US20090220508A1 - Treatment Of Paroxysmal Nocturnal Hemoglobinuria Patients By An Inhibitor Of Complement - Google Patents

Treatment Of Paroxysmal Nocturnal Hemoglobinuria Patients By An Inhibitor Of Complement Download PDF

Info

Publication number
US20090220508A1
US20090220508A1 US12/225,040 US22504007A US2009220508A1 US 20090220508 A1 US20090220508 A1 US 20090220508A1 US 22504007 A US22504007 A US 22504007A US 2009220508 A1 US2009220508 A1 US 2009220508A1
Authority
US
United States
Prior art keywords
patient
antibody
antibodies
life
complement
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
US12/225,040
Other languages
English (en)
Inventor
Leonard Bell
Russell P. Rother
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
Family has litigation
First worldwide family litigation filed litigation Critical https://patents.darts-ip.com/?family=38222520&utm_source=google_patent&utm_medium=platform_link&utm_campaign=public_patent_search&patent=US20090220508(A1) "Global patent litigation dataset” by Darts-ip is licensed under a Creative Commons Attribution 4.0 International License.
Priority to US12/225,040 priority Critical patent/US20090220508A1/en
Application filed by Alexion Pharmaceuticals Inc filed Critical Alexion Pharmaceuticals Inc
Assigned to ALEXION PHARMACEUTICALS, INC. reassignment ALEXION PHARMACEUTICALS, INC. ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: ROTHER, RUSSELL P., BELL, LEONARD
Publication of US20090220508A1 publication Critical patent/US20090220508A1/en
Priority to US13/426,973 priority patent/US20120237515A1/en
Priority to US15/148,839 priority patent/US9718880B2/en
Priority to US15/260,888 priority patent/US9725504B2/en
Priority to US15/284,015 priority patent/US9732149B2/en
Priority to US15/642,096 priority patent/US10590189B2/en
Priority to US16/750,978 priority patent/US20200392216A1/en
Priority to US16/804,567 priority patent/US10703809B1/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
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/70Carbohydrates; Sugars; Derivatives thereof
    • A61K31/7088Compounds having three or more nucleosides or nucleotides
    • A61K31/7105Natural ribonucleic acids, i.e. containing only riboses attached to adenine, guanine, cytosine or uracil and having 3'-5' phosphodiester links
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/395Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/0012Galenical forms characterised by the site of application
    • A61K9/0019Injectable compositions; Intramuscular, intravenous, arterial, subcutaneous administration; Compositions to be administered through the skin in an invasive manner
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • A61P1/14Prodigestives, e.g. acids, enzymes, appetite stimulants, antidyspeptics, tonics, antiflatulents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P13/00Drugs for disorders of the urinary system
    • A61P13/02Drugs for disorders of the urinary system of urine or of the urinary tract, e.g. urine acidifiers
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P15/00Drugs for genital or sexual disorders; Contraceptives
    • A61P15/10Drugs for genital or sexual disorders; Contraceptives for impotence
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P17/00Drugs for dermatological disorders
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P19/00Drugs for skeletal disorders
    • 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
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/04Centrally acting analgesics, e.g. opioids
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/18Antipsychotics, i.e. neuroleptics; Drugs for mania or schizophrenia
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/20Hypnotics; Sedatives
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/26Psychostimulants, e.g. nicotine, cocaine
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/28Drugs for disorders of the nervous system for treating neurodegenerative disorders of the central nervous system, e.g. nootropic agents, cognition enhancers, drugs for treating Alzheimer's disease or other forms of dementia
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P29/00Non-central analgesic, antipyretic or antiinflammatory agents, e.g. antirheumatic agents; Non-steroidal antiinflammatory drugs [NSAID]
    • A61P29/02Non-central analgesic, antipyretic or antiinflammatory agents, e.g. antirheumatic agents; Non-steroidal antiinflammatory drugs [NSAID] without antiinflammatory effect
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P3/00Drugs for disorders of the metabolism
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P3/00Drugs for disorders of the metabolism
    • A61P3/02Nutrients, e.g. vitamins, minerals
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/04Antibacterial agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/02Immunomodulators
    • A61P37/04Immunostimulants
    • 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
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P7/00Drugs for disorders of the blood or the extracellular fluid
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P7/00Drugs for disorders of the blood or the extracellular fluid
    • A61P7/02Antithrombotic agents; Anticoagulants; Platelet aggregation inhibitors
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P7/00Drugs for disorders of the blood or the extracellular fluid
    • A61P7/06Antianaemics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • A61P9/12Antihypertensives
    • 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
    • A61K38/00Medicinal preparations containing peptides
    • 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
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/50Immunoglobulins specific features characterized by immunoglobulin fragments
    • C07K2317/51Complete heavy chain or Fd fragment, i.e. VH + CH1
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/50Immunoglobulins specific features characterized by immunoglobulin fragments
    • C07K2317/515Complete light chain, i.e. VL + CL
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/50Immunoglobulins specific features characterized by immunoglobulin fragments
    • C07K2317/56Immunoglobulins specific features characterized by immunoglobulin fragments variable (Fv) region, i.e. VH and/or VL
    • C07K2317/565Complementarity determining region [CDR]

