ⁇ 1.6 [ ⁇ 2.28, ⁇ 0.89]; p ⁇ 0.0001).
  • the majority of the overall treatment effect was achieved by week 4 ( ⁇ 2.4[ ⁇ 3.19, ⁇ 1.71]; p ⁇ 0.0001) during the blind induction phase, and was sustained through week 52 ( ⁇ 2.7 [ ⁇ 3.73, ⁇ 1.63]; p ⁇ 0.0001).

Landscapes

  • Health & Medical Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • General Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Engineering & Computer Science (AREA)
  • Veterinary Medicine (AREA)
  • Public Health (AREA)
  • Animal Behavior & Ethology (AREA)
  • Heart & Thoracic Surgery (AREA)
  • Molecular Biology (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Immunology (AREA)
  • Biochemistry (AREA)
  • Biophysics (AREA)
  • Genetics & Genomics (AREA)
  • Pharmacology & Pharmacy (AREA)
  • General Chemical & Material Sciences (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Anesthesiology (AREA)
  • Vascular Medicine (AREA)
  • Biomedical Technology (AREA)
  • Hematology (AREA)
  • Neurology (AREA)
  • Orthopedic Medicine & Surgery (AREA)
  • Physical Education & Sports Medicine (AREA)
  • Medicines Containing Antibodies Or Antigens For Use As Internal Diagnostic Agents (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)
  • Peptides Or Proteins (AREA)
US15/595,890 2016-05-27 2017-05-15 Methods for Treatment of Refractory Generalized Myasthenia Gravis Abandoned US20170342139A1 (en)

Priority Applications (2)

Application Number Priority Date Filing Date Title
US15/595,890 US20170342139A1 (en) 2016-05-27 2017-05-15 Methods for Treatment of Refractory Generalized Myasthenia Gravis
US17/150,005 US20210246196A1 (en) 2016-05-27 2021-01-15 Methods for treatment of refractory generalized myasthenia gravis

Applications Claiming Priority (5)

Application Number Priority Date Filing Date Title
US201662342321P 2016-05-27 2016-05-27
US201662346168P 2016-06-06 2016-06-06
US201762489240P 2017-04-24 2017-04-24
US201762500643P 2017-05-03 2017-05-03
US15/595,890 US20170342139A1 (en) 2016-05-27 2017-05-15 Methods for Treatment of Refractory Generalized Myasthenia Gravis

Related Child Applications (1)

Application Number Title Priority Date Filing Date
US17/150,005 Continuation US20210246196A1 (en) 2016-05-27 2021-01-15 Methods for treatment of refractory generalized myasthenia gravis

Publications (1)

Publication Number Publication Date
US20170342139A1 true US20170342139A1 (en) 2017-11-30

Family

ID=60412593

Family Applications (2)

Application Number Title Priority Date Filing Date
US15/595,890 Abandoned US20170342139A1 (en) 2016-05-27 2017-05-15 Methods for Treatment of Refractory Generalized Myasthenia Gravis
US17/150,005 Pending US20210246196A1 (en) 2016-05-27 2021-01-15 Methods for treatment of refractory generalized myasthenia gravis

Family Applications After (1)

Application Number Title Priority Date Filing Date
US17/150,005 Pending US20210246196A1 (en) 2016-05-27 2021-01-15 Methods for treatment of refractory generalized myasthenia gravis

Country Status (11)

Country Link
US (2) US20170342139A1 (fr)
EP (2) EP3463461A4 (fr)
JP (2) JP2019517473A (fr)
KR (1) KR20190012199A (fr)
CN (1) CN109475630A (fr)
AU (1) AU2017269839A1 (fr)
BR (1) BR112018073945A2 (fr)
CA (1) CA3024618A1 (fr)
MX (1) MX2018014560A (fr)
RU (1) RU2018141451A (fr)
WO (1) WO2017205101A1 (fr)

Cited By (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2020058759A1 (fr) * 2018-09-17 2020-03-26 Kyoto University Administration d'un agent anti-c5 pour le traitement d'une lésion ou d'une insuffisance hépatique
CN113614106A (zh) * 2019-02-14 2021-11-05 亚力兄制药公司 抗c5抗体用于治疗全身型重症肌无力的剂量和施用

Families Citing this family (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP3463461A4 (fr) * 2016-05-27 2020-05-20 Alexion Pharmaceuticals, Inc. Procédés de traitement de la myasthénie grave généralisée réfractaire
KR102534895B1 (ko) 2016-06-14 2023-05-24 리제너론 파아마슈티컬스, 인크. 항-c5 항체 및 이의 용도
MX2020006113A (es) 2017-12-13 2020-08-24 Regeneron Pharma Combinaciones de anticuerpos anti-c5 y usos de las mismas.
WO2020106724A1 (fr) * 2018-11-20 2020-05-28 Alexion Pharmaceuticals, Inc. Méthodes de traitement de la myasthénie grave généralisée réfractaire chez des patients pédiatriques
WO2021026160A1 (fr) * 2019-08-05 2021-02-11 Alexion Pharmaceuticals, Inc. Méthodes de traitement de la myasthénie grave généralisée réfractaire avec de l'eculizumab