Definitions

  • Paroxysmal nocturnal hemoglobinuria is an acquired hematologic disease that results from clonal expansion of hematopoietic stem cells with somatic mutations in the X-linked gene called PIG-A. 1,2 Mutations in PIG-A lead to an early block in the synthesis of glycosylphosphatidylinositol (GPI)-anchors, which are required to tether many proteins to the cell surface. Consequently, PNH blood cells have a partial (type II) or complete (type III) deficiency of GPI-anchored proteins.
  • GPI glycosylphosphatidylinositol
  • Intravascular hemolysis is a prominent feature of PNH and a direct result of the absence of the GPI-anchored complement regulatory protein CD59. 3,4 Under normal circumstances, CD59 blocks the formation of the terminal complement complex (also called the membrane attack complex) on the cell surface, thereby preventing erythrocyte lysis and platelet activation.
  • the terminal complement complex also called the membrane attack complex
  • Eculizumab is a humanized monoclonal antibody directed against the terminal complement protein C5. 11 In a preliminary, 12-week, open-label clinical study in 111 PNH patients, eculizumab was shown to reduce intravascular hemolysis and transfusion requirements. 12 However, this unblinded study involved a small number of patients with no control arm and without protocol-driven transfusion standards.
  • the application provides a method to improve at least one aspect of the quality of life of a patient suffering from paroxysmal nocturnal hemoglobinuria, said method comprising administering to said patient in need thereof a compound which inhibits complement or inhibits formation of C5b-9.
  • the application provides a method to improve at least one aspect of the quality of life of a patient suffering from paroxysmal nocturnal hemoglobinuria, said method comprising administering to said patient in need thereof a compound which inhibits intravascular hemolysis. In certain embodiments, said method results in a greater than 30% reduction in LDH in said patient.
  • the application provides a method to improve at least one aspect of the quality of life of an anemic patient whose anemia results at least in part from hemolysis, said method comprising administering to said patient in need thereof a compound which inhibits intravascular hemolysis, wherein said patient remains anemic. In certain embodiments, said method results in a greater than 30% reduction in LDH in said patient.
  • the application provides a method of prolonging the health-adjusted life expectancy of a patient comprising administering to said patient in need thereof a compound which inhibits formation of C5b-9.
  • said patient is anemic.
  • said patient remains anemic following treatment.
  • said patient has a hemoglobin level less than i) 14 g/dL if a man or ii) 12 g/dL if a woman.
  • said patient has a hemoglobin level less than i) 13 g/dL if a man or ii) 11 g/dL if a woman.
  • said patient has a hemoglobin level less than i) 12 g/dL if a man or ii) 10 g/dL if a woman. In certain embodiments, said patient suffers from paroxysmal nocturnal hemoglobinuria.
  • the application provides a pharmaceutical composition comprising an antibody that binds C5 or an active antibody fragment thereof.
  • the antibody that binds C5 or an active antibody fragment thereof is eculizumab.
  • the antibody that binds C5 or an active antibody fragment thereof is pexelizumab.
  • the pharmaceutical formulations of the application may be administered to a subject, particularly a subject having PNH.
  • the application provides a method of treating a patient suffering from paroxysmal nocturnal hemoglobinuria by administering a pharmaceutical composition comprising an antibody that binds C5 or an active antibody fragment thereof.
  • a pharmaceutical composition comprising an antibody that binds C5 or an active antibody fragment thereof.
  • the antibody that binds C5 or an active antibody fragment thereof is eculizumab.
  • the antibody that binds C5 or an active antibody fragment thereof is pexelizumab.
  • the pharmaceutical formulations of the application may be administered to a subject, particularly a subject having PNH.
  • kits comprising a pharmaceutical composition of the application.
  • the kit further comprises at least one component of a closed sterile system.
  • Components of the closed sterile system include, but are not limited to, needles, syringes, catheter based syringes, needle based injection devices, needle-less injection devices, filters, tubing, valves and cannulas.
  • the kit comprise components for the removal of a preservative from the composition. Such components include filters, syringes, vials, containers, tubing, etc.
  • said quality of life is measured by a FACIT-Fatigue score.
  • the FACIT-Fatigue score increases by at least 3 points. In certain embodiments, the FACIT-Fatigue score increases by ⁇ 4 points.
  • said quality of life is measured by an EORTC QLQ-C30 score.
  • said EORTC QLQ-C30 score improves by ⁇ 10% of the pretreatment score.
  • said aspect of the quality of life as measured by an EORTC QLQ-C30 score is selected from the group consisting of a) global health status, b) physical functioning, c) emotional functioning, d) cognitive functioning, e) role functioning, f) social functioning, g) fatigue, h) pain, i) dyspnea, j) appetite loss, and k) insomnia.
  • said aspect of quality of life is fatigue.
  • said compound is selected from the group consisting of CR1, LEX-CR1, MCP, DAF, CD59, Factor H, cobra venom factor, FUT-175, complestatin, and K76 COOH.
  • said compound is a steroid that suppresses complement.
  • said compound is selected from the group consisting of antibodies, active antibody fragments, soluble complement inhibitory compounds, proteins, soluble complement inhibitors with a lipid tail, protein fragments, peptides, small organic compounds, RNA aptamers, L-RNA aptamers, spiegelmers, antisense compounds, serine protease inhibitors, double stranded RNA, small interfering RNA, locked nucleic acid inhibitors, and peptide nucleic acid inhibitors.
  • said compound is an antibody or an active antibody fragment.
  • said antibody or active antibody fragment is selected from the group consisting of a) polyclonal antibodies, b) monoclonal antibodies, c) single chain antibodies, d) chimeric antibodies, e) humanized antibodies, f) Fabs, g) F(ab′)s, h) F(ab′) 2 s, i) Fvs, j) diabodies, and k) human antibodies.
  • said antibody or an active antibody fragment thereof binds C5. In certain embodiments, said antibody or active antibody fragment blocks C5 cleavage. In certain embodiments, said antibody or active antibody fragment inhibits the formation of C5b-9. In certain embodiments, said antibody is eculizumab. In certain embodiments, said antibody or active antibody fragment is administered for at least 6 months. In certain embodiments, said patient has aplastic anemia or myelodysplastic syndrome.
  • said antibody that binds C5 or an active antibody fragment thereof is administered in a single unit dosage form.
  • the single unit dosage form is a 300 mg unit dosage form.
  • the single unit dosage form is lyophilized.
  • the single unit dosage form is a sterile solution.
  • the single unit dosage form is a preservative free formulation.
  • the 300 mg single-use dosage form comprises 30 ml of a 10 mg/ml sterile, preservative free solution.
  • the antibody that binds C5 or an active antibody fragment thereof comprises an altered constant region, wherein said antibody or antigen-binding fragment exhibits decreased effector function relative to an anti-CDCP1 antibody with a native constant region.
  • decreased effector function comprises one or more properties of the following group: a) decreased antibody-dependent cell-mediated cytotoxicity (ADCC), and b) decreased complement dependent cytotoxicity (CDC), compared to an anti-CDCP1 antibody with a native constant region.
  • the altered constant region comprises a G2/G4 construct in place of the G1 domain.
  • the antibody that binds C5 or an active antibody fragment thereof comprises a heavy chain variable region and a light chain variable region, wherein the heavy chain variable region comprises one or more CDR regions having an amino acid sequence selected from the group consisting of SEQ ID NO:5, SEQ ID NO:6, or SEQ ID NO:7, and wherein the light chain variable region comprises one or more CDR regions having an amino acid sequence selected from the group consisting of SEQ ID NO:8, SEQ ID NO:9, or SEQ ID NO:10.
  • the antibody that binds C5 or an active antibody fragment thereof comprises a heavy chain variable region and a light chain variable region, wherein the heavy chain variable region consists of SEQ ID NO: 1 and the light chain variable region consists of SEQ ID NO: 3.
  • the pharmaceutical composition comprises eculizumab. In certain embodiments, the pharmaceutical composition comprises pexelizumab. In certain embodiments, the antibody that binds C5 or an active antibody fragment thereof comprises a heavy chain and a light chain, wherein the heavy chain consists of SEQ ID NO: 2 and the light chain consists of SEQ TD NO: 4.
  • said patient is anemic. In certain embodiments, said patient remains anemic following treatment. In certain embodiments, said patient has a hemoglobin level less than i) 14 g/dL if a man or ii) 12 g/dL if a woman. In certain embodiments, said patient has a hemoglobin level less than i) 13 g/dL if a man or ii) 11 g/dL if a woman. In certain embodiments, said patient has a hemoglobin level less than i) 12 g/dL if a man or ii) 10 g/dL if a woman.
  • said health-adjusted life expectancy is measured according to a unit selected from the group consisting of Years of potential life lost, Disability-free life expectancy, Health-adjusted life year, Quality adjusted life year, Healthy years equivalents, Healthy days gained, Episode-free day, Q-TWiST, Health Utilities Index, or Years of healthy life.
  • the health-adjusted life expectancy in a subject is prolonged by at least one day. In certain embodiments, the health-adjusted life expectancy in a subject is prolonged by at least week. In certain embodiments, the health-adjusted life expectancy in a subject is prolonged by at least one month. In certain embodiments, the health-adjusted life expectancy in a subject is prolonged by at least one year.
  • the pharmaceutical composition is in a single unit dosage form. In certain embodiments, the single unit dosage form is a 300 mg unit dosage form. In certain embodiments, the pharmaceutical composition is lyophilized. In certain embodiments, the pharmaceutical composition is a sterile solution. In certain embodiments, the pharmaceutical composition is a preservative free formulation. In certain embodiments, the pharmaceutical composition comprises a 300 mg single-use formulation of 30 ml of a 10 mg/ml sterile, preservative free solution. In certain embodiments, the pharmaceutical composition comprises an antibody that binds C5 or an active antibody fragment thereof.
  • FIGS. 1A-B show that eculizumab treatment decreases intravascular hemolysis and increases PNH type III erythrocytes.
  • FIG. 1A shows the degree of intravascular hemolysis in PNH patients, demonstrated by mean lactate dehydrogenase (LDH) levels.
  • FIG. 1B shows the mean proportion of PNH type III erythrocytes assessed for placebo- and eculizumab-treated patients.
  • LDH lactate dehydrogenase
  • FIG. 2 shows the effect of eculizumab treatment on transfusion requirements in PNH patients. This is a Kaplan-Meier plot of time to first transfusion for eculizumab- and placebo-treated patients from baseline through week 26. The P value is from the log rank analysis.
  • FIG. 3 shows the effect of eculizumab on fatigue assessed by the FACIT-Fatigue Instrument. Quality of Life scores were assessed using the Functional Assessment of Chronic Illness Therapy-Fatigue (FACIT-Fatigue) instrument. Values for change from baseline to 26 weeks represent least-square means. A positive change indicates an improvement and a negative change indicates deterioration in the FACIT-Fatigue measures of quality of life.
  • FACIT-Fatigue Functional Assessment of Chronic Illness Therapy-Fatigue
  • derived from means “obtained from” or “produced by” or “descending from”.
  • genetically altered antibodies means antibodies wherein the amino acid sequence has been varied from that of a native antibody. Because of the relevance of recombinant DNA techniques to this application, one need not be confined to the sequences of amino acids found in natural antibodies; antibodies can be redesigned to obtain desired characteristics. The possible variations are many and range from the changing of just one or a few amino acids to the complete redesign of, for example, the variable or constant region. Changes in the constant region will, in general, be made in order to improve or alter characteristics, such as complement fixation, interaction with membranes and other effector functions. Changes in the variable region will be made in order to improve the antigen binding characteristics.
  • an antigen-binding fragment of an antibody refers to any portion of an antibody that retains the binding utility to the antigen.
  • An exemplary antigen-binding fragment of an antibody is the heavy chain and/or light chain CDR, or the heavy and/or light chain variable region.
  • homologous in the context of two nucleic acids or polypeptides refers to two or more sequences or subsequences that have at least about 85%, at least 90%, at least 95%, or higher nucleotide or amino acid residue identity, when compared and aligned for maximum correspondence, as measured using the following sequence comparison method and/or by visual inspection.
  • the “homolog” exists over a region of the sequences that is about 50 residues in length, at least about 100 residues, at least about 150 residues, or over the full length of the two sequences to be compared.
  • Percent (%) sequence identity with respect to a specified subject sequence, or a specified portion thereof, may be defined as the percentage of nucleotides or amino acids in the candidate derivative sequence identical with the nucleotides or amino acids in the subject sequence (or specified portion thereof), after aligning the sequences and introducing gaps, if necessary to achieve the maximum percent sequence identity, as generated by the program WU-BLAST-2.0a19 (Altschul et al., J. Mol. Biol. 215:403-410 (1997); http://blast.wustl.edu/blast/README.htm-l) with search parameters set to default values.
  • the HSP S and HSP S2 parameters are dynamic values and are established by the program itself depending upon the composition of the particular sequence and composition of the particular database against which the sequence of interest is being searched.
  • a “% identity value” is determined by the number of matching identical nucleotides or amino acids divided by the sequence length for which the percent identity is being reported.
  • the present disclosure relates to a method of treating paroxysmal nocturnal hemoglobinuria (“PNH”), more specifically to improving certain aspects of quality of life which are impaired in PNH patients, and other hemolytic diseases in mammals.
  • PNH paroxysmal nocturnal hemoglobinuria
  • the methods of treating hemolytic diseases, which are described herein involve using compounds which bind to or otherwise block the generation and/or activity of one or more complement components.
  • the present methods have been found to provide surprising results. For instance, hemolysis rapidly ceases upon administration of the compound which binds to or otherwise blocks the generation and/or activity of one or more complement components, with hemoglobinuria being significantly reduced after treatment.
  • hemolytic patients can be rendered less dependent on transfusions or transfusion-independent for extended periods (twelve months or more), well beyond the 120 day life cycle of red blood cells.
  • type III red blood cell count can be increased dramatically in the midst of other mechanisms of red blood cell lysis (non-complement mediated and/or earlier complement component mediated e.g., Cb3).
  • Another example of a surprising result is that symptoms resolved, indicating that NO serum levels were increased enough even in the presence of other mechanisms of red blood cell lysis.
  • the complement system acts in conjunction with other immunological systems of the body to defend against intrusion of cellular and viral pathogens.
  • complement proteins There are at least 25 complement proteins, which are found as a complex collection of plasma proteins and membrane cofactors.
  • the plasma proteins make up about 10% of the globulins in vertebrate serum.
  • Complement components achieve their immune defensive functions by interacting in a series of intricate but precise enzymatic cleavage and membrane binding events.
  • the resulting complement cascade leads to the production of products with opsonic, immunoregulatory, and lytic functions.
  • the complement cascade progresses via the classical pathway or the alternative pathway. These pathways share many components and, while they differ in their initial steps, they converge and share the same “terminal complement” components (C5 through C9) responsible for the activation and destruction of target cells.
  • the classical complement pathway is typically initiated by antibody recognition of and binding to an antigenic site on a target cell.
  • the alternative pathway is usually antibody independent and can be initiated by certain molecules on pathogen surfaces. Both pathways converge at the point where complement component C3 is cleaved by an active protease (which is different in each pathway) to yield C3a and C3b. Other pathways activating complement attack can act later in the sequence of events leading to various aspects of complement function.
  • C3a is an anaphylatoxin.
  • C3b binds to bacterial and other cells, as well as to certain viruses and immune complexes, and tags them for removal from the circulation.
  • C3b in this role is known as opsonin.
  • the opsonic function of C3b is considered to be the most important anti-infective action of the complement system. Patients with genetic lesions that block C3b function are prone to infection by a broad variety of pathogenic organisms, while patients with lesions later in the complement cascade sequence, i.e., patients with lesions that block C5 functions, are found to be more prone only to Neisseria infection, and then only somewhat more prone (Fearon, 1983).
  • C3b also forms a complex with other components unique to each pathway to form classical or alternative C5 convertase, which cleaves C5 into C5a and C5b.
  • C3 is thus regarded as the central protein in the complement reaction sequence since it is essential to both the alternative and classical pathways (Wurzner et al., 1991).
  • This property of C3b is regulated by the serun protease Factor 1, which acts on C3b to produce iC3b. While still functional as opsonin, iC3b cannot form an active C5 convertase.
  • C5 is a 190 kDa beta globulin found in normal serum at approximately 75 ⁇ g/mL (0.4 ⁇ M). C5 is glycosylated, with about 1.5-3 percent of its mass attributed to carbohydrate. Mature C5 is a heterodimer of a 999 amino acid 115 kDa alpha chain that is disulfide linked to a 656 amino acid 75 kDa beta chain. C5 is synthesized as a single chain precursor protein product of a single copy gene (Haviland et al., 1991). The cDNA sequence of the transcript of this gene predicts a secreted pro-C5 precursor of 1659 amino acids along with an 18 amino acid leader sequence.
  • the pro-C5 precursor is cleaved after amino acid 655 and 659, to yield the beta chain as an amino terminal fragment (amino acid residues +1 to 655) and the alpha chain as a carboxyl terminal fragment (amino acid residues 660 to 1658), with four amino acids deleted between the two.
  • C5a is cleaved from the alpha chain of C5 by either alternative or classical C5 convertase as an amino terminal fragment comprising the first 74 amino acids of the alpha chain (i.e., amino acid residues 660-733). Approximately 20 percent of the II kDa mass of C5a is attributed to carbohydrate. The cleavage site for convertase action is at or immediately adjacent to amino acid residue 733. A compound that would bind at or adjacent to this cleavage site would have the potential to block access of the C5 convertase enzymes to the cleavage site and thereby act as a complement inhibitor.
  • C5 can also be activated by means other than C5 convertase activity. Limited trypsin digestion (Minta and Man, 1977; Wetsel and Kolb, 1982) and acid treatment (Yamamoto and Gewurz, 1978; Vogt et al., 1989) can also cleave C5 and produce active C5b.
  • C5a is another anaphylatoxin.
  • C5b combines with C6, C7, and C8 to form the C5b-8 complex at the surface of the target cell.
  • the membrane attack complex (MAC, C5b-9, terminal complement complex-TCC) is formed.
  • MAC membrane attack complex
  • C5b-9 terminal complement complex-TCC
  • MAC pores mediate rapid osmotic lysis of the target cells.
  • non-lytic concentrations of MACs can produce other effects.
  • membrane insertion of small numbers of the C5b-9 complexes into endothelial cells and platelets can cause deleterious cell activation. In some cases activation may precede cell lysis.
  • C3a and C5a are anaphylatoxins. These activated complement components can trigger mast cell degranulation, which releases histamine and other mediators of inflammation, resulting in smooth muscle contraction, increased vascular permeability, leukocyte activation, and other inflammatory phenomena including cellular proliferation resulting in hypercellularity.
  • C5a also functions as a chemotactic peptide that serves to attract pro-inflammatory granulocytes to the site of complement activation.
  • anti-C5 mAb has previously been reported in several experimental models including myocardial reperfusion (Vakeva et al., 1998), systemic lupus erythematosus (Wang et al., 1996) and rheumatoid arthritis (Wang et al., 1995); as well as in human clinical trials (Kirschfink, 2001) of autoimmune disease, cardiopulmonary bypass and acute myocardial infarction.
  • MMSE Mini-Mental State Examination
  • EORTC European Organization for Research and Treatment of Cancer
  • FACIT questionnaires and subscales including fatigue and anemia the Likert Scale, and Borg Scale (Tombaugh, et al., J. Am. Geriatr. Soc. 40:922, 1992; Cummings, JAMA. 269(18):2420, 1993; Crum, et al., JAMA. 269(18):2386, 1993; Folstein, et. al., J. Psychiat. Res.
  • improvement in quality of life due to administration of a compound which inhibits complement or inhibits formation of C5b-9 is measured by the Functional Assessment of Chronic Illness Therapy (FACIT) Measurement System.
  • FACIT Functional Assessment of Chronic Illness Therapy
  • improvement in quality of life is measured by: a) full scales; b) stand-alone subscales; and c) symptom indices.
  • improvement in quality of life due to administration of a compound which inhibits complement or inhibits formation of C5b-9 is measured by a European Organization for Research and Treatment of Cancer (EORTC) Quality of Life Questionnaire.
  • EORTC European Organization for Research and Treatment of Cancer
  • the EORTC questionnaire is the QLQ-C30.
  • HALE health-adjusted life expectancy
  • QALY quality-adjusted life years
  • Quality-adjusted life years is a health index that weighs each year of life on a scale from 1 to 0 (Weinstein M C and Stason W B, N Engl J Med, 296:716-721 (1977)). Perfect health is rated as 1, death is rated as 0, and disability and pain are rated based on severity.
  • QALY is determined by multiplying the number of years at each health status.
  • improvement in quality of life is measured by the following instruments: Years of potential life lost, Disability-free life expectancy, Health-adjusted life year, Quality adjusted life year, Healthy years equivalents, Healthy days gained, Episode-free day, Q-TWiST, Health Utilities Index, and Years of healthy life. These measurements account for both changes in mortality as well as changes in morbidity and disability. Any of these measurements may be used to assess the change in quality of life due to administration of a compound which inhibits complement or inhibits formation of C5b-9.
  • the disclosed methods improve the quality of life of a patient for at least one day, at least one week, at least two weeks, at least three weeks, at least one month, at least two months, at least three months, at least 6 months, at least one year, at least 18 months, at least two years, at least 30 months, or at least three years, or the duration of treatment.
  • the symptoms used to measure quality of life are scaled for intensity. In certain embodiments, the symptoms are scaled for frequency. In certain embodiments, the symptoms are scaled for intensity and frequency.
  • the application provides a method for prolonging the health-adjusted life expectancy of a subject comprising administering to the subject a compound which inhibits complement or inhibits formation of C5b-9.
  • the above measurements account for both changes in mortality as well as changes in morbidity and disability. Any of these measurements may be used to assess the change in quality-adjusted life expectancy due to administration of a compound which inhibits complement or inhibits formation of C5b-9.
  • the disclosed methods prolong the health-adjusted life expectancy in a subject by at least one day, at least one week, at least two weeks, at least three weeks, at least one month, at least two months, at least three months, at least 6 months, at least one year, at least 18 months, at least two years, at least 30 months, or at least three years as measured by the health-adjusted life expectancy (HALE) index as described in Wilkins et al. Am J Public Health, 73:1073-1080 (1983).
  • HALE health-adjusted life expectancy
  • Health-adjusted life expectancy is an average of the quality-adjusted life years (QALY) for a given population and can be used to evaluate the therapeutic value of a medical intervention.
  • Quality-adjusted life years is a health index that weighs each year of life on a scale from 1 to 0 (Weinstein et al., N Engl J Med, 296:716-721 (1977)). Perfect health is rated as 1, death is rated as 0, and disability and pain are rated based on severity. QALY is determined by multiplying the number of years at each health status.
  • a complement inhibitor may be a small molecule (up to 6,000 Da in molecular weight), a nucleic acid or nucleic acid analog, a peptidomimetic, or a macromolecule that is not a nucleic acid, a serine protease inhibitor, or a protein.
  • RNA aptamers including ARC187 (which is commercially available from Archemix Corp., Cambridge, Mass.), L-RNA aptamers, Spiegelmers, antisense compounds, molecules which may be utilized in RNA interference (RNAi) such as double stranded RNA including small interfering RNA (siRNA), locked nucleic acid (LNA) inhibitors, peptide nucleic acid (PNA) inhibitors.
  • RNAi RNA interference
  • siRNA small interfering RNA
  • LNA locked nucleic acid
  • PNA peptide nucleic acid
  • a complement inhibitor may be a protein or protein fragment. Proteins are known which inhibit the complement cascade, including CD59, CD55, CD46 and other inhibitors of C8 and C9 (see, e.g., U.S. Pat. No. 6,100,443). Proteins known as complement receptors and which bind complement are also known (see, Published PCT Patent Application WO 92/10205 and U.S. Pat. No. 6,057,131). Use of soluble forms of complement receptors, e.g., soluble CR1, can inhibit the consequences of complement activation such as neutrophil oxidative burst, complement mediated neural injury, and C3a and C5a production.
  • complement activation such as neutrophil oxidative burst, complement mediated neural injury, and C3a and C5a production.
  • a complement inhibitor may be naturally occurring or soluble forms of complement inhibitory compounds such as CR1, LEX-CR1, MCP, DAF, CD59, Factor H, cobra venom factor, FUT-175, complestatin, and K76 COOH.
  • complement inhibitory compounds such as CR1, LEX-CR1, MCP, DAF, CD59, Factor H, cobra venom factor, FUT-175, complestatin, and K76 COOH.
  • a complement inhibitor may be an antibody capable of inhibiting complement, such as an antibody that can block the formation of MAC.
  • an antibody complement inhibitor may include an antibody that binds C5.
  • Such anti-C5 antibodies may directly interact with C5 and/or C5b, so as to inhibit the formation of and/or physiologic function of C5b.
  • Suitable anti-C5 antibodies are known to those of skill in the art. Antibodies can be made to individual components of activated complement, e.g., antibodies to C7, C9, etc. (see, e.g., U.S. Pat. No. 6,534,058; published U.S. patent application US 2003/0129187; and U.S. Pat. No. 5,660,825). U.S. Pat. No. 6,355,245 teaches an antibody which binds to C5 and inhibits cleavage into C5a and C5b thereby decreasing the formation not only of C5a but also the downstream complement components.
  • the concentration and/or physiologic activity of C5a and C5b in a body fluid can be measured by methods well known in the art.
  • C5a such methods include chemotaxis assays, RIAs, or ELISAs (see, for example, Ward and Zvaifler, J Clin Invest. 1971 March; 50(3):606-16; Wurzner, et al., Complement Inflamm. 8:328-340, 1991).
  • C5b hemolytic assays or assays for soluble C5b-9 as discussed herein can be used. Other assays known in the art can also be used.
  • candidate antibodies capable of inhibiting complement such as anti-C5 antibodies, now known or subsequently identified, can be screened in order to 1) identify compounds that are useful in the practice of the application and 2) determine the appropriate dosage levels of such compounds.
  • An antibody capable of inhibiting complement such as an antibody that binds C5 affecting C5b is preferably used at concentrations providing substantial reduction (i.e., reduction by at least about 25% as compared to that in the absence of the antibody that binds C5) in the C5b levels present in at least one blood-derived fluid of the patient following activation of complement within the fluid.
  • concentrations can be conveniently determined by measuring the cell-lysing ability (e.g., hemolytic activity) of complement present in the fluid or the levels of soluble C5b-9 present in the fluid.
  • a specific concentration for an antibody that affects C5b is one that results in a substantial reduction (i.e., a reduction by at least about 25%) in the cell-lysing ability of the complement present in at least one of the patient's blood-derived fluids.
  • Reductions of the cell-lysing ability of complement present in the patient's body fluids can be measured by methods well known in the art such as, for example, by a conventional hemolytic assay such as the hemolysis assay described by Kabat and Mayer (eds), “Experimental Immunochemistry, 2d Edition”, 135-240, Springfield, Ill., CC Thomas (1961), pages 135-139, or a conventional variation of that assay such as the chicken erythrocyte hemolysis method described below.
  • Specific antibodies capable of inhibiting complement are relatively specific and do not block the functions of early complement components.
  • specific agents will not substantially impair the opsonization functions associated with complement component C3b, which functions provide a means for clearance of foreign particles and substances from the body.
  • C3b is generated by the cleavage of C3, which is carried out by classical and/or alternative C3 convertases and results in the generation of both C3a and C3b. Therefore, in order not to impair the opsonization functions associated with C3b, specific antibodies capable of inhibiting complement such as an antibody that binds C5 do not substantially interfere with the cleavage of complement component C3 in a body fluid of the patient (e.g., serum) into C3a and C3b. Such interference with the cleavage of C3 can be detected by measuring body fluid levels of C3a and/or C3b, which are produced in equimolar ratios by the actions of the C3 convertases.
  • a body fluid of the patient e.g., serum
  • Such measurements are informative because C3a and C3b levels will be reduced (compared to a matched sample without the antibody capable of inhibiting complement such as an antibody that binds C5) if cleavage is interfered with by an antibody capable of inhibiting complement such as an antibody that binds C5.
  • C3a levels in a body fluid can be measured by methods well known in the art such as, for example, by using a commercially available C3a EIA kit, e.g., that sold by Quidel Corporation, San Diego, Calif., according to the manufacturer's specifications.
  • C3a EIA kit e.g., that sold by Quidel Corporation, San Diego, Calif.
  • Particularly specific antibodies capable of inhibiting complement such as an antibody that binds C5 produce essentially no reduction in body fluid C3a levels following complement activation when tested in such assays.
  • Certain antibodies of the disclosure will prevent the cleavage of C5 to form C5a and C5b, thus preventing the generation of the anaphylatoxic activity associated with C5a and preventing the assembly of the membrane attack complex associated with C5b. As discussed above, in a particular embodiment, these anti-C5 antibodies will not impair the opsonization function associated with the action of C3b.
  • a preferred method of inhibiting complement activity is to use a monoclonal antibody which binds to complement C5 and inhibits cleavage. This decreases the formation of both C5a and C5b while at the same time allowing the formation of C3a and C3b which are beneficial to the recipient.
  • Such antibodies which are specific to human complement are known (U.S. Pat. No. 6,355,245). These antibodies disclosed in U.S. Pat. No. 6,355,245 include a preferred whole antibody (now named eculizumab).
  • a similar antibody against mouse C5 is called BB5.1 (Frei et al., Mol. Cell. Probes. 1:141-149 (1987)).
  • Antibodies to inhibit complement activity need not be monoclonal antibodies.
  • antibodies can be, e.g., polyclonal antibodies. They may additionally be antibody fragments.
  • An antibody fragment includes, but is not limited to, an Fab, F(ab′), F(ab′) 2 , single-chain antibody, and F ⁇ .
  • antibodies can be humanized (Jones et al., Nature 321:522-S (1986)), chimerized, or deimmunized.
  • the antibodies to be used in the present disclosure may be any of these. It is preferable to use humanized antibodies.
  • a therapeutic agent of the disclosure comprises an antibody or antibody fragment.
  • Antibodies and fragments thereof may be made by any conventional method, such as those methods described herein. Antibodies are found in multiple forms, e.g., IgA, IgG, IgM, etc. Additionally, antibodies can be engineered in numerous ways. They can be made as single-chain antibodies (including small modular immunopharmaceuticals or SMIPsTM), Fab and F(ab′) 2 fragments, etc. Antibodies can be humanized, chimerized, deimmunized, or fully human. Numerous publications set forth the many types of antibodies and the methods of engineering such antibodies. For example, see U.S. Pat. Nos. 6,355,245; 6,180,370; 5,693,762; 6,407,213, 6,548,640; 5,565,332; 5,225,539; 6,103,889; and 5,260,203.
  • This invention provides fragments of anti-C5 antibodies, which may comprise a portion of an intact antibody, preferably the antigen-binding or variable region of the intact antibody.
  • antibody fragments include Fab, Fab′, F(ab′) 2 , and Fv fragments; diabodies; linear antibodies (Zapata et al., Protein Eng. 8:1057-1062 (1995)); single-chain antibody molecules; and multispecific antibodies formed from antibody fragments.
  • Papain digestion of antibodies produces two identical antigen-binding fragments, called “Fab” fragments, each with a single antigen-binding site, and a residual “Fc” fragment, whose name reflects its ability to crystallize readily.
  • Pepsin treatment of an antibody yields an F(ab′) 2 fragment that has two antigen-combining sites and is still capable of cross-linking antigen.
  • “Fv” refers to the minimum antibody fragment that contains a complete antigen-recognition and -binding site. This region consists of a dimer of one heavy- and one light-chain variable domain in tight, non-covalent association. It is in this configuration that the three CDRs of each variable domain interact to define an antigen-binding site on the surface of the V H -V L dimer Collectively, the six CDRs confer antigen-binding specificity to the antibody. However, even a single variable domain (or half of an Fv comprising only three CDRs specific for an antigen) has the ability to recognize and bind antigen, although likely at a lower affinity than the entire binding site.
  • the Fab fragment also contains the constant domain of the light chain and the first constant domain (CH1) of the heavy chain.
  • Fab′ fragments differ from Fab fragments by the addition of a few residues at the carboxy terminus of the heavy chain CH1 domain including one or more cysteines from the antibody hinge region.
  • Fab′-SH is the designation herein for Fab′ in which the cysteine residue(s) of the constant domains bear a free thiol group.
  • F(ab′) 2 antibody fragments originally were produced as pairs of Fab′ fragments that have hinge cysteines between them. Other chemical couplings of antibody fragments are also known.
  • Single-chain Fv or “scFv” antibody fragments comprise the V H and V L domains of an antibody, wherein these domains are present in a single polypeptide chain.
  • the Fv polypeptide further comprises a polypeptide linker between the V H and V L domains that enables the scFv to form the desired structure for antigen binding.
  • SMIPs are a class of single-chain peptide engineered to include a target binding region, effector domain (CH2 and CH3 domains). See, e.g., U.S. Patent Application Publication No. 20050238646.
  • the target binding region may be derived from the variable region or CDRs of an antibody, e.g., an antibody that binds C5 of the application.
  • the target binding region is derived from a protein that binds C5.
  • diabodies refers to small antibody fragments with two antigen-binding sites, which fragments comprise a heavy-chain variable domain (V H ) connected to a light-chain variable domain (V L ) in the same polypeptide chain (V H -V L ).
  • V H heavy-chain variable domain
  • V L light-chain variable domain
  • the domains are forced to pair with the complementary domains of another chain and create two antigen-binding sites.
  • Diabodies are described more fully in, for example, EP 404,097; WO 93/11161; and Hollinger et al., Proc. Natl. Acad. Sci. USA, 90: 6444-6448 (1993).
  • the Fc portions of antibodies are recognized by specialized receptors expressed by immune effector cells.
  • the Fc portions of IgG1 and IgG3 antibodies are recognized by Fc receptors present on the surface of phagocytic cells such as macrophages and neutrophils, which can thereby bind and engulf the molecules or pathogens coated with antibodies of these isotypes (C. A. Janeway et al., Immunobiology 5th edition, page 147, Garland Publishing (New York, 2001)).
  • a monoclonal antibody can be obtained from a population of substantially homogeneous antibodies, i.e., the individual antibodies comprising the population are identical except for possible naturally-occurring mutations that may be present in minor amounts. Monoclonal antibodies are highly specific, being directed against a single antigenic site. Furthermore, in contrast to conventional (polyclonal) antibody preparations that typically include different antibodies directed against different determinants (epitopes), each monoclonal antibody is directed against a single determinant on the antigen. In addition to their specificity, the monoclonal antibodies are advantageous in that they are often synthesized by the hybridoma culture, uncontaminated by other immunoglobulins.
  • Monoclonal antibodies may also be produced in transfected cells, such as CHO cells and NS0 cells.
  • the modifier “monoclonal” indicates the character of the antibody as being obtained from a substantially homogeneous population of antibodies and does not require production of the antibody by any particular method.
  • the monoclonal antibodies to be used in accordance with the present disclosure may be made by the hybridoma method first described by Kohler et al., Nature 256:495-497 (1975), or may be made by recombinant DNA methods (see, e.g., U.S. Pat. Nos. 4,816,567 and 6,331,415).
  • the “monoclonal antibodies” may also be isolated from phage antibody libraries using the techniques described in Clackson et al., Nature 352:624-628 (1991) and Marks et al., J. Mol. Biol. 222:581-597 (1991), for example.
  • oligoclonal antibodies refers to a predetermined mixture of distinct monoclonal antibodies. See, e.g., PCT publication WO 95/20401; U.S. Pat. Nos. 5,789,208 and 6,335,163.
  • oligoclonal antibodies consisting of a predetermined mixture of antibodies against one or more epitopes are generated in a single cell.
  • oligoclonal antibodies comprise a plurality of heavy chains capable of pairing with a common light chain to generate antibodies with multiple specificities (e.g., PCT publication WO 04/009618).
  • Oligoclonal antibodies are particularly useful when it is desired to target multiple epitopes on a single target molecule (e.g., C5).
  • a single target molecule e.g., C5
  • those skilled in the art can generate or select antibodies or mixtures of antibodies that are applicable for an intended purpose and desired need.
  • one or more of the CDRs are derived from an anti-human C5 antibody.
  • all of the CDRs are derived from an anti-human C5 antibody.
  • the CDRs from more than one anti-human C5 antibody are mixed and matched in a chimeric antibody.
  • a chimeric antibody may comprise a CDR1 from the light chain of a first anti-human C5 antibody combined with CDR2 and CDR3 from the light chain of a second anti-human C5 antibody, and the CDRs from the heavy chain may be derived from a third anti-human C5 antibody.
  • the framework regions may be derived from one of the same anti-human C5 antibodies, from one or more different antibodies, such as a human antibody, or from a humanized antibody. Human or humanized antibodies are specific for administration to human patients.
  • single chain antibodies, and chimeric, humanized or primatized (CDR-grafted) antibodies, as well as chimeric or CDR-grafted single chain antibodies, comprising portions derived from different species, are also encompassed by the present disclosure as antigen-binding fragments of an antibody.
  • the various portions of these antibodies can be joined together chemically by conventional techniques, or can be prepared as a contiguous protein using genetic engineering techniques.
  • nucleic acids encoding a chimeric or humanized chain can be expressed to produce a contiguous protein. See, e.g., U.S. Pat. Nos. 4,816,567 and 6,331,415; U.S. Pat. No. 4,816,397; European Patent No.
  • functional fragments of antibodies including fragments of chimeric, humanized, primatized or single chain antibodies, can also be produced.
  • Functional fragments of the subject antibodies retain at least one binding function and/or modulation function of the full-length antibody from which they are derived.
  • Preferred functional fragments retain an antigen-binding function of a corresponding full-length antibody (such as for example, ability of antibody that binds C5 to bind C5).
  • Methods of the application may be used to treat paroxysmal nocturnal hemoglobinuria associated symptoms. Methods of the application may be used to treat anemia associated symptoms. Treatment of paroxysmal nocturnal hemoglobinuria and/or anemia may be administered by standard means. Treatments of the application may be used in combination with other treatments of the application or known treatments for paroxysmal nocturnal hemoglobinuria and/or anemia. Treatments of the application may be co-administered with other treatments that treat symptoms of paroxysmal nocturnal hemoglobinuria and/or anemia.
  • the antibodies can be administered in a variety of unit dosage forms.