Family Cites Families (10)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6074642A (en) 1994-05-02 2000-06-13 Alexion Pharmaceuticals, Inc. Use of antibodies specific to human complement component C5 for the treatment of glomerulonephritis
JP4021194B2 (ja) 2001-12-28 2007-12-12 シャープ株式会社 薄膜トランジスタ装置の製造方法
US9415102B2 (en) * 2002-09-06 2016-08-16 Alexion Pharmaceuticals, Inc. High concentration formulations of anti-C5 antibodies
US7807378B2 (en) * 2005-12-29 2010-10-05 Industrial Technology Research Institute (Itri) Method of diagnosing myasthenia gravis and kits therefor
WO2010015608A1 (fr) 2008-08-05 2010-02-11 Novartis Ag Compositions et procédés pour des anticorps ciblant une protéine du complément c5
EP3101031A1 (fr) * 2008-11-10 2016-12-07 Alexion Pharmaceuticals, Inc. Procédés et compositions de traitement de troubles associés à un complément
TWI622597B (zh) 2011-03-28 2018-05-01 賽諾菲公司 具有交叉結合區定向之雙重可變區類抗體結合蛋白
NZ631007A (en) * 2014-03-07 2015-10-30 Alexion Pharma Inc Anti-c5 antibodies having improved pharmacokinetics
EP3463461A4 (fr) * 2016-05-27 2020-05-20 Alexion Pharmaceuticals, Inc. Procédés de traitement de la myasthénie grave généralisée réfractaire
WO2018044858A2 (fr) * 2016-08-29 2018-03-08 The Cleveland Clinic Foundation Immunisation à l'aide d'épitopes c5 pour la production d'anticorps anti-c5 autologues

Cited By (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2020058759A1 (fr) * 2018-09-17 2020-03-26 Kyoto University Administration d'un agent anti-c5 pour le traitement d'une lésion ou d'une insuffisance hépatique
CN113614106A (zh) * 2019-02-14 2021-11-05 亚力兄制药公司 抗c5抗体用于治疗全身型重症肌无力的剂量和施用

Also Published As

Publication number Publication date
EP3463461A4 (fr) 2020-05-20
JP2019517473A (ja) 2019-06-24
JP2022126773A (ja) 2022-08-30
EP3909609A1 (fr) 2021-11-17
EP3463461A1 (fr) 2019-04-10
MX2018014560A (es) 2019-08-12
US20210246196A1 (en) 2021-08-12
RU2018141451A3 (fr) 2020-09-21
CA3024618A1 (fr) 2017-11-30
RU2018141451A (ru) 2020-06-29
KR20190012199A (ko) 2019-02-08
WO2017205101A1 (fr) 2017-11-30
BR112018073945A2 (pt) 2019-02-26
AU2017269839A1 (en) 2018-11-22
CN109475630A (zh) 2019-03-15

Similar Documents

Publication Publication Date Title
US20210246196A1 (en) Methods for treatment of refractory generalized myasthenia gravis
US9018170B2 (en) Reduced volume formulation of glatiramer acetate and methods of administration
JP2024069373A (ja) 視神経脊髄炎の処置のためのエクリズマブ
US20200331993A1 (en) Dosage and administration of anti-c5 antibodies for treatment of generalized myasthenia gravis
US20240059783A1 (en) Anti-ox40 antagonistic antibodies and dosage for the treatment of ox40-mediated disorders
US20220259305A1 (en) Methods for treatment of refractory generalized myasthenia gravis with eculizumab
EP3979983A1 (fr) Composition de toxine botulique liquide pour le traitement de rides glabellaires et de rides canthales latérales modérées à très sévères
CA3101767A1 (fr) Compositions et procedes de traitement du psoriasis
CN114401742B (zh) 通过转换疗法治疗rms
US20220002393A1 (en) Methods for treatment of refractory generalized myasthenia gravis in pediatric patients
Ryan et al. Providing pharmaceutical care to the multiple sclerosis patient
US12016921B2 (en) Methods for treatment of psoriasis with an anti-Apo B100 antibody
WO2023235545A2 (fr) Dosage et administration de polypeptides de fusion pour le traitement de la myasthénie grave
WO2023144699A1 (fr) Méthodes de traitement de sujets atteints du psoriasis en plaques du cuir chevelu
TW202348621A (zh) 用TACI-Fc融合蛋白治療重症肌無力的方法
EP4370547A1 (fr) Dosage et administration d'anticorps anti-c5 pour le traitement de la myasthénie gravis
EA046524B1 (ru) Доза и введение антител к c5 для лечения генерализованной миастении гравис
WO2023215384A2 (fr) Utilisation d'un inhibiteur de la myostatine pour le traitement de l'amyotrophie spinale

Legal Events

Date Code Title Description
AS Assignment

Owner name: ALEXION PHARMACEUTICALS, INC., CONNECTICUT

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:BEDROSIAN, CAMILLE;O'BRIEN, FANNY;WANG, JING JING;SIGNING DATES FROM 20170517 TO 20170531;REEL/FRAME:042553/0038

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

Free format text: NON FINAL ACTION MAILED

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

Free format text: RESPONSE TO NON-FINAL OFFICE ACTION ENTERED AND FORWARDED TO EXAMINER

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

Free format text: FINAL REJECTION MAILED

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: RESPONSE TO NON-FINAL OFFICE ACTION ENTERED AND FORWARDED TO EXAMINER

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

Free format text: FINAL REJECTION MAILED

STCB Information on status: application discontinuation

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