  • the dose will vary according to the particular antibody. For example, different antibodies may have different masses and/or affinities, and thus require different dosage levels.
  • Antibodies prepared as Fab fragments will also require differing dosages than the equivalent intact immunoglobulins, as they are of considerably smaller mass than intact immunoglobulins, and thus require lower dosages to reach the same molar levels in the patient's blood.
  • Dosage levels of the antibodies for human subjects are generally between about 1 mg per kg and about 100 mg per kg per patient per treatment, and preferably between about 5 mg per kg and about 50 mg per kg per patient per treatment.
  • the antibody concentrations are preferably in the range from about 25 ⁇ g/mL to about 500 ⁇ g/mL. However, greater amounts may be required for extreme cases and smaller amounts may be sufficient for milder cases.
  • the pharmaceutical composition is in a single unit dosage form.
  • the single unit dosage form is a 300 mg unit dosage form.
  • the pharmaceutical composition is lyophilized.
  • the pharmaceutical composition is a sterile solution.
  • the pharmaceutical composition is a preservative free formulation.
  • the pharmaceutical composition comprises a 300 mg single-use formulation of 30 ml of a 10 mg/ml sterile, preservative free solution.
  • the antibody is administered according to the following protocol: 600 mg via 25 to 45 minute IV infusion every 7 ⁇ 2 days for the first 4 weeks, followed by 900 mg for the fifth dose 7 ⁇ 2 days later, then 900 mg every 14 ⁇ 2 days thereafter. Antibody is administered via IV infusion over 25 to 45 minute.
  • Administration of the anti-C5 antibodies will generally be performed by an intravascular route, e.g., via intravenous infusion by injection. Other routes of administration may be used if desired but an intravenous route will be the most preferable.
  • Formulations suitable for injection are found in Remington's Pharmaceutical Sciences, Mack Publishing Company, Philadelphia, Pa., 17th ed. (1985). Such formulations must be sterile and non-pyrogenic, and generally will include a pharmaceutically effective carrier, such as saline, buffered (e.g., phosphate buffered) saline, Hank's solution, Ringer's solution, dextrose/saline, glucose solutions, and the like.
  • the formulations may contain pharmaceutically acceptable auxiliary substances as required, such as, tonicity adjusting agents, wetting agents, bactericidal agents, preservatives, stabilizers, and the like.
  • complement inhibitors such as eculizumab may be administered via IV infusion and diluted to a final concentration of 5 mg/ml prior to administration.
  • Administration of the antibodies capable of inhibiting complement such as an antibody that binds C5 will generally be performed by a parenteral route, typically via injection such as intra-articular or intravascular injection (e.g., intravenous infusion) or intramuscular injection. Other routes of administration, e.g., oral (p.o.), may be used if desired and practicable for the particular antibody capable of inhibiting complement to be administered.
  • Antibodies capable of inhibiting complement such as an antibody that binds C5 can also be administered in a variety of unit dosage forms and their dosages will also vary with the size, potency, and in vivo half-life of the particular antibody capable of inhibiting complement being administered. Doses of antibodies capable of inhibiting complement such as an antibody that binds C5 will also vary depending on the manner of administration, the particular symptoms of the patient being treated, the overall health, condition, size, and age of the patient, and the judgment of the prescribing physician.
  • a typical therapeutic treatment includes a series of doses, which will usually be administered concurrently with the monitoring of clinical endpoints with the dosage levels adjusted as needed to achieve the desired clinical outcome.
  • treatment is administered in multiple dosages over at least a week.
  • treatment is administered in multiple dosages over at least a month.
  • treatment is administered in multiple dosages over at least a year.
  • treatment is administered in multiple dosages over the remainder of the patient's life.
  • the frequency of administration may also be adjusted according to various parameters. These include the clinical response, the plasma half-life of the therapeutic of the disclosure, and the levels of the antibody in a body fluid, such as, blood, plasma, serum, or synovial fluid. To guide adjustment of the frequency of administration, levels of the therapeutic of the disclosure in the body fluid may be monitored during the course of treatment.
  • the frequency of administration may be adjusted according to an assay measuring cell-lysing ability of complement present in one or more of the patient's body fluids.
  • the cell-lysing ability can be measured as percent hemolysis in hemolytic assays of the types described herein.
  • a 10% or 25% or 50% reduction in the cell-lysing ability of complement present in a body fluid after treatment with the antibody capable of inhibiting complement used in the practice of the application means that the percent hemolysis after treatment is 90, 75, or 50 percent, respectively, of the percent hemolysis before treatment.
  • a large initial dose is specific, i.e., a single initial dose sufficient to yield a substantial reduction, and more preferably an at least about 50% reduction, in the hemolytic activity of the patient's serum.
  • a large initial dose is preferably followed by regularly repeated administration of tapered doses as needed to maintain substantial reductions of serum hemolytic titer.
  • the initial dose is given by both local and systemic routes, followed by repeated systemic administration of tapered doses as described above.
  • the liquid formulations of the application are substantially free of surfactant and/or inorganic salts.
  • the liquid formulations have a pH ranging from about 5.0 to about 7.0.
  • the liquid formulations comprise histidine at a concentration ranging from about 1 mM to about 100 mM.
  • the liquid formulations comprise histidine at a concentration ranging from 1 mM to 100 mM.
  • liquid formulations may further comprise one or more excipients such as a saccharide, an amino acid (e.g., arginine, lysine, and methionine) and a polyol.
  • excipients such as a saccharide, an amino acid (e.g., arginine, lysine, and methionine) and a polyol.
  • wetting agents such as sodium lauryl sulfate and magnesium stearate, as well as coloring agents, release agents, coating agents, sweetening, flavoring and perfuming agents, preservatives and antioxidants can also be present in the pharmaceutical compositions of the application.
  • formulations of the subject antibodies are pyrogen-free formulations which are substantially free of endotoxins and/or related pyrogenic substances.
  • Endotoxins include toxins that are confined inside microorganisms and are released when the microorganisms are broken down or die.
  • Pyrogenic substances also include fever-inducing, thermostable substances (glycoproteins) from the outer membrane of bacteria and other microorganisms. Both of these substances can cause fever, hypotension and shock if administered to humans. Due to the potential harmful effects, it is advantageous to remove even low amounts of endotoxins from intravenously administered pharmaceutical drug solutions.
  • FDA Food & Drug Administration
  • EU endotoxin units
  • Formulations of the subject antibodies include those suitable for oral, dietary, topical, parenteral (e.g., intravenous, intraarterial, intramuscular, subcutaneous injection), ophthalmologic (e.g., topical or intraocular), inhalation (e.g., intrabronchial, intranasal or oral inhalation, intranasal drops), rectal, and/or intravaginal administration.
  • parenteral e.g., intravenous, intraarterial, intramuscular, subcutaneous injection
  • ophthalmologic e.g., topical or intraocular
  • inhalation e.g., intrabronchial, intranasal or oral inhalation, intranasal drops
  • rectal e.g., rectal, and/or intravaginal administration.
  • Other suitable methods of administration can also include rechargeable or biodegradable devices and controlled release polymeric devices.
  • Stents in particular, may be coated with a controlled release polymer mixed with an agent
  • the amount of the formulation which will be therapeutically effective can be determined by standard clinical techniques.
  • in vitro assays may optionally be employed to help identify optimal dosage ranges.
  • the precise dose to be employed in the formulation will also depend on the route of administration, and the seriousness of the disease or disorder, and should be decided according to the judgment of the practitioner and each patient's circumstances. Effective doses may be extrapolated from dose-response curves derived from in vitro or animal model test systems.
  • the dosage of the compositions to be administered can be determined by the skilled artisan without undue experimentation in conjunction with standard dose-response studies.
  • anti-C5 antibodies can be administered in a variety of unit dosage forms.
  • the dose will vary according to the particular antibody. For example, different antibodies may have different masses and/or affinities, and thus require different dosage levels.
  • Antibodies prepared as Fab′ fragments or single chain antibodies will also require differing dosages than the equivalent native immunoglobulins, as they are of considerably smaller mass than native immunoglobulins, and thus require lower dosages to reach the same molar levels in the patient's blood.
  • therapeutics of the disclosure can also be administered in a variety of unit dosage forms and their dosages will also vary with the size, potency, and in vivo half-life of the particular therapeutic being administered.
  • Doses of therapeutics of the disclosure will also vary depending on the manner of administration, the particular symptoms of the patient being treated, the overall health, condition, size, and age of the patient, and the judgment of the prescribing physician.
  • the formulations of the application can be distributed as articles of manufacture comprising packaging material and a pharmaceutical agent which comprises the antibody capable of inhibiting complement and a pharmaceutically acceptable carrier as appropriate to the mode of administration.
  • the packaging material may include a label which indicates that the formulation is for use in the treatment of hemolytic diseases such as PNH.
  • hemolytic diseases such as PNH.
  • antibodies are preferred, especially anti-C5 antibodies which have already been shown to be safe and effective at decreasing the accumulation of downstream complement components in persons, the use of other complement inhibitors is also contemplated by this disclosure.
  • the pharmaceutical formulations and uses of the disclosure may be combined with any known complement inhibitors or hemolytic diseases treatments known in the art.
  • kits comprising a pharmaceutical composition of the application.
  • the kit further comprises at least one component of a closed sterile system.
  • Components of the closed sterile system include, but are not limited to, needles, syringes, catheter based syringes, needle based injection devices, needle-less injection devices, filters, tubing, valves and cannulas.
  • the kit comprise components for the removal of a preservative from the composition. Such components include filters, syringes, vials, containers, tubing, etc.
  • the TRIUMPH trial consisted of a 2-week screening period, an observation period of up to 3 months duration, and a 2-week treatment period.
  • patients were evaluated with respect to inclusion and exclusion criteria. Men and women, 18 years or older, diagnosed as having PNH with a type III erythrocyte population of ⁇ 10%, and who had received at least 4 transfusions in the previous 12 months were eligible. Concomitant administration of erythropoietin, immunosuppressants, corticosteroids, coumadin, low molecular weight heparin, iron supplements, and folic acid were not reasons for exclusion, provided the doses were steady prior to the first visit and throughout the duration of the study. Because of the increased frequency of neisserial infections in individuals genetically deficient in terminal complement proteins, all patients were vaccinated against Neisseria meningitides. Patients were to avoid conception.
  • the protocol was approved by an Investigational Review Board at each clinical site and written informed consent was obtained from all patients enrolled. Patients transfused with a mean pre-transfusion hemoglobin level >10.5 g/dL over the previous 12 months, and those who showed evidence of having a suppressed immune response, complement deficiency, or active bacterial infection, including any history of meningococcal disease, were excluded from the study. Patients were also not eligible if they had previously received a bone marrow transplant or if they had participated in another trial or received another investigational drug within 30 days of the first visit.
  • a platelet count ⁇ 100,000/mL and a LDH level ⁇ 1.5 times the upper limit of the normal range were also required either at screening or during the observation period for eligibility.
  • eculizumab placebo or eculizumab (SolirisTM, Alexion Pharmaceuticals, Inc.) within 10 days of the qualifying transfusion.
  • Study medication was dosed in a blinded fashion as follows: 600 mg eculizumab for patients randomly assigned to active drug, or placebo for those patients randomly assigned to placebo, respectively via IV infusion every 7 ⁇ 1 days for 4 doses; followed by 900 mg eculizumab, or placebo, respectively, via IV infusion 7 ⁇ 1 day later; followed by a maintenance dose of 900 mg eculizumab, or placebo, respectively, via IV infusion every 14 ⁇ 2 days for a total of 26 weeks of treatment.
  • Pre-specified secondary endpoints included transfusion avoidance, hemolysis as measured by LDH area under the curve from baseline to 26 weeks, and QoL changes as measured from baseline to 26 weeks using the Functional Assessment of Chronic Illness Therapy-Fatigue (FACIT-Fatigue) instrument. 13 Pre-specified exploratory analyses included assessment of the EORTC QLQ-C30 instrument, 14 the change in LDH from baseline through week 26, and thrombosis. Other pre-specified measurements included pharmacokinetics, pharmacodynamics, and immunogenicity of eculizumab. Time to first transfusion during the 26-week treatment phase and the proportion of PNH type III blood cells were also assessed.
  • FACIT-Fatigue Functional Assessment of Chronic Illness Therapy-Fatigue
  • Treatment-emergent adverse events e.g., serum chemical analyses and complete blood counts
  • electrocardiogram data e.g., electrocardiogram data
  • vital signs were assessed.
  • Adverse events were defined using the MedDRA preferred terms and tabulated as incidence rates per treatment group.
  • Stable usage of concomitant medications at baseline in the eculizuumab- and placebo-treated groups included the following: erythropoietin, 3 patients and 0 patients; cyclosporine, 1 and 1; anticoagulants (coumarins or heparins) 21 and 11; and steroids (glucocorticoids or androgenic steroids), 12 and 12, respectively.
  • the mean LDH level decreased from 2199.7 ⁇ 157.7 IU/L at baseline to 327.3 ⁇ 67.6 IU/L by 26 weeks in eculizumab-treated patients while levels in placebo-treated patients remained consistently elevated with values of 2259.0 ⁇ 158.5 IU/L at baseline and 2418.9 ⁇ 140.3 IU/L at 26 weeks (P ⁇ 0.001, for eculizumab versus placebo).
  • Haptoglobin levels were statistically significantly increased in eculizumab- as compared to placebo-treated patients but mean levels of haptoglobin were still below normal levels in eculizumab-treated patients (data not shown).
  • FIG. 1A shows the degree of intravascular hemolysis in PNH patients, demonstrated by mean lactate dehydrogenase (LDH) levels ( ⁇ standard error) from baseline (study week 0) to week 26 for both eculizumab- and placebo-treated patient populations. Screening occurred up to 3 months prior to study week 0. The upper limit of the normal range (103-223 IU/L) for LDH is indicated by a dashed line. LDH was reduced to a mean level just above the upper limit of normal at week 26 for eculizumab-treated patients; 15 of 41 patients who completed the study demonstrated LDH levels within the normal range. All placebo-treated patients remained at least 5 times above the upper limit of normal at week 26.
  • LDH lactate dehydrogenase
  • FIG. 1B shows the mean proportion ( ⁇ standard error) of PNH type III erythrocytes assessed for placebo- and eculizumab-treated patients.
  • the screening visit occurred up to 3 months prior to study week 0.
  • the P value is based on a mixed model analysis from baseline through week 26.
  • FIG. 1B A corollary to the reduction in intravascular hemolysis during eculizumab treatment was an observed increase in the PNH type III erythrocyte population ( FIG. 1B ).
  • the mean proportions of type III erythrocytes increased from 28.1 ⁇ 2.0% at baseline to 56.9 ⁇ 3.6% by week 26 for eculizumab-treated patients while proportions in the placebo group remained constant with mean values of 35.7 ⁇ 2.8% before treatment to 35.5 ⁇ 2.8% at 26 weeks (P ⁇ 0.001, for eculizumab versus placebo).
  • the proportions of PNH type III granulocytes and monocytes did not change significantly between the treatment groups during the treatment period and were greater than 90% at week 26.
  • the co-primary efficacy endpoints in the TRIUMPH trial were stabilization of hemoglobin levels and reduction in PRBC units transfused.
  • 48.8% of eculizumab-treated patients had maintained levels of hemoglobin above the pre-specified set-point (median set-point value of 7.7 g/dL for both treatment groups) in the absence of transfusions, whereas stabilization of hemoglobin did not occur in any of the patients in the placebo group (P ⁇ 0.001; Table 1).
  • the median of PRBC units transfused per patient was 0 in the eculizumab group and 10.0 in the placebo cohort (P ⁇ 0.001), while the mean of PRBC units transfused was 3.0 and 11.0 in the eculizumab and placebo cohorts, respectively.
  • the median of PRBC units transfused per patient was 9.0 in the eculizumab cohort and 8.5 in placebo patients while the mean of PRBC units transfused was 9.6 ⁇ 0.6 and 9.7 ⁇ 0.7, respectively.
  • the median time to first transfusion was not reached during the study period in eculizumab-treated patients (it was greater than 26 weeks) while the placebo group reached the median time to first transfusion in only 4 weeks (P ⁇ 0.001; FIG. 2 ).
  • Transfusion avoidance was achieved in 51.2% and 0% of the eculizumab and placebo cohorts, respectively (P ⁇ 0.001).
  • the total PRBC units transfused were 131 in eculizumab-treated patients versus 482 in the placebo group (Table 1).
  • total PRBC units transfused in the eculizumab- and placebo-cohorts were 413 and 417, respectively.
  • SAEs Serious adverse events
  • Lactate dehydrogenase a biochemical marker of hemolysis in PNH, 9 was immediately and consistently decreased in all eculizumab-treated patients, while patients in the placebo cohort continued to hemolyze with levels of LDH exceeding 5 times the upper limit of the normal range in all patients at the study end. Levels of LDH were reduced into the normal range in approximately one-third of eculizumab-treated patients, while the remainder stabilized at a level just above the upper limit of normal suggesting residual low level hemolysis in some patients. Levels of haptoglobin, a more sensitive marker of the presence of cell free hemoglobin in the circulation, were undetectable in most patients.
  • Patients with PNH generally experience markedly impaired quality of life characterized by fatigue, anemia, thrombosis, and pulmonary hypertension as well as smooth muscle dystonia including abdominal pain, dysphagia, and erectile dysfunction. 9,10,18 These symptoms have been attributed to both excessive intravascular hemolysis and downstream scavenging of nitric oxide by cell free hemoglobin in plasma.
  • the reduction of intravascular hemolysis in eculizumab-treated patients in the current study was associated with significant improvements in the fatigue component of quality of life relative to placebo-treated patients as assessed via the FACIT-Fatigue instrument. Further, eculizumab therapy was associated with a median increase of 6.4 points over baseline values established before treatment.
  • Eculizumab was safe and well-tolerated. There were no deaths in the study and only a single thrombotic event which occurred in a placebo patient in a site (the hepatic veins) which is typical of the thrombosis in PNH. The relative brief duration of this study was not sufficient to address the relevant issue of a possible protection from thrombosis by terminal complement inhibition with eculizumab.
  • Additional safety assessments, as well as efficacy measures, are being examined in an ongoing multi-center, open-label Phase III safety trial of eculizumab (SHEPHERD) in approximately 95 patients with PNH.
  • results from the current randomized, double-blind, placebo-controlled, global study show that terminal complement inhibition with eculizumab appears to be a safe and effective therapy for patients with the rare disorder PNH.
  • Treatment with eculizumab reduced intravascular hemolysis, and stabilized hemoglobin levels despite a reduction of transfusions, to the point where most PNH patients were rendered transfusion independent. Substantial and clinically meaningful improvements in fatigue and other key quality of life parameters were also demonstrated. All of the 85 patients who completed the study elected to receive eculizumab in an open-label extension study and all currently remain on drug.
  • the results of the TRIUMPH study indicate that terminal complement inhibition with eculizumab safely and effectively addresses an important consequence of the underlying genetic defect in PNH hematopoietic stem cells by providing a therapeutic replacement for the terminal complement inhibitor deficiency.

Landscapes

  • Health & Medical Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • General Health & Medical Sciences (AREA)
  • Veterinary Medicine (AREA)
  • Public Health (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Animal Behavior & Ethology (AREA)
  • Organic Chemistry (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Engineering & Computer Science (AREA)
  • General Chemical & Material Sciences (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Neurology (AREA)
  • Neurosurgery (AREA)
  • Biomedical Technology (AREA)
  • Immunology (AREA)
  • Diabetes (AREA)
  • Hematology (AREA)
  • Epidemiology (AREA)
  • Molecular Biology (AREA)
  • Biochemistry (AREA)
  • Psychiatry (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Dermatology (AREA)
  • Physical Education & Sports Medicine (AREA)
  • Obesity (AREA)
  • Nutrition Science (AREA)
  • Pain & Pain Management (AREA)
  • Genetics & Genomics (AREA)
  • Biophysics (AREA)
  • Heart & Thoracic Surgery (AREA)
  • Oncology (AREA)
  • Hospice & Palliative Care (AREA)
  • Orthopedic Medicine & Surgery (AREA)
  • Anesthesiology (AREA)
  • Urology & Nephrology (AREA)
  • Endocrinology (AREA)
US12/225,040 2006-03-15 2007-03-15 Treatment Of Paroxysmal Nocturnal Hemoglobinuria Patients By An Inhibitor Of Complement Abandoned US20090220508A1 (en)

Priority Applications (8)

Application Number Priority Date Filing Date Title
US12/225,040 US20090220508A1 (en) 2006-03-15 2007-03-15 Treatment Of Paroxysmal Nocturnal Hemoglobinuria Patients By An Inhibitor Of Complement
US13/426,973 US20120237515A1 (en) 2006-03-15 2012-03-22 Treatment of paroxysmal nocturnal hemoglobinuria patients by an inhibitor of complement
US15/148,839 US9718880B2 (en) 2006-03-15 2016-05-06 Treatment of paroxysmal nocturnal hemoglobinuria patients by an inhibitor of complement
US15/260,888 US9725504B2 (en) 2006-03-15 2016-09-09 Treatment of paroxysmal nocturnal hemoglobinuria patients by an inhibitor of complement
US15/284,015 US9732149B2 (en) 2006-03-15 2016-10-03 Treatment of paroxysmal nocturnal hemoglobinuria patients by an inhibitor of complement
US15/642,096 US10590189B2 (en) 2006-03-15 2017-07-05 Treatment of paroxysmal nocturnal hemoglobinuria patients by an inhibitor of complement
US16/750,978 US20200392216A1 (en) 2006-03-15 2020-01-23 Treatment of paroxysmal nocturnal hemoglobinuria patients by an inhibitor of complement
US16/804,567 US10703809B1 (en) 2006-03-15 2020-02-28 Treatment of paroxysmal nocturnal hemoglobinuria patients by an inhibitor of complement

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
US78307006P 2006-03-15 2006-03-15
PCT/US2007/006606 WO2007106585A1 (en) 2006-03-15 2007-03-15 Treatment of paroxysmal nocturnal hemoglobinuria patients by an inhibitor of complement
USPCT/US07/06606 2007-03-15
US12/225,040 US20090220508A1 (en) 2006-03-15 2007-03-15 Treatment Of Paroxysmal Nocturnal Hemoglobinuria Patients By An Inhibitor Of Complement

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2007/006606 A-371-Of-International WO2007106585A1 (en) 2006-03-15 2007-03-15 Treatment of paroxysmal nocturnal hemoglobinuria patients by an inhibitor of complement

Related Child Applications (1)

Application Number Title Priority Date Filing Date
US13/426,973 Continuation US20120237515A1 (en) 2006-03-15 2012-03-22 Treatment of paroxysmal nocturnal hemoglobinuria patients by an inhibitor of complement

Publications (1)

Publication Number Publication Date
US20090220508A1 true US20090220508A1 (en) 2009-09-03

Family

ID=38222520

Family Applications (8)

Application Number Title Priority Date Filing Date
US12/225,040 Abandoned US20090220508A1 (en) 2006-03-15 2007-03-15 Treatment Of Paroxysmal Nocturnal Hemoglobinuria Patients By An Inhibitor Of Complement
US13/426,973 Abandoned US20120237515A1 (en) 2006-03-15 2012-03-22 Treatment of paroxysmal nocturnal hemoglobinuria patients by an inhibitor of complement
US15/148,839 Active US9718880B2 (en) 2006-03-15 2016-05-06 Treatment of paroxysmal nocturnal hemoglobinuria patients by an inhibitor of complement
US15/260,888 Active US9725504B2 (en) 2006-03-15 2016-09-09 Treatment of paroxysmal nocturnal hemoglobinuria patients by an inhibitor of complement
US15/284,015 Active US9732149B2 (en) 2006-03-15 2016-10-03 Treatment of paroxysmal nocturnal hemoglobinuria patients by an inhibitor of complement
US15/642,096 Active 2027-09-08 US10590189B2 (en) 2006-03-15 2017-07-05 Treatment of paroxysmal nocturnal hemoglobinuria patients by an inhibitor of complement
US16/750,978 Pending US20200392216A1 (en) 2006-03-15 2020-01-23 Treatment of paroxysmal nocturnal hemoglobinuria patients by an inhibitor of complement
US16/804,567 Active US10703809B1 (en) 2006-03-15 2020-02-28 Treatment of paroxysmal nocturnal hemoglobinuria patients by an inhibitor of complement

Family Applications After (7)

Application Number Title Priority Date Filing Date
US13/426,973 Abandoned US20120237515A1 (en) 2006-03-15 2012-03-22 Treatment of paroxysmal nocturnal hemoglobinuria patients by an inhibitor of complement
US15/148,839 Active US9718880B2 (en) 2006-03-15 2016-05-06 Treatment of paroxysmal nocturnal hemoglobinuria patients by an inhibitor of complement
US15/260,888 Active US9725504B2 (en) 2006-03-15 2016-09-09 Treatment of paroxysmal nocturnal hemoglobinuria patients by an inhibitor of complement
US15/284,015 Active US9732149B2 (en) 2006-03-15 2016-10-03 Treatment of paroxysmal nocturnal hemoglobinuria patients by an inhibitor of complement
US15/642,096 Active 2027-09-08 US10590189B2 (en) 2006-03-15 2017-07-05 Treatment of paroxysmal nocturnal hemoglobinuria patients by an inhibitor of complement
US16/750,978 Pending US20200392216A1 (en) 2006-03-15 2020-01-23 Treatment of paroxysmal nocturnal hemoglobinuria patients by an inhibitor of complement
US16/804,567 Active US10703809B1 (en) 2006-03-15 2020-02-28 Treatment of paroxysmal nocturnal hemoglobinuria patients by an inhibitor of complement

Country Status (17)

Country Link
US (8) US20090220508A1 (pl)
EP (5) EP4316465A3 (pl)
JP (8) JP2009530299A (pl)
KR (2) KR20080110800A (pl)
AU (1) AU2007225044C1 (pl)
BR (2) BR122017024057B8 (pl)
CA (2) CA2645810C (pl)
DK (1) DK2359834T5 (pl)
ES (2) ES2612118T3 (pl)
HK (1) HK1161119A1 (pl)
HU (1) HUE030105T2 (pl)
IL (4) IL302157A (pl)
MX (2) MX340784B (pl)
PL (1) PL2359834T3 (pl)
PT (1) PT2359834T (pl)
SI (1) SI2359834T1 (pl)
WO (1) WO2007106585A1 (pl)

Cited By (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20130273052A1 (en) * 2010-10-01 2013-10-17 Alexion Pharmaceuticals, Inc. Polypeptides that bind to human complement component c5
US9718880B2 (en) 2006-03-15 2017-08-01 Alexion Pharmaceuticals, Inc. Treatment of paroxysmal nocturnal hemoglobinuria patients by an inhibitor of complement
US9771418B2 (en) * 2008-11-10 2017-09-26 Alexion Pharmaceuticals, Inc. Methods for treating complement-associated disorders
KR101796886B1 (ko) * 2014-03-07 2017-11-10 알렉시온 파마슈티칼스, 인코포레이티드 개선된 약동학을 가지는 항-c5 항체
WO2018109588A3 (en) * 2016-12-16 2018-08-02 Samsung Bioepis Co., Ltd Stable aqueous anti-c5 antibody composition
US10125191B2 (en) 2005-05-11 2018-11-13 Alexion Pharmaceutiacls, Inc. Compositions comprising an anti-C5 antibody

Families Citing this family (110)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP2845865A1 (en) 2004-11-12 2015-03-11 Xencor Inc. Fc variants with altered binding to FcRn
JP5624276B2 (ja) 2006-03-31 2014-11-12 中外製薬株式会社 抗体の血中動態を制御する方法
EP3689912A1 (en) 2007-09-26 2020-08-05 Chugai Seiyaku Kabushiki Kaisha Method of modifying isoelectric point of antibody via amino acid substitution in cdr
EP3575317A1 (en) * 2007-12-26 2019-12-04 Xencor, Inc. Fc variants with altered binding to fcrn
TWI564021B (zh) 2008-04-11 2017-01-01 Chugai Pharmaceutical Co Ltd Repeated binding of antigen to antigen binding molecules
MX356218B (es) 2008-08-05 2018-05-18 Novartis Ag Composiciones y métodos para anticuerpos que se dirigen a la proteína de complemento c5.
WO2011057250A1 (en) * 2009-11-09 2011-05-12 Alexion Pharmaceuticals, Inc. Reagents and methods for detecting pnh type ii white blood cells and their identification as risk factors for thrombotic disorders
US20120100166A1 (en) 2010-07-15 2012-04-26 Zyngenia, Inc. Ang-2 Binding Complexes and Uses Thereof
MX365235B (es) 2010-11-30 2019-05-28 Chugai Pharmaceutical Co Ltd Molécula de unión a antígeno capaz de unir repetidamente a la pluralidad de moléculas de antígeno.
US20140088019A1 (en) 2011-02-11 2014-03-27 Zyngenia, Inc. Monovalent and Multivalent Multispecific Complexes and Uses Thereof
CN106432506A (zh) 2011-05-24 2017-02-22 泽恩格尼亚股份有限公司 多价和单价多特异性复合物及其用途
CN106831985A (zh) 2011-12-21 2017-06-13 诺华股份有限公司 用于抗体靶定p因子的组合物和方法
KR102336895B1 (ko) 2012-02-20 2021-12-08 스위디쉬 오르펀 바이오비트럼 에이비 (피유비엘) 인간 보체 c5에 결합하는 폴리펩티드
CN104205155A (zh) * 2012-03-16 2014-12-10 亚力克恩制药公司 给接受补体抑制剂的患者分配补体抑制药物的方法
TWI635098B (zh) 2013-02-01 2018-09-11 再生元醫藥公司 含嵌合恆定區之抗體
EA201890895A1 (ru) 2013-03-15 2019-02-28 Зинджения, Инк. Мультивалентные и моновалентные мультиспецифические комплексы и их применение
US20160051673A1 (en) * 2013-03-29 2016-02-25 Alexion Pharmaceuticals, Inc. Compositions and methods for increasing the serum half-life of a therapeutic agent targeting complement c5
DK3039033T3 (da) 2013-08-28 2019-08-26 Affibody Ab Bindende polypeptider, der har et muteret skelet
RU2733897C2 (ru) * 2013-08-28 2020-10-09 Сведиш Орфан Биовитрум Аб (Пабл) Стабильные полипептиды, связывающиеся с с5 комплемента человека
US20150104445A1 (en) 2013-10-10 2015-04-16 Viropharma Holdings Limited Methods of inhibiting the alternative pathway of complement immune system activation and compositions used therein
TWI701042B (zh) 2014-03-19 2020-08-11 美商再生元醫藥公司 用於腫瘤治療之方法及抗體組成物
NZ727420A (en) 2014-06-12 2018-06-29 Ra Pharmaceuticals Inc Modulation of complement activity
IL279606B (en) 2014-08-08 2022-08-01 Alector Llc Anti-trem2 antibodies and methods of using them
DK3200791T3 (da) 2014-09-29 2020-05-25 Chemocentryx Inc Fremgangsmåder og mellemprodukter i fremstilling af C5AR- antagonister
JP6948942B2 (ja) 2014-10-15 2021-10-13 アレクシオン ファーマシューティカルズ, インコーポレイテッド タンパク質産物の等電性プロファイルをシフトさせる方法およびその使用
JP6778185B2 (ja) * 2014-10-15 2020-10-28 アレクシオン ファーマシューティカルズ, インコーポレイテッド 細胞を培養する方法
TR201903267T4 (tr) 2014-10-15 2019-03-21 Alexion Pharma Inc Büyük ölçekli eculizumab üretim hücre kültürünün replikasyon metotları.
KR102614189B1 (ko) 2014-11-17 2023-12-18 리제너론 파아마슈티컬스, 인크. Cd3xcd20 이특이적 항체를 이용한 종양의 치료 방법
US20160168237A1 (en) 2014-12-12 2016-06-16 Alexion Pharmaceuticals, Inc. Method for treating a complement mediated disorder caused by an infectious agent in a patient
CN113045650A (zh) 2014-12-19 2021-06-29 中外制药株式会社 抗-c5抗体及使用方法
CN107428823B (zh) 2015-01-22 2021-10-26 中外制药株式会社 两种以上抗-c5抗体的组合与使用方法
PT3250230T (pt) 2015-01-28 2021-12-07 Ra Pharmaceuticals Inc Moduladores da atividade do complemento
EP3274359B1 (en) 2015-03-23 2022-11-02 Alexion Pharmaceuticals, Inc. Virus filtration
CA2981312C (en) 2015-03-30 2023-09-26 Regeneron Pharmaceuticals, Inc. Heavy chain constant regions with reduced binding to fc gamma receptors
WO2016160756A2 (en) * 2015-03-31 2016-10-06 Alexion Pharmaceuticlas, Inc. Identifying and treating subpopulations of paroxysmal nocturnal hemoglobinuria (pnh) patents
NZ736022A (en) 2015-04-06 2024-02-23 Subdomain Llc De novo binding domain containing polypeptides and uses thereof
PL3280441T3 (pl) 2015-04-07 2022-02-21 Alector Llc Przeciwciała przeciwko sortilinie i sposoby ich stosowania
US11840564B2 (en) 2015-06-09 2023-12-12 Children's Hospital Medical Center Dosing algorithm for complement inhibitor
JP7497953B2 (ja) 2015-06-12 2024-06-11 アレクトル エルエルシー 抗cd33抗体及びその使用方法
EP3307771A2 (en) 2015-06-12 2018-04-18 Alector LLC Anti-cd33 antibodies and methods of use thereof
EP3313437A1 (en) * 2015-06-26 2018-05-02 Alexion Pharmaceuticals, Inc. A method for treating a patient in compliance with vaccination with eculizumab or an eculizumab variant
WO2017035401A1 (en) 2015-08-26 2017-03-02 Achillion Pharmaceuticals, Inc. Amide compounds for treatment of immune and inflammatory disorders
WO2017035405A1 (en) 2015-08-26 2017-03-02 Achillion Pharmaceuticals, Inc. Amino compounds for treatment of immune and inflammatory disorders
JP2018525999A (ja) 2015-08-28 2018-09-13 アレクトル エルエルシー 抗Siglec−7抗体及びその使用方法
JP2018528218A (ja) 2015-09-11 2018-09-27 アンドリアン、ブルース 組換えグリコシル化エクリズマブおよびエクリズマブ変異体
SG10201912150TA (en) 2015-10-06 2020-02-27 Alector Llc Anti-trem2 antibodies and methods of use thereof
EP3359566A1 (en) 2015-10-07 2018-08-15 Alexion Pharmaceuticals, Inc. A method for treating age-related macular degeneration in a patient
CN116003596A (zh) 2015-10-29 2023-04-25 艾利妥 抗siglec-9抗体及其使用方法
EP3389692B1 (en) 2015-12-16 2020-03-04 RA Pharmaceuticals, Inc. Modulators of complement activity
EP3402486A4 (en) * 2016-01-14 2019-08-28 ChemoCentryx, Inc. PROCESS FOR TREATING C3 GLOMERULOPATHY
US11472877B2 (en) 2016-03-04 2022-10-18 Alector Llc Anti-TREM1 antibodies and methods of use thereof
EP3426680A4 (en) 2016-03-10 2020-03-11 Acceleron Pharma Inc. TYPE 2 ACTIVIN RECEPTOR BINDING PROTEINS AND USES THEREOF
JP7102353B2 (ja) 2016-06-14 2022-07-19 リジェネロン・ファーマシューティカルズ・インコーポレイテッド 抗c5抗体及びそれらの使用
CA3026050A1 (en) 2016-08-05 2018-02-08 Chugai Seiyaku Kabushiki Kaisha Composition for prophylaxis or treatment of il-8 related diseases
JP7096240B2 (ja) 2016-10-19 2022-07-05 アレクシオン ファーマシューティカルズ, インコーポレイテッド 試料中の非結合c5の定量化方法
US11123399B2 (en) 2016-12-07 2021-09-21 Ra Pharmaceuticals, Inc. Modulators of complement activity
TWI828672B (zh) 2017-01-31 2024-01-11 日商中外製藥股份有限公司 用於治療或預防c5相關疾病的醫藥組合物以及治療或預防c5相關疾病的方法
WO2018160891A1 (en) 2017-03-01 2018-09-07 Achillion Pharmaceutical, Inc. Pharmaceutical compounds for treatment of medical disorders
EP3985002A1 (en) 2017-03-01 2022-04-20 Achillion Pharmaceuticals, Inc. Aryl, heteroary, and heterocyclic pharmaceutical compounds for treatment of medical disorders
WO2018213316A1 (en) 2017-05-16 2018-11-22 Alector Llc Anti-siglec-5 antibodies and methods of use thereof
JP7194698B2 (ja) 2017-05-22 2022-12-22 アレクシオン ファーマシューティカルズ, インコーポレイテッド 患者においてタンパク質漏出性腸症を処置するための抗c5抗体の投薬量および投与
DK3658184T3 (da) 2017-07-27 2023-11-27 Alexion Pharma Inc Højkoncentrerede anti-c5-antistofformuleringer
CN111163767A (zh) * 2017-08-02 2020-05-15 艾其林医药公司 治疗阵发性睡眠性血红蛋白尿症的治疗方案
JP7299160B2 (ja) 2017-08-03 2023-06-27 アレクトル エルエルシー 抗cd33抗体及びその使用方法
NZ758301A (en) 2017-08-03 2023-07-28 Alector Llc Anti-trem2 antibodies and methods of use thereof
KR20200096769A (ko) 2017-11-14 2020-08-13 아르셀엑스, 인크. D-도메인 함유 폴리펩타이드 및 이의 용도
WO2019112984A1 (en) * 2017-12-04 2019-06-13 Ra Pharmaceuticals, Inc. Modulators of complement activity
MA51147A (fr) 2017-12-13 2021-03-24 Regeneron Pharma Associations d'anticorps anti-c5 et utilisations associées
CN111886246A (zh) 2017-12-29 2020-11-03 艾莱克特有限责任公司 抗tmem106b抗体及其使用方法
CN111971301A (zh) 2018-01-31 2020-11-20 艾莱克特有限责任公司 抗ms4a4a抗体及其使用方法
CA3098710A1 (en) 2018-05-25 2019-11-28 Alector Llc Anti-sirpa antibodies and methods of use thereof
CN112512638A (zh) 2018-06-08 2021-03-16 艾利妥 抗siglec-7抗体及其使用方法
WO2019246293A2 (en) * 2018-06-19 2019-12-26 Atarga, Llc Antibody molecules to complement component 5 and uses thereof
KR20210025614A (ko) 2018-06-29 2021-03-09 알렉터 엘엘씨 항-sirp-베타1 항체 및 그의 사용 방법
RS64034B1 (sr) 2018-07-13 2023-04-28 Alector Llc Anti-sortilin antitela i načini njihove upotrebe
MX2021000933A (es) 2018-07-27 2021-05-27 Alector Llc Anticuerpos anti-siglec-5 y métodos para su uso.
WO2020027279A1 (en) 2018-08-01 2020-02-06 Chugai Seiyaku Kabushiki Kaisha A pharmaceutical composition for use in the treatment or prevention of a c5-related disease and a method for treating or preventing a c5-related disease
CN112771077A (zh) 2018-08-31 2021-05-07 瑞泽恩制药公司 针对cd3/c20双特异性抗体的减轻细胞因子释放综合征的给药策略
SG11202101552SA (en) 2018-08-31 2021-03-30 Alector Llc Anti-cd33 antibodies and methods of use thereof
EP3847174A4 (en) 2018-09-06 2022-06-15 Achillion Pharmaceuticals, Inc. MORPHIC FORMS OF COMPLEMENT D FACTOR INHIBITORS
EP3846803A4 (en) 2018-09-06 2022-08-10 Achillion Pharmaceuticals, Inc. MACROCYCLIC COMPOUNDS FOR THE TREATMENT OF MEDICAL DISORDERS
MX2021003425A (es) 2018-09-25 2021-07-16 Achillion Pharmaceuticals Inc Formas morficas de los inhibidores del factor d del complemento.
TW202029980A (zh) 2018-10-26 2020-08-16 美商免疫遺傳股份有限公司 E p C A M 抗體、可活化抗體及免疫偶聯物以及其用途
AU2020291527A1 (en) 2019-06-11 2022-01-20 Alector Llc Anti-Sortilin antibodies for use in therapy
US20220275070A1 (en) 2019-07-31 2022-09-01 Hoffmann-La Roche Inc. Dosage and administration regimen for the treatment or prevention of c5-related diseases by the use of the anti-c5 antibody crovalimab
AU2020321969A1 (en) 2019-07-31 2022-03-17 Alector Llc Anti-MS4A4A antibodies and methods of use thereof
WO2021019033A1 (en) 2019-07-31 2021-02-04 F. Hoffmann-La Roche Ag Dosage and administration regimen for the treatment or prevention of c5-related diseases by the use of the anti-c5 antibody crovalimab
US20220259305A1 (en) 2019-08-05 2022-08-18 Alexion Pharmaceuticals, Inc. Methods for treatment of refractory generalized myasthenia gravis with eculizumab
EP3986937A1 (en) 2019-12-05 2022-04-27 Alector LLC Methods of use of anti-trem2 antibodies
MX2022007156A (es) 2019-12-12 2022-09-07 Alector Llc Metodos de uso de anticuerpos anti-cd33.
EP4072680A1 (en) 2019-12-13 2022-10-19 Alector LLC Anti-mertk antibodies and methods of use thereof
JP2023515478A (ja) 2020-02-24 2023-04-13 アレクトル エルエルシー 抗trem2抗体の使用方法
EP4126937A1 (en) 2020-03-31 2023-02-08 Alector LLC Anti-mertk antibodies and methods of use thereof
AU2021247286A1 (en) 2020-04-03 2022-10-20 Alector Llc Methods of use of anti-TREM2 antibodies
JP2023522208A (ja) 2020-04-16 2023-05-29 アシスタンス ピュブリック-オピト ド パリ ウイルスによって引き起こされる補体媒介性障害を処置する方法
KR20230009431A (ko) 2020-05-12 2023-01-17 알렉시온 파마슈티칼스, 인코포레이티드 발작성 야간 혈색소뇨증의 치료를 위한 보체 인자 d 저해제를 단독으로 또는 항-c5 항체와 조합한 용도
JP2023553399A (ja) 2020-12-02 2023-12-21 アレクトル エルエルシー 抗ソルチリン抗体の使用法
US20240174746A1 (en) 2021-03-18 2024-05-30 Alector Llc Anti-tmem106b antibodies and methods of use thereof
EP4314063A1 (en) 2021-03-23 2024-02-07 Alector LLC Anti-tmem106b antibodies for treating and preventing coronavirus infections
EP4348263A1 (en) 2021-05-28 2024-04-10 Alexion Pharmaceuticals, Inc. Methods for detecting cm-tma biomarkers
EP4355786A1 (en) 2021-06-16 2024-04-24 Alector LLC Bispecific anti-mertk and anti-pdl1 antibodies and methods of use thereof
WO2022266221A1 (en) 2021-06-16 2022-12-22 Alector Llc Monovalent anti-mertk antibodies and methods of use thereof
WO2023023220A1 (en) 2021-08-20 2023-02-23 Alexion Pharmaceuticals, Inc. Methods for treating sickle cell disease or beta thalassemia using a complement alternative pathway inhibitor
WO2023069919A1 (en) 2021-10-19 2023-04-27 Alector Llc Anti-cd300lb antibodies and methods of use thereof
WO2023081898A1 (en) 2021-11-08 2023-05-11 Alector Llc Soluble cd33 as a biomarker for anti-cd33 efficacy
KR20230105972A (ko) * 2022-01-05 2023-07-12 주식회사 카나프테라퓨틱스 혈관신생 억제제가 결합된 항-C3b 항체 또는 항-C5 항체 및 이의 용도
WO2023164516A1 (en) 2022-02-23 2023-08-31 Alector Llc Methods of use of anti-trem2 antibodies
WO2024015830A1 (en) 2022-07-12 2024-01-18 Cytomx Therapeutics, Inc. Epcam immunoconjugates and uses thereof
TW202415679A (zh) 2022-07-29 2024-04-16 美商阿列克特有限責任公司 抗gpnmb抗體及其使用方法
WO2024086796A1 (en) 2022-10-20 2024-04-25 Alector Llc Anti-ms4a4a antibodies with amyloid-beta therapies

Citations (7)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4686100A (en) * 1985-04-02 1987-08-11 The Board Of Trustees Of The Leland Stanford Junior University Method for the treatment of adult respiratory distress syndrome
US5135916A (en) * 1989-06-12 1992-08-04 Oklahoma Medical Research Foundation Inhibition of complement mediated inflammatory response
US6355245B1 (en) * 1994-05-02 2002-03-12 Alexion Pharmaceuticals, Inc. C5-specific antibodies for the treatment of inflammatory diseases
US20030049683A1 (en) * 2000-12-05 2003-03-13 Bowdish Katherine S. Rationally designed antibodies
US20050191298A1 (en) * 2004-02-03 2005-09-01 Leonard Bell Method of treating hemolytic disease
US20050221382A1 (en) * 2002-03-18 2005-10-06 Rother Russell P Stratification of patient populations having or suspected of having rheumatoid arthritis
US7361339B2 (en) * 2003-01-09 2008-04-22 Alexion Pharmaceuticals, Inc. Methods for reducing morality associated with acute myocardial infarction

Family Cites Families (64)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
GB8308235D0 (en) 1983-03-25 1983-05-05 Celltech Ltd Polypeptides
US4816567A (en) 1983-04-08 1989-03-28 Genentech, Inc. Recombinant immunoglobin preparations
GB8422238D0 (en) 1984-09-03 1984-10-10 Neuberger M S Chimeric proteins
GB8607679D0 (en) 1986-03-27 1986-04-30 Winter G P Recombinant dna product
US5225539A (en) 1986-03-27 1993-07-06 Medical Research Council Recombinant altered antibodies and methods of making altered antibodies
US6548640B1 (en) 1986-03-27 2003-04-15 Btg International Limited Altered antibodies
US5260203A (en) 1986-09-02 1993-11-09 Enzon, Inc. Single polypeptide chain binding molecules
US4946778A (en) 1987-09-21 1990-08-07 Genex Corporation Single polypeptide chain binding molecules
US5530101A (en) 1988-12-28 1996-06-25 Protein Design Labs, Inc. Humanized immunoglobulins
US5705732A (en) 1989-06-12 1998-01-06 Oklahoma Medical Research Foundation Universal donor cells
DE3920358A1 (de) 1989-06-22 1991-01-17 Behringwerke Ag Bispezifische und oligospezifische, mono- und oligovalente antikoerperkonstrukte, ihre herstellung und verwendung
DE3924924A1 (de) 1989-07-27 1991-02-07 Goetze Otto Verfahren zum nachweis und/oder der quantitativen bestimmung von komplementpeptid c5a und/oder c5adesarg
WO1992010205A1 (en) 1990-12-06 1992-06-25 T Cell Sciences, Inc. Synergistic compositions of soluble complement receptors and compounds that inhibit complement and/or suppress immune activity
CA2103059C (en) 1991-06-14 2005-03-22 Paul J. Carter Method for making humanized antibodies
US6329509B1 (en) 1991-08-14 2001-12-11 Genentech, Inc. Anti-IgE antibodies
US5565332A (en) 1991-09-23 1996-10-15 Medical Research Council Production of chimeric antibodies - a combinatorial approach
US6025165A (en) 1991-11-25 2000-02-15 Enzon, Inc. Methods for producing multivalent antigen-binding proteins
DK1136556T3 (da) 1991-11-25 2005-10-03 Enzon Inc Fremgangsmåde til fremstilling af multivalente antigen-bindende proteiner
US5456909A (en) 1992-08-07 1995-10-10 T Cell Sciences, Inc. Glycoform fractions of recombinant soluble complement receptor 1 (sCR1) having extended half-lives in vivo
ATE243745T1 (de) 1994-01-31 2003-07-15 Univ Boston Bibliotheken aus polyklonalen antikörpern
ES2252746T3 (es) 1994-09-23 2006-05-16 Alexion Pharmaceuticals, Inc. Procedimientos para el tratamiento de la enfermedad inflamatoria articular.
US6685940B2 (en) 1995-07-27 2004-02-03 Genentech, Inc. Protein formulation
JP2002514895A (ja) 1995-09-28 2002-05-21 アレクション、ファーマスーティカルズ、インコーポレーテッド ブタ細胞相互作用タンパク質
US6171586B1 (en) 1997-06-13 2001-01-09 Genentech, Inc. Antibody formulation
US6991790B1 (en) 1997-06-13 2006-01-31 Genentech, Inc. Antibody formulation
AU2001234962A1 (en) 2000-02-10 2001-08-20 Alexion Pharmaceuticals, Inc. Mixtures of caspase inhibitors and complement inhibitors and methods of use thereof
PT1325033E (pt) 2000-10-10 2010-04-15 Genentech Inc Inibição da activação do complemento c5 para o tratamento e a prevenção da rejeição diferida de um xenoenxertos ou de uma rejeição vascular aguda
US7396917B2 (en) 2000-12-05 2008-07-08 Alexion Pharmaceuticals, Inc. Rationally designed antibodies
US20040253242A1 (en) 2000-12-05 2004-12-16 Bowdish Katherine S. Rationally designed antibodies
US20030133939A1 (en) 2001-01-17 2003-07-17 Genecraft, Inc. Binding domain-immunoglobulin fusion proteins
CA2454562A1 (en) 2001-07-26 2003-02-06 Alexion Pharmaceuticals, Inc. Method of improving cognitive function
ES2283594T5 (es) 2001-08-17 2016-03-15 Genentech, Inc. Inhibidores de la ruta del complemento que se unen a C5 y C5a sin impedir la formación de C5b
US7393648B2 (en) 2001-12-03 2008-07-01 Alexion Pharmaceuticals, Inc. Hybrid antibodies
US7425618B2 (en) 2002-06-14 2008-09-16 Medimmune, Inc. Stabilized anti-respiratory syncytial virus (RSV) antibody formulations
PT2314629E (pt) 2002-07-18 2014-01-22 Merus B V Produção recombinante de misturas de anticorpos
AU2003265361A1 (en) 2002-08-28 2004-03-19 Pharmacia Corporation Stable ph optimized formulation of a modified antibody
US9415102B2 (en) 2002-09-06 2016-08-16 Alexion Pharmaceuticals, Inc. High concentration formulations of anti-C5 antibodies
US20050271660A1 (en) 2002-09-06 2005-12-08 Alexion Pharmaceuticals, Inc. Nebulization of monoclonal antibodies for treating pulmonary diseases
DK2345671T3 (en) 2002-09-27 2016-02-15 Xencor Inc Optimized Fc variants and methods for their formation
EP3284753B1 (en) 2002-10-17 2019-06-05 Genmab A/S Human monoclonal antibodies against cd20 for use in the treatment of multiple sclerosis
US20040146948A1 (en) 2002-10-18 2004-07-29 Centenary Institute Of Cancer Medicine And Cell Biology Compositions and methods for targeting antigen-presenting cells with antibody single-chain variable region fragments
EP1576812A1 (en) 2002-12-16 2005-09-21 Koninklijke Philips Electronics N.V. Personal video recorder method and user interface
CA2512174A1 (en) 2003-01-30 2004-08-12 Christian B. Allan Anti-integrin .alpha..nu..beta.3 antibody formulations and uses thereof
JP4728948B2 (ja) 2003-02-10 2011-07-20 エラン ファーマシューティカルズ,インコーポレイテッド 免疫グロブリン製剤およびその調製の方法
EP1610820B2 (en) 2003-04-04 2013-08-21 Genentech, Inc. High concentration antibody and protein formulations
JP2007525176A (ja) * 2003-04-25 2007-09-06 イミュネックス・コーポレーション 組換えタンパク質発現の誘導剤
EP1635872A4 (en) 2003-05-30 2008-01-02 Alexion Pharma Inc ANTIBODIES AND FUSION PROTEINS COMPRISING MODIFIED CONSTANT REGIONS
AU2004245038A1 (en) 2003-06-02 2004-12-16 Alexion Pharmaceuticals, Inc. De-immunized anti-CD3 antibody
US20050027166A1 (en) 2003-06-17 2005-02-03 Shinya Matsumoto Endoscope system for fluorescent observation
AT412877B (de) 2003-07-01 2005-08-25 Miba Gleitlager Gmbh Schichtwerkstoff
US20070116710A1 (en) 2004-02-03 2007-05-24 Leonard Bell Methods of treating hemolytic anemia
ES2376556T3 (es) 2004-03-24 2012-03-14 Abbott Biotherapeutics Corp. Utilización de la anticuerpos anti-alfa5beta1 para inhibir la proliferación de las células cancerosas.
SI1755674T1 (sl) 2004-05-14 2015-04-30 Alexion Pharmaceuticals, Inc. Podaljšanje preživetja alotransplantata z inhibiranjem aktivnosti komplementa
EP2287195B1 (en) 2004-07-01 2019-05-15 Novo Nordisk A/S Pan-kir2dl nk-receptor antibodies and their use in diagnostik and therapy
US7470521B2 (en) 2004-07-20 2008-12-30 Critical Therapeutics, Inc. RAGE protein derivatives
WO2006127517A2 (en) 2005-05-20 2006-11-30 Genentech, Inc. Pretreatment of a biological sample from an autoimmune disease subject
WO2007002571A2 (en) 2005-06-23 2007-01-04 Alexion Pharmaceuticals, Inc. Use of an anti c5 complement antibody to treat patients with sickle cell disease
US8309690B2 (en) 2005-07-01 2012-11-13 Medimmune, Llc Integrated approach for generating multidomain protein therapeutics
KR20080098504A (ko) 2006-02-03 2008-11-10 메디뮨 엘엘씨 단백질 제제
SI1988882T1 (sl) 2006-03-02 2015-07-31 Alexion Pharmaceuticals, Inc. Podaljšanje preživetja alografta z inhibiranjem aktivnosti komplementa
US20090220508A1 (en) 2006-03-15 2009-09-03 Alexion Pharmaceuticals, Inc. Treatment Of Paroxysmal Nocturnal Hemoglobinuria Patients By An Inhibitor Of Complement
EP2013235A1 (en) 2006-05-01 2009-01-14 Alexion Pharmaceuticals, Inc. Methods for the treatment of muscular dystrophy associated with dysferlin-deficiency
EP2913342A1 (en) 2006-09-05 2015-09-02 Alexion Pharmaceuticals, Inc. Compositions for use in the treatment of antibody mediated neuropathies
TWI629483B (zh) 2010-06-11 2018-07-11 協和醱酵麒麟有限公司 anti-TIM-3 antibody

Patent Citations (8)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4686100A (en) * 1985-04-02 1987-08-11 The Board Of Trustees Of The Leland Stanford Junior University Method for the treatment of adult respiratory distress syndrome
US5135916A (en) * 1989-06-12 1992-08-04 Oklahoma Medical Research Foundation Inhibition of complement mediated inflammatory response
US5660825A (en) * 1989-06-12 1997-08-26 Oklahoma Medical Research Foundation Method of inhibition of complement mediated inflammatory response
US6355245B1 (en) * 1994-05-02 2002-03-12 Alexion Pharmaceuticals, Inc. C5-specific antibodies for the treatment of inflammatory diseases
US20030049683A1 (en) * 2000-12-05 2003-03-13 Bowdish Katherine S. Rationally designed antibodies
US20050221382A1 (en) * 2002-03-18 2005-10-06 Rother Russell P Stratification of patient populations having or suspected of having rheumatoid arthritis
US7361339B2 (en) * 2003-01-09 2008-04-22 Alexion Pharmaceuticals, Inc. Methods for reducing morality associated with acute myocardial infarction
US20050191298A1 (en) * 2004-02-03 2005-09-01 Leonard Bell Method of treating hemolytic disease

Cited By (15)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US10125191B2 (en) 2005-05-11 2018-11-13 Alexion Pharmaceutiacls, Inc. Compositions comprising an anti-C5 antibody
US10927166B2 (en) 2005-05-11 2021-02-23 Alexion Pharmaceuticals, Inc. Compositions comprising an anti-C5 antibody
US10479828B2 (en) 2005-05-11 2019-11-19 Alexion Pharmaceuticals, Inc. Compositions comprising an anti-C5 antibody
US10590189B2 (en) 2006-03-15 2020-03-17 Alexion Pharmaceuticals, Inc. Treatment of paroxysmal nocturnal hemoglobinuria patients by an inhibitor of complement
US9718880B2 (en) 2006-03-15 2017-08-01 Alexion Pharmaceuticals, Inc. Treatment of paroxysmal nocturnal hemoglobinuria patients by an inhibitor of complement
US9725504B2 (en) 2006-03-15 2017-08-08 Alexion Pharmaceuticals, Inc. Treatment of paroxysmal nocturnal hemoglobinuria patients by an inhibitor of complement
US9732149B2 (en) 2006-03-15 2017-08-15 Alexion Pharmaceuticals, Inc. Treatment of paroxysmal nocturnal hemoglobinuria patients by an inhibitor of complement
US10703809B1 (en) 2006-03-15 2020-07-07 Alexion Pharmaceuticals, Inc. Treatment of paroxysmal nocturnal hemoglobinuria patients by an inhibitor of complement
US9771418B2 (en) * 2008-11-10 2017-09-26 Alexion Pharmaceuticals, Inc. Methods for treating complement-associated disorders
US20130273052A1 (en) * 2010-10-01 2013-10-17 Alexion Pharmaceuticals, Inc. Polypeptides that bind to human complement component c5
KR101796886B1 (ko) * 2014-03-07 2017-11-10 알렉시온 파마슈티칼스, 인코포레이티드 개선된 약동학을 가지는 항-c5 항체
WO2018109588A3 (en) * 2016-12-16 2018-08-02 Samsung Bioepis Co., Ltd Stable aqueous anti-c5 antibody composition
CN110087683A (zh) * 2016-12-16 2019-08-02 三星Bioepis股份有限公司 稳定水性抗c5抗体组合物
KR20190088081A (ko) * 2016-12-16 2019-07-25 삼성바이오에피스 주식회사 안정한 액상의 항-c5 항체 조성물
KR102579940B1 (ko) * 2016-12-16 2023-09-15 삼성바이오에피스 주식회사 안정한 액상의 항-c5 항체 조성물

Also Published As

Publication number Publication date
AU2007225044C1 (en) 2018-03-29
EP2359834A1 (en) 2011-08-24
EP3124029A1 (en) 2017-02-01
KR20080110800A (ko) 2008-12-19
US9718880B2 (en) 2017-08-01
IL252260B (en) 2018-04-30
JP2009530299A (ja) 2009-08-27
EP3167888A1 (en) 2017-05-17
WO2007106585A1 (en) 2007-09-20
PT2359834T (pt) 2016-12-23
US20160244516A1 (en) 2016-08-25
CA2645810A1 (en) 2007-09-20
MX340784B (es) 2016-07-26
CA3022097C (en) 2020-10-27
BRPI0708909B8 (pt) 2021-05-25
US9725504B2 (en) 2017-08-08
JP2020002176A (ja) 2020-01-09
EP2359834B1 (en) 2016-11-09
JP2018165277A (ja) 2018-10-25
EP3167888C0 (en) 2024-05-01
ES2965283T1 (es) 2024-04-11
JP2023026536A (ja) 2023-02-24
US9732149B2 (en) 2017-08-15
US10703809B1 (en) 2020-07-07
EP3167888B1 (en) 2024-05-01
MX2008011735A (es) 2009-02-06
CA3022097A1 (en) 2007-09-20
SI2359834T1 (sl) 2017-02-28
BR122017024057B8 (pt) 2021-05-25
EP4316465A3 (en) 2024-04-24
HUE030105T2 (en) 2017-04-28
ES2612118T3 (es) 2017-05-12
US20120237515A1 (en) 2012-09-20
BRPI0708909B1 (pt) 2021-02-09
AU2007225044A1 (en) 2007-09-20
CA2645810C (en) 2018-12-11
US20160376355A1 (en) 2016-12-29
JP2014015484A (ja) 2014-01-30
DK2359834T3 (en) 2017-01-16
PL2359834T3 (pl) 2017-09-29
IL194041A (en) 2017-10-31
KR20140057635A (ko) 2014-05-13
US20200392216A1 (en) 2020-12-17
EP4316465A2 (en) 2024-02-07
IL194041A0 (en) 2011-08-01
JP2016034978A (ja) 2016-03-17
BR122017024057B1 (pt) 2021-02-17
BRPI0708909A2 (pt) 2011-06-14
JP2021073319A (ja) 2021-05-13
DK2359834T5 (en) 2017-02-06
US10590189B2 (en) 2020-03-17
MX2022002589A (es) 2023-01-05
EP2001490A1 (en) 2008-12-17
JP6224059B2 (ja) 2017-11-01
JP2017082017A (ja) 2017-05-18
US20170349652A1 (en) 2017-12-07
ES2965283T3 (es) 2024-05-28
IL252260A0 (en) 2017-07-31
HK1161119A1 (zh) 2012-08-24
IL302157A (en) 2023-06-01
IL258548A (en) 2018-05-31
AU2007225044B2 (en) 2013-03-07
US20170015741A1 (en) 2017-01-19
US20200199211A1 (en) 2020-06-25

Similar Documents

Publication Publication Date Title
US10703809B1 (en) Treatment of paroxysmal nocturnal hemoglobinuria patients by an inhibitor of complement
WO2009149306A2 (en) Complement depleting compounds and methods of treating cancer comprising monoclonal antibody therapy and said complement depleting compounds
AU2013206214A1 (en) Treatment of paroxysmal nocturnal hemoglobinuria patients by an inhibitor of complement

Legal Events

Date Code Title Description
AS Assignment

Owner name: ALEXION PHARMACEUTICALS, INC., CONNECTICUT

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:BELL, LEONARD;ROTHER, RUSSELL P.;REEL/FRAME:022527/0623;SIGNING DATES FROM 20081017 TO 20081027

STCB Information on status: application discontinuation

